51
|
Isolation of mitochondria-derived mitovesicles and subpopulations of microvesicles and exosomes from brain tissues. Nat Protoc 2022; 17:2517-2549. [PMID: 35962195 PMCID: PMC9633367 DOI: 10.1038/s41596-022-00719-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 04/29/2022] [Indexed: 11/08/2022]
Abstract
Extracellular vesicles (EVs) are nanoscale vesicles secreted into the extracellular space by all cell types, including neurons and astrocytes in the brain. EVs play pivotal roles in physiological and pathophysiological processes such as waste removal, cell-to-cell communication and transport of either protective or pathogenic material into the extracellular space. Here we describe a detailed protocol for the reliable and consistent isolation of EVs from both murine and human brains, intended for anyone with basic laboratory experience and performed in a total time of 27 h. The method includes a mild extracellular matrix digestion of the brain tissue, a series of filtration and centrifugation steps to purify EVs and an iodixanol-based high-resolution density step gradient that fractionates different EV populations, including mitovesicles, a newly identified type of EV of mitochondrial origin. We also report detailed downstream protocols for the characterization and analysis of brain EV preparations using nanotrack analysis, electron microscopy and western blotting, as well as for measuring mitovesicular ATP kinetics. Furthermore, we compared this novel iodixanol-based high-resolution density step gradient to the previously described sucrose-based gradient. Although the yield of total EVs recovered was similar, the iodixanol-based gradient better separated distinct EV species as compared with the sucrose-based gradient, including subpopulations of microvesicles, exosomes and mitovesicles. This technique allows quantitative, highly reproducible analyses of brain EV subtypes under normal physiological processes and pathological brain conditions, including neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease.
Collapse
|
52
|
Zou Y, Mu D, Ma X, Wang D, Zhong J, Gao J, Yu S, Qiu L. Review on the roles of specific cell-derived exosomes in Alzheimer's disease. Front Neurosci 2022; 16:936760. [PMID: 35968378 PMCID: PMC9366882 DOI: 10.3389/fnins.2022.936760] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/08/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the sixth leading cause of death worldwide and cannot be effectively cured or prevented; thus, early diagnosis, and intervention are important. The importance of exosomes, membrane-bound extracellular vesicles produced in the endosome of eukaryotic cells, in the development, diagnosis, and treatment of AD has been recognized; however, their specific functions remain controversial and even unclear. With the development of exosome extraction, isolation, and characterization, many studies have focused on exosomes derived from different cells and body fluids. In this study, we summarized the roles of exosomes derived from different body fluids and cells, such as neuron, glial, stem, and endothelial cells, in the development, diagnosis, monitoring, and treatment of AD. We also emphasize the necessity to focus on exosomes from biological fluids and specific cells that are less invasive to target. Moreover, aside from the concentrations of classic and novel biomarkers in exosomes, the size and number of exosomes may also influence early and differential diagnosis of AD.
Collapse
Affiliation(s)
- Yutong Zou
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Danni Mu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Xiaoli Ma
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
- Medical Science Research Center (MRC), Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Danchen Wang
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Jian Zhong
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
| | - Jing Gao
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Songlin Yu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
- Songlin Yu
| | - Ling Qiu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Ling Qiu
| |
Collapse
|
53
|
Aβ and Tau Regulate Microglia Metabolism via Exosomes in Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10081800. [PMID: 35892700 PMCID: PMC9332859 DOI: 10.3390/biomedicines10081800] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 12/17/2022] Open
Abstract
One of the most striking hallmarks shared by various neurodegenerative diseases, including Alzheimer’s disease (AD), is microglia-mediated neuroinflammation. The main pathological features of AD are extracellular amyloid-β (Aβ) plaques and intracellular tau-containing neurofibrillary tangles in the brain. Amyloid-β (Aβ) peptide and tau protein are the primary components of the plaques and tangles. The crosstalk between microglia and neurons helps maintain brain homeostasis, and the metabolic phenotype of microglia determines its polarizing phenotype. There are currently many research and development efforts to provide disease-modifying therapies for AD treatment. The main targets are Aβ and tau, but whether there is a causal relationship between neurodegenerative proteins, including Aβ oligomer and tau oligomer, and regulation of microglia metabolism in neuroinflammation is still controversial. Currently, the accumulation of Aβ and tau by exosomes or other means of propagation is proposed as a regulator in neurological disorders, leading to metabolic disorders of microglia that can play a key role in the regulation of immune cells. In this review, we propose that the accumulation of Aβ oligomer and tau oligomer can propagate to adjacent microglia through exosomes and change the neuroinflammatory microenvironment by microglia metabolic reprogramming. Clarifying the relationship between harmful proteins and microglia metabolism will help people to better understand the mechanism of crosstalk between neurons and microglia, and provide new ideas for the development of AD drugs.
Collapse
|
54
|
Congdon EE, Jiang Y, Sigurdsson EM. Targeting tau only extracellularly is likely to be less efficacious than targeting it both intra- and extracellularly. Semin Cell Dev Biol 2022; 126:125-137. [PMID: 34896021 PMCID: PMC9680670 DOI: 10.1016/j.semcdb.2021.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022]
Abstract
Aggregation of the tau protein is thought to be responsible for the neurodegeneration and subsequent functional impairments in diseases that are collectively named tauopathies. Alzheimer's disease is the most common tauopathy, but the group consists of over 20 different diseases, many of which have tau pathology as their primary feature. The development of tau therapies has mainly focused on preventing the formation of and/or clearing these aggregates. Of these, immunotherapies that aim to either elicit endogenous tau antibodies or deliver exogenous ones are the most common approach in clinical trials. While their mechanism of action can involve several pathways, both extra- and intracellular, pharmaceutical companies have primarily focused on antibody-mediated clearance of extracellular tau. As we have pointed out over the years, this is rather surprising because it is well known that most of pathological tau protein is found intracellularly. It has been repeatedly shown by several groups over the past decades that antibodies can enter neurons and that their cellular uptake can be enhanced by various means, particularly by altering their charge. Here, we will briefly describe the potential extra- and intracellular mechanisms involved in antibody-mediated clearance of tau pathology, discuss these in the context of recent failures of some of the tau antibody trials, and finally provide a brief overview of how the intracellular efficacy of tau antibodies can potentially be further improved by certain modifications that aim to enhance tau clearance via specific intracellular degradation pathways.
Collapse
Affiliation(s)
- Erin E Congdon
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, United States.
| | - Yixiang Jiang
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, United States
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, United States; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, United States.
| |
Collapse
|
55
|
Ali Moussa HY, Manaph N, Ali G, Maacha S, Shin KC, Ltaief SM, Gupta V, Tong Y, Ponraj J, Salloum-Asfar S, Mansour S, Al-Shaban FA, Kim HG, Stanton LW, Grivel JC, Abdulla SA, Al-Shammari AR, Park Y. Single Extracellular Vesicle Analysis Using Flow Cytometry for Neurological Disorder Biomarkers. Front Integr Neurosci 2022; 16:879832. [PMID: 35655952 PMCID: PMC9152125 DOI: 10.3389/fnint.2022.879832] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane vesicles released from cells to the extracellular space, involved in cell-to-cell communication by the horizontal transfer of biomolecules such as proteins and RNA. Because EVs can cross the blood-brain barrier (BBB), circulating through the bloodstream and reflecting the cell of origin in terms of disease prognosis and severity, the contents of plasma EVs provide non-invasive biomarkers for neurological disorders. However, neuronal EV markers in blood plasma remain unclear. EVs are very heterogeneous in size and contents, thus bulk analyses of heterogeneous plasma EVs using Western blot and ELISA have limited utility. In this study, using flow cytometry to analyze individual neuronal EVs, we show that our plasma EVs isolated by size exclusion chromatography are mainly CD63-positive exosomes of endosomal origin. As a neuronal EV marker, neural cell adhesion molecule (NCAM) is highly enriched in EVs released from induced pluripotent stem cells (iPSCs)-derived cortical neurons and brain organoids. We identified the subpopulations of plasma EVs that contain NCAM using flow cytometry-based individual EV analysis. Our results suggest that plasma NCAM-positive neuronal EVs can be used to discover biomarkers for neurological disorders.
Collapse
Affiliation(s)
- Houda Yasmine Ali Moussa
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Nimshitha Manaph
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Gowher Ali
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Selma Maacha
- Deep Phenotyping Core, Research Branch, Sidra Medicine, Doha, Qatar
| | - Kyung Chul Shin
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Samia M. Ltaief
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Vijay Gupta
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Yongfeng Tong
- Qatar Environment and Energy Research Institute (QEERI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Janarthanan Ponraj
- Qatar Environment and Energy Research Institute (QEERI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Salam Salloum-Asfar
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Said Mansour
- Qatar Environment and Energy Research Institute (QEERI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Fouad A. Al-Shaban
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Hyung-Goo Kim
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Lawrence W. Stanton
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | | | - Sara A. Abdulla
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Abeer R. Al-Shammari
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- Abeer R. Al-Shammari,
| | - Yongsoo Park
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- *Correspondence: Yongsoo Park,
| |
Collapse
|
56
|
Younas N, Fernandez Flores LC, Hopfner F, Höglinger GU, Zerr I. A new paradigm for diagnosis of neurodegenerative diseases: peripheral exosomes of brain origin. Transl Neurodegener 2022; 11:28. [PMID: 35527262 PMCID: PMC9082915 DOI: 10.1186/s40035-022-00301-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/09/2022] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases are a heterogeneous group of maladies, characterized by progressive loss of neurons. These diseases involve an intricate pattern of cross-talk between different types of cells to maintain specific signaling pathways. A component of such intercellular cross-talk is the exchange of various types of extracellular vesicles (EVs). Exosomes are a subset of EVs, which are increasingly being known for the role they play in the pathogenesis and progression of neurodegenerative diseases, e.g., synucleinopathies and tauopathies. The ability of the central nervous system exosomes to cross the blood–brain barrier into blood has generated enthusiasm in their study as potential biomarkers. However, the lack of standardized, efficient, and ultra-sensitive methods for the isolation and detection of brain-derived exosomes has hampered the development of effective biomarkers. Exosomes mirror heterogeneous biological changes that occur during the progression of these incurable illnesses, potentially offering a more comprehensive outlook of neurodegenerative disease diagnosis, progression and treatment. In this review, we aim to discuss the challenges and opportunities of peripheral biofluid-based brain-exosomes in the diagnosis and biomarker discovery of Alzheimer’s and Parkinson’s diseases. In the later part, we discuss the traditional and emerging methods used for the isolation of exosomes and compare their advantages and disadvantages in clinical settings.
Collapse
|
57
|
Ferrer I, Andrés-Benito P, Garcia-Esparcia P, López-Gonzalez I, Valiente D, Jordán-Pirla M, Carmona M, Sala-Jarque J, Gil V, del Rio JA. Differences in Tau Seeding in Newborn and Adult Wild-Type Mice. Int J Mol Sci 2022; 23:4789. [PMID: 35563179 PMCID: PMC9099670 DOI: 10.3390/ijms23094789] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 12/10/2022] Open
Abstract
Alzheimer's disease (AD) and other tauopathies are common neurodegenerative diseases in older adults; in contrast, abnormal tau deposition in neurons and glial cells occurs only exceptionally in children. Sarkosyl-insoluble fractions from sporadic AD (sAD) containing paired helical filaments (PHFs) were inoculated unilaterally into the thalamus in newborn and three-month-old wild-type C57BL/6 mice, which were killed at different intervals from 24 h to six months after inoculation. Tau-positive cells were scanty and practically disappeared at three months in mice inoculated at the age of a newborn. In contrast, large numbers of tau-positive cells, including neurons and oligodendrocytes, were found in the thalamus of mice inoculated at three months and killed at the ages of six months and nine months. Mice inoculated at the age of newborn and re-inoculated at the age of three months showed similar numbers and distribution of positive cells in the thalamus at six months and nine months. This study shows that (a) differences in tau seeding between newborn and young adults may be related to the ratios between 3Rtau and 4Rtau, and the shift to 4Rtau predominance in adults, together with the immaturity of connections in newborn mice, and (b) intracerebral inoculation of sAD PHFs in newborn mice does not protect from tau seeding following intracerebral inoculation of sAD PHFs in young/adult mice.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; (P.A.-B.); (P.G.-E.); (I.L.-G.); (D.V.); (M.J.-P.); (M.C.)
- Bellvitge Biomedical Research Centre—IDIBELL, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases—CIBERNED, Institute of Health Carlos III, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | - Pol Andrés-Benito
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; (P.A.-B.); (P.G.-E.); (I.L.-G.); (D.V.); (M.J.-P.); (M.C.)
- Bellvitge Biomedical Research Centre—IDIBELL, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases—CIBERNED, Institute of Health Carlos III, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | - Paula Garcia-Esparcia
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; (P.A.-B.); (P.G.-E.); (I.L.-G.); (D.V.); (M.J.-P.); (M.C.)
- Bellvitge Biomedical Research Centre—IDIBELL, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases—CIBERNED, Institute of Health Carlos III, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | - Irene López-Gonzalez
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; (P.A.-B.); (P.G.-E.); (I.L.-G.); (D.V.); (M.J.-P.); (M.C.)
- Bellvitge Biomedical Research Centre—IDIBELL, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases—CIBERNED, Institute of Health Carlos III, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | - Diego Valiente
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; (P.A.-B.); (P.G.-E.); (I.L.-G.); (D.V.); (M.J.-P.); (M.C.)
- Bellvitge Biomedical Research Centre—IDIBELL, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases—CIBERNED, Institute of Health Carlos III, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | - Mónica Jordán-Pirla
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; (P.A.-B.); (P.G.-E.); (I.L.-G.); (D.V.); (M.J.-P.); (M.C.)
- Bellvitge Biomedical Research Centre—IDIBELL, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases—CIBERNED, Institute of Health Carlos III, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | - Margarita Carmona
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain; (P.A.-B.); (P.G.-E.); (I.L.-G.); (D.V.); (M.J.-P.); (M.C.)
- Bellvitge Biomedical Research Centre—IDIBELL, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
- Network Centre of Biomedical Research of Neurodegenerative Diseases—CIBERNED, Institute of Health Carlos III, Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | - Julia Sala-Jarque
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia, Barcelona Institute for Science and Technology, Parc Científic de Barcelona, Baldiri Reixac sn, 08020 Barcelona, Spain; (J.S.-J.); (V.G.); (J.A.d.R.)
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Baldiri Reixac sn, 08020 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Vanessa Gil
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia, Barcelona Institute for Science and Technology, Parc Científic de Barcelona, Baldiri Reixac sn, 08020 Barcelona, Spain; (J.S.-J.); (V.G.); (J.A.d.R.)
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Baldiri Reixac sn, 08020 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - José Antonio del Rio
- Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia, Barcelona Institute for Science and Technology, Parc Científic de Barcelona, Baldiri Reixac sn, 08020 Barcelona, Spain; (J.S.-J.); (V.G.); (J.A.d.R.)
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Baldiri Reixac sn, 08020 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| |
Collapse
|
58
|
Mato-Basalo R, Lucio-Gallego S, Alarcón-Veleiro C, Sacristán-Santos M, Quintana MDPM, Morente-López M, de Toro FJ, Silva-Fernández L, González-Rodríguez A, Arufe MC, Labora JAF. Action Mechanisms of Small Extracellular Vesicles in Inflammaging. Life (Basel) 2022; 12:life12040546. [PMID: 35455036 PMCID: PMC9028066 DOI: 10.3390/life12040546] [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: 03/14/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 02/05/2023] Open
Abstract
The accumulation process of proinflammatory components in the body due to aging influences intercellular communication and is known as inflammaging. This biological mechanism relates the development of inflammation to the aging process. Recently, it has been reported that small extracellular vesicles (sEVs) are mediators in the transmission of paracrine senescence involved in inflammatory aging. For this reason, their components, as well as mechanisms of action of sEVs, are relevant to develop a new therapy called senodrugs (senolytics and senomorphic) that regulates the intercellular communication of inflammaging. In this review, we include the most recent and relevant studies on the role of sEVs in the inflammatory aging process and in age-related diseases such as cancer and type 2 diabetes.
Collapse
|
59
|
Li TR, Yang Q, Hu X, Han Y. Biomarkers and Tools for Predicting Alzheimer's Disease in the Preclinical Stage. Curr Neuropharmacol 2022; 20:713-737. [PMID: 34030620 PMCID: PMC9878962 DOI: 10.2174/1570159x19666210524153901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/27/2021] [Accepted: 05/08/2021] [Indexed: 11/22/2022] Open
Abstract
Alzheimer's disease (AD) is the only leading cause of death for which no disease-modifying therapy is currently available. Over the past decade, a string of disappointing clinical trial results has forced us to shift our focus to the preclinical stage of AD, which represents the most promising therapeutic window. However, the accurate diagnosis of preclinical AD requires the presence of brain β- amyloid deposition determined by cerebrospinal fluid or amyloid-positron emission tomography, significantly limiting routine screening and diagnosis in non-tertiary hospital settings. Thus, an easily accessible marker or tool with high sensitivity and specificity is highly needed. Recently, it has been discovered that individuals in the late stage of preclinical AD may not be truly "asymptomatic" in that they may have already developed subtle or subjective cognitive decline. In addition, advances in bloodderived biomarker studies have also allowed the detection of pathologic changes in preclinical AD. Exosomes, as cell-to-cell communication messengers, can reflect the functional changes of their source cell. Methodological advances have made it possible to extract brain-derived exosomes from peripheral blood, making exosomes an emerging biomarker carrier and liquid biopsy tool for preclinical AD. The eye and its associated structures have rich sensory-motor innervation. In this regard, studies have indicated that they may also provide reliable markers. Here, our report covers the current state of knowledge of neuropsychological and eye tests as screening tools for preclinical AD and assesses the value of blood and brain-derived exosomes as carriers of biomarkers in conjunction with the current diagnostic paradigm.
Collapse
Affiliation(s)
- Tao-Ran Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Qin Yang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Xiaochen Hu
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, 50924, Germany
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China;,Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China;,National Clinical Research Center for Geriatric Disorders, Beijing, 100053, China;,School of Biomedical Engineering, Hainan University, Haikou, 570228, China;,Address correspondence to this author at the Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China; Tel: +86 13621011941; E-mail:
| |
Collapse
|
60
|
Nicholson S, Baccarelli A, Prada D. Role of brain extracellular vesicles in air pollution-related cognitive impairment and neurodegeneration. ENVIRONMENTAL RESEARCH 2022; 204:112316. [PMID: 34728237 PMCID: PMC8671239 DOI: 10.1016/j.envres.2021.112316] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 09/15/2021] [Accepted: 10/28/2021] [Indexed: 05/07/2023]
Abstract
A relationship between environmental exposure to air pollution and cognitive impairment and neurological disorders has been described. Previous literature has focused on the direct effects of the air pollution components on neuronal and glial cells, as well as on involvement of oxidative stress and neuroinflammation on microglia and astrocyte reactivity. However, other mechanisms involved in the air pollution effects on central nervous system (CNS) toxicity can be playing critical roles. Increasingly, extracellular vesicle's (EVs) mediated intercellular communication is being recognized as impacting the development of cognitive impairment and neurological disorders like Alzheimer's disease and others. Here we describe the available evidence about toxic air pollutants and its components on brain, an involvement of brain cells specific and EVs types (based in the origin or in the size of EVs) in the initiation, exacerbation, and propagation of the neurotoxic effects (inflammation, neurodegeneration, and accumulation of neurotoxic proteins) induced by air pollution in the CNS. Additionally, we discuss the identification and isolation of neural-derived EVs from human plasma, the most common markers for neural-derived EVs, and their potential for use as diagnostic or therapeutic molecules for air pollution-related cognitive impairment and neurodegeneration.
Collapse
Affiliation(s)
- Stacia Nicholson
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, 10032, USA
| | - Andrea Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, 10032, USA
| | - Diddier Prada
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, 10032, USA; Instituto Nacional de Cancerología, Mexico City, 14080, Mexico.
| |
Collapse
|
61
|
Tracy TE, Madero-Pérez J, Swaney DL, Chang TS, Moritz M, Konrad C, Ward ME, Stevenson E, Hüttenhain R, Kauwe G, Mercedes M, Sweetland-Martin L, Chen X, Mok SA, Wong MY, Telpoukhovskaia M, Min SW, Wang C, Sohn PD, Martin J, Zhou Y, Luo W, Trojanowski JQ, Lee VMY, Gong S, Manfredi G, Coppola G, Krogan NJ, Geschwind DH, Gan L. Tau interactome maps synaptic and mitochondrial processes associated with neurodegeneration. Cell 2022; 185:712-728.e14. [PMID: 35063084 PMCID: PMC8857049 DOI: 10.1016/j.cell.2021.12.041] [Citation(s) in RCA: 161] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/20/2021] [Accepted: 12/22/2021] [Indexed: 12/21/2022]
Abstract
Tau (MAPT) drives neuronal dysfunction in Alzheimer disease (AD) and other tauopathies. To dissect the underlying mechanisms, we combined an engineered ascorbic acid peroxidase (APEX) approach with quantitative affinity purification mass spectrometry (AP-MS) followed by proximity ligation assay (PLA) to characterize Tau interactomes modified by neuronal activity and mutations that cause frontotemporal dementia (FTD) in human induced pluripotent stem cell (iPSC)-derived neurons. We established interactions of Tau with presynaptic vesicle proteins during activity-dependent Tau secretion and mapped the Tau-binding sites to the cytosolic domains of integral synaptic vesicle proteins. We showed that FTD mutations impair bioenergetics and markedly diminished Tau’s interaction with mitochondria proteins, which were downregulated in AD brains of multiple cohorts and correlated with disease severity. These multimodal and dynamic Tau interactomes with exquisite spatial resolution shed light on Tau’s role in neuronal function and disease and highlight potential therapeutic targets to block Tau-mediated pathogenesis. By combining APEX and AP-MS proteomic approaches, Tau interactome mapping reveals that Tau interactors are modified by neuronal activity and FTD mutations in human iPSC-derived neurons.
Collapse
Affiliation(s)
- Tara E Tracy
- Gladstone Institutes, San Francisco, CA 94158, USA; Buck Institute for Research on Aging, Novato, CA 94945, USA.
| | - Jesus Madero-Pérez
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.
| | - Danielle L Swaney
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Timothy S Chang
- Department of Neurology, Movement Disorders Program and Program in Neurogenetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Michelle Moritz
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA
| | - Csaba Konrad
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Erica Stevenson
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ruth Hüttenhain
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA
| | - Grant Kauwe
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Maria Mercedes
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Lauren Sweetland-Martin
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Xu Chen
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Sue-Ann Mok
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Man Ying Wong
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | | | - Sang-Won Min
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Chao Wang
- Gladstone Institutes, San Francisco, CA 94158, USA
| | | | | | - Yungui Zhou
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Virginia M Y Lee
- Center for Neurodegenerative Disease Research, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shiaoching Gong
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Giovanni Coppola
- Department of Neurology, Movement Disorders Program and Program in Neurogenetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA 90095, USA
| | - Nevan J Krogan
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA
| | - Daniel H Geschwind
- Department of Neurology, Movement Disorders Program and Program in Neurogenetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute of Precision Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.
| |
Collapse
|
62
|
Leroux E, Perbet R, Caillierez R, Richetin K, Lieger S, Espourteille J, Bouillet T, Bégard S, Danis C, Loyens A, Toni N, Déglon N, Deramecourt V, Schraen-Maschke S, Buée L, Colin M. Extracellular vesicles: Major actors of heterogeneity in tau spreading among human tauopathies. Mol Ther 2022; 30:782-797. [PMID: 34563677 PMCID: PMC8821971 DOI: 10.1016/j.ymthe.2021.09.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/12/2021] [Accepted: 09/20/2021] [Indexed: 02/04/2023] Open
Abstract
Tauopathies are neurodegenerative diseases characterized by tau inclusions in brain cells. Seed-competent tau species have been suggested to spread from cell to cell in a stereotypical manner, indicating that this may involve a prion-like mechanism. Although the intercellular mechanisms of transfer are unclear, extracellular vesicles (EVs) could be potential shuttles. We assessed this in humans by preparing vesicles from fluids (brain-derived enriched EVs [BD-EVs]). These latter were isolated from different brain regions in various tauopathies, and their seeding potential was assessed in vitro and in vivo. We observed considerable heterogeneity among tauopathies and brain regions. The most striking evidence was coming mainly from Alzheimer's disease where the BD-EVs clearly contain pathological species that can induce tau lesions in vivo. The results support the hypothesis that BD-EVs participate in the prion-like propagation of tau pathology among tauopathies, and there may be implications for diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Elodie Leroux
- Université de Lille, INSERM, CHU-Lille, Lille Neuroscience & Cognition, 59000 Lille, France
| | - Romain Perbet
- Université de Lille, INSERM, CHU-Lille, Lille Neuroscience & Cognition, 59000 Lille, France
| | - Raphaëlle Caillierez
- Université de Lille, INSERM, CHU-Lille, Lille Neuroscience & Cognition, 59000 Lille, France
| | - Kevin Richetin
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland,Lausanne University Hospital (CHUV) and University of Lausanne, Neuroscience Research Center (CRN), Laboratory of Cellular and Molecular Neurotherapies, 1011 Lausanne, Switzerland,Lausanne University Hospital (CHUV) and University of Lausanne, Department of Clinical Neuroscience (DNC), Laboratory of Cellular and Molecular Neurotherapies, 1011 Lausanne, Switzerland
| | - Sarah Lieger
- Université de Lille, INSERM, CHU-Lille, Lille Neuroscience & Cognition, 59000 Lille, France
| | - Jeanne Espourteille
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
| | - Thomas Bouillet
- Université de Lille, INSERM, CHU-Lille, Lille Neuroscience & Cognition, 59000 Lille, France
| | - Séverine Bégard
- Université de Lille, INSERM, CHU-Lille, Lille Neuroscience & Cognition, 59000 Lille, France
| | - Clément Danis
- Université de Lille, INSERM, CHU-Lille, Lille Neuroscience & Cognition, 59000 Lille, France
| | - Anne Loyens
- Université de Lille, INSERM, CHU-Lille, Lille Neuroscience & Cognition, 59000 Lille, France
| | - Nicolas Toni
- Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, 1011 Lausanne, Switzerland
| | - Nicole Déglon
- Lausanne University Hospital (CHUV) and University of Lausanne, Neuroscience Research Center (CRN), Laboratory of Cellular and Molecular Neurotherapies, 1011 Lausanne, Switzerland,Lausanne University Hospital (CHUV) and University of Lausanne, Department of Clinical Neuroscience (DNC), Laboratory of Cellular and Molecular Neurotherapies, 1011 Lausanne, Switzerland
| | - Vincent Deramecourt
- Université de Lille, INSERM, CHU-Lille, Lille Neuroscience & Cognition, 59000 Lille, France
| | | | - Luc Buée
- Université de Lille, INSERM, CHU-Lille, Lille Neuroscience & Cognition, 59000 Lille, France,Corresponding author: Luc Buée, PhD, Université de Lille, INSERM, CHU-Lille, Lille Neuroscience & Cognition, Bâtiment Biserte, rue Polonovski, 59045 Lille Cedex, France.
| | - Morvane Colin
- Université de Lille, INSERM, CHU-Lille, Lille Neuroscience & Cognition, 59000 Lille, France,Corresponding author: Morvane Colin, Université de Lille, INSERM, CHU-Lille, Lille Neuroscience & Cognition, Bâtiment Biserte, rue Polonovski, 59045 Lille Cedex, France.
| |
Collapse
|
63
|
Longobardi A, Nicsanu R, Bellini S, Squitti R, Catania M, Tiraboschi P, Saraceno C, Ferrari C, Zanardini R, Binetti G, Di Fede G, Benussi L, Ghidoni R. Cerebrospinal Fluid EV Concentration and Size Are Altered in Alzheimer’s Disease and Dementia with Lewy Bodies. Cells 2022; 11:cells11030462. [PMID: 35159272 PMCID: PMC8834088 DOI: 10.3390/cells11030462] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/21/2022] [Accepted: 01/28/2022] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD), dementia with Lewy bodies (DLB) and frontotemporal dementia (FTD) represent the three major neurodegenerative dementias characterized by abnormal brain protein accumulation. In this study, we investigated extracellular vesicles (EVs) and neurotrophic factors in the cerebrospinal fluid (CSF) of 120 subjects: 36 with AD, 30 with DLB, 34 with FTD and 20 controls. Specifically, CSF EVs were analyzed by Nanoparticle Tracking Analysis and neurotrophic factors were measured with ELISA. We found higher EV concentration and lower EV size in AD and DLB groups compared to the controls. Classification tree analysis demonstrated EV size as the best parameter able to discriminate the patients from the controls (96.7% vs. 3.3%, respectively). The diagnostic performance of the EV concentration/size ratio resulted in a fair discrimination level with an area under the curve of 0.74. Moreover, the EV concentration/size ratio was associated with the p-Tau181/Aβ42 ratio in AD patients. In addition, we described altered levels of cystatin C and progranulin in the DLB and AD groups. We did not find any correlation between neurotrophic factors and EV parameters. In conclusion, the results of this study suggest a common involvement of the endosomal pathway in neurodegenerative dementias, giving important insight into the molecular mechanisms underlying these pathologies.
Collapse
Affiliation(s)
- Antonio Longobardi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
| | - Roland Nicsanu
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
| | - Sonia Bellini
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
| | - Rosanna Squitti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
| | - Marcella Catania
- Neurology 5 and Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (M.C.); (P.T.); (G.D.F.)
| | - Pietro Tiraboschi
- Neurology 5 and Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (M.C.); (P.T.); (G.D.F.)
| | - Claudia Saraceno
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
| | - Clarissa Ferrari
- Service of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy;
| | - Roberta Zanardini
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
| | - Giuliano Binetti
- MAC Memory Clinic and Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy;
| | - Giuseppe Di Fede
- Neurology 5 and Neuropathology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (M.C.); (P.T.); (G.D.F.)
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (A.L.); (R.N.); (S.B.); (R.S.); (C.S.); (R.Z.); (L.B.)
- Correspondence: ; Tel.: +39-030-3501725
| |
Collapse
|
64
|
Alvarez XA, Winston CN, Barlow JW, Sarsoza FM, Alvarez I, Aleixandre M, Linares C, García-Fantini M, Kastberger B, Winter S, Rissman RA. Modulation of Amyloid-β and Tau in Alzheimer's Disease Plasma Neuronal-Derived Extracellular Vesicles by Cerebrolysin® and Donepezil. J Alzheimers Dis 2022; 90:705-717. [PMID: 36155516 PMCID: PMC9697063 DOI: 10.3233/jad-220575] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Plasma neuronal-derived extracellular vesicles (NDEV) contain proteins of pathological, diagnostic, and therapeutic relevance. OBJECTIVE We investigated the associations of six plasma NDEV markers with Alzheimer's disease (AD) severity, cognition and functioning, and changes in these biomarkers after Cerebrolysin®, donepezil, and a combination therapy in AD. METHODS Plasma NDEV levels of Aβ42, total tau, P-T181-tau, P-S393-tau, neurogranin, and REST were determined in: 1) 116 mild to advanced AD patients and in 20 control subjects; 2) 110 AD patients treated with Cerebrolysin®, donepezil, or combination therapy in a randomized clinical trial (RCT). Samples for NDEV determinations were obtained at baseline in the NDEV study and at baseline and study endpoint in the RCT. Cognition and functioning were assessed at the same time points. RESULTS NDEV levels of Aβ42, total tau, P-T181-tau, and P-S393-tau were higher and those of neurogranin and REST were lower in mild-to-moderate AD than in controls (p < 0.05 to p < 0.001). NDEV total tau, neurogranin, and REST increased with AD severity (p < 0.05 to p < 0.001). NDEV Aβ42 and P-T181-tau correlated negatively with serum BDNF (p < 0.05), and total-tau levels were associated to plasma TNF-α (p < 0.01) and cognitive impairment (p < 0.05). Combination therapy reduced NDEV Aβ42 with respect to monotherapies (p < 0.05); and NDEV total tau, P-T181-tau, and P-S396-tau were decreased in Cerebrolysin-treated patients compared to those on donepezil monotherapy (p < 0.05). CONCLUSION The present results demonstrate the utility of NDEV determinations of pathologic and synaptic proteins as effective AD biomarkers, as markers of AD severity, and as potential tools for monitoring the effects of anti-AD drugs.
Collapse
Affiliation(s)
- X. Anton Alvarez
- Medinova Institute of Neurosciences, Clinica Reha Salud, A Coruña, Spain
- Clinical Research Department, QPS Holdings, A Coruña, Spain
| | | | - James W. Barlow
- Department of Neurosciences, University of California, San Diego, CA, USA
| | - Floyd M. Sarsoza
- Department of Neurosciences, University of California, San Diego, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Irene Alvarez
- Medinova Institute of Neurosciences, Clinica Reha Salud, A Coruña, Spain
| | | | | | | | | | | | - Robert A. Rissman
- Department of Neurosciences, University of California, San Diego, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
65
|
Gabrielli M, Prada I, Joshi P, Falcicchia C, D’Arrigo G, Rutigliano G, Battocchio E, Zenatelli R, Tozzi F, Radeghieri A, Arancio O, Origlia N, Verderio C. OUP accepted manuscript. Brain 2022; 145:2849-2868. [PMID: 35254410 PMCID: PMC9420022 DOI: 10.1093/brain/awac083] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 01/17/2022] [Accepted: 02/13/2022] [Indexed: 11/27/2022] Open
Abstract
Synaptic dysfunction is an early mechanism in Alzheimer’s disease that involves progressively larger areas of the brain over time. However, how it starts and propagates is unknown. Here we show that amyloid-β released by microglia in association with large extracellular vesicles (Aβ-EVs) alters dendritic spine morphology in vitro, at the site of neuron interaction, and impairs synaptic plasticity both in vitro and in vivo in the entorhinal cortex–dentate gyrus circuitry. One hour after Aβ-EV injection into the mouse entorhinal cortex, long-term potentiation was impaired in the entorhinal cortex but not in the dentate gyrus, its main target region, while 24 h later it was also impaired in the dentate gyrus, revealing a spreading of long-term potentiation deficit between the two regions. Similar results were obtained upon injection of extracellular vesicles carrying Aβ naturally secreted by CHO7PA2 cells, while neither Aβ42 alone nor inflammatory extracellular vesicles devoid of Aβ were able to propagate long-term potentiation impairment. Using optical tweezers combined to time-lapse imaging to study Aβ-EV–neuron interaction, we show that Aβ-EVs move anterogradely at the axon surface and that their motion can be blocked through annexin-V coating. Importantly, when Aβ-EV motility was inhibited, no propagation of long-term potentiation deficit occurred along the entorhinal–hippocampal circuit, implicating large extracellular vesicle motion at the neuron surface in the spreading of long-term potentiation impairment. Our data indicate the involvement of large microglial extracellular vesicles in the rise and propagation of early synaptic dysfunction in Alzheimer’s disease and suggest a new mechanism controlling the diffusion of large extracellular vesicles and their pathogenic signals in the brain parenchyma, paving the way for novel therapeutic strategies to delay the disease.
Collapse
Affiliation(s)
| | - Ilaria Prada
- CNR Institute of Neuroscience, Vedano al Lambro, MB 20854, Italy
| | - Pooja Joshi
- CNR Institute of Neuroscience, Vedano al Lambro, MB 20854, Italy
| | | | - Giulia D’Arrigo
- CNR Institute of Neuroscience, Vedano al Lambro, MB 20854, Italy
| | - Grazia Rutigliano
- Institute of Life Sciences, Sant’Anna School of Advanced Studies, Pisa 56127, Italy
- CNR Institute of Clinical Physiology, Pisa 56124, Italy
| | - Elisabetta Battocchio
- CNR Institute of Neuroscience, Vedano al Lambro, MB 20854, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Monza 20900, Italy
| | - Rossella Zenatelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
| | - Francesca Tozzi
- Bio@SNS Laboratory, Scuola Normale Superiore, Pisa, 56124, Italy
| | - Annalisa Radeghieri
- Department of Molecular and Translational Medicine, University of Brescia, Brescia 25123, Italy
- Consorzio Sistemi a Grande Interfase (CSGI), Department of Chemistry, University of Florence, Sesto Fiorentino, FI 50019, Italy
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York 10032, NY, USA
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Nicola Origlia
- Correspondence may also be addressed to: Nicola Origlia CNR Institute of Neuroscience, via Moruzzi 1 Pisa, 56124, Italy E-mail:
| | - Claudia Verderio
- Correspondence to: Claudia Verderio CNR Institute of Neuroscience via Raoul Follereau 3, Vedano al Lambro MB, 20854, Italy E-mail:
| |
Collapse
|
66
|
You Y, Muraoka S, Jedrychowski MP, Hu J, McQuade AK, Young‐Pearse T, Aslebagh R, Shaffer SA, Gygi SP, Blurton‐Jones M, Poon WW, Ikezu T. Human neural cell type-specific extracellular vesicle proteome defines disease-related molecules associated with activated astrocytes in Alzheimer's disease brain. J Extracell Vesicles 2022; 11:e12183. [PMID: 35029059 PMCID: PMC8758831 DOI: 10.1002/jev2.12183] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/10/2021] [Indexed: 12/30/2022] Open
Abstract
In neurodegenerative diseases, extracellular vesicles (EVs) transfer pathogenic molecules and are consequently involved in disease progression. We have investigated the proteomic profiles of EVs that were isolated from four different human-induced pluripotent stem cell-derived neural cell types (excitatory neurons, astrocytes, microglia-like cells, and oligodendrocyte-like cells). Novel cell type-specific EV protein markers were then identified for the excitatory neurons (ATP1A3, NCAM1), astrocytes (LRP1, ITGA6), microglia-like cells (ITGAM, LCP1), and oligodendrocyte-like cells (LAMP2, FTH1), as well as 16 pan-EV marker candidates, including integrins and annexins. To further demonstrate how cell-type-specific EVs may be involved in Alzheimer's disease (AD), we performed protein co-expression network analysis and conducted cell type assessments for the proteomes of brain-derived EVs from the control, mild cognitive impairment, and AD cases. A protein module enriched in astrocyte-specific EV markers was most significantly associated with the AD pathology and cognitive impairment, suggesting an important role in AD progression. The hub protein from this module, integrin-β1 (ITGB1), was found to be significantly elevated in astrocyte-specific EVs enriched from the total brain-derived AD EVs and associated with the brain β-amyloid and tau load in independent cohorts. Thus, our study provides a featured framework and rich resource for the future analyses of EV functions in neurodegenerative diseases in a cell type-specific manner.
Collapse
Affiliation(s)
- Yang You
- Department of Pharmacology & Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
- Department of NeuroscienceMayo Clinic FloridaJacksonvilleFloridaUSA
| | - Satoshi Muraoka
- Department of Pharmacology & Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
| | | | - Jianqiao Hu
- Department of Pharmacology & Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
| | - Amanda K. McQuade
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Tracy Young‐Pearse
- Department of Neurology, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Roshanak Aslebagh
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
- Mass Spectrometry FacilityUniversity of Massachusetts Medical SchoolShrewsburyMassachusettsUSA
| | - Scott A. Shaffer
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
- Mass Spectrometry FacilityUniversity of Massachusetts Medical SchoolShrewsburyMassachusettsUSA
| | - Steven P. Gygi
- Department of Cell BiologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Mathew Blurton‐Jones
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Wayne W. Poon
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological DisordersUniversity of CaliforniaIrvineCaliforniaUSA
| | - Tsuneya Ikezu
- Department of Pharmacology & Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
- Department of NeuroscienceMayo Clinic FloridaJacksonvilleFloridaUSA
- The Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
67
|
Tallon C, Picciolini S, Yoo SW, Thomas AG, Pal A, Alt J, Carlomagno C, Gualerzi A, Rais R, Haughey NJ, Bedoni M, Slusher BS. Inhibition of neutral sphingomyelinase 2 reduces extracellular vesicle release from neurons, oligodendrocytes, and activated microglial cells following acute brain injury. Biochem Pharmacol 2021; 194:114796. [PMID: 34678224 DOI: 10.1016/j.bcp.2021.114796] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 12/16/2022]
Abstract
Extracellular Vesicles (EVs) are implicated in the spread of pathogenic proteinsin a growing number of neurological diseases. Given this, there is rising interest in developing inhibitors of Neutral Sphingomyelinase 2 (nSMase2), an enzyme critical in EV biogenesis. Our group recently discovered phenyl(R)-(1-(3-(3,4-dimethoxyphenyl)-2,6-dimethylimidazo[1,2-b]pyridazin-8-yl)pyrrolidin-3-yl)carbamate (PDDC), the first potent, selective, orally-available, and brain-penetrable nSMase2 inhibitor, capable of dose-dependently reducing EVs release in vitro and in vivo. Herein, using multiplexed Surface Plasmon Resonance imaging (SPRi), we evaluated which brain cell-derived EVs were affected by PDDC following acute brain injury. Mice were fed PDDC-containing chow at doses which gave steady PDDC brain exposures exceeding its nSMase2 IC50. Mice were then administered an intra-striatal IL-1β injection and two hours later plasma and brain were collected. IL-1β injection significantly increased striatal nSMase2 activity which was completely normalized by PDDC. Using SPRi, we found that IL-1β-induced injury selectively increased plasma levels of CD171 + and PLP1 + EVs; this EV increase was normalized by PDDC. In contrast, GLAST1 + EVs were unchanged by IL-1β or PDDC. IL-1β injection selectively increased EVs released from activated versus non-activated microglia, indicated by the CD11b+/IB4 + ratio. The increase in EVs from CD11b + microglia was dramatically attenuated with PDDC. Taken together, our data demonstrate that following acute injury, brain nSMase2 activity is elevated. EVs released from neurons, oligodendrocytes, and activated microglial are increased in plasma and inhibition of nSMase2 with PDDC reduced these IL-1β-induced changes implicating nSMase2 inhibition as a therapeutic target for acute brain injury.
Collapse
Affiliation(s)
- Carolyn Tallon
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Silvia Picciolini
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Laboratory of Nanomedicine and Clinical Biophotonics (LABION), Milan, Italy
| | - Seung-Wan Yoo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ajit G Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Arindom Pal
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jesse Alt
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Cristiano Carlomagno
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Laboratory of Nanomedicine and Clinical Biophotonics (LABION), Milan, Italy
| | - Alice Gualerzi
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Laboratory of Nanomedicine and Clinical Biophotonics (LABION), Milan, Italy
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Marzia Bedoni
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Laboratory of Nanomedicine and Clinical Biophotonics (LABION), Milan, Italy.
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
68
|
Koychev I, Jansen K, Dette A, Shi L, Holling H. Blood-Based ATN Biomarkers of Alzheimer's Disease: A Meta-Analysis. J Alzheimers Dis 2021; 79:177-195. [PMID: 33252080 DOI: 10.3233/jad-200900] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The Amyloid Tau Neurodegeneration (ATN) framework was proposed to define the biological state underpinning Alzheimer's disease (AD). Blood-based biomarkers offer a scalable alternative to the costly and invasive currently available biomarkers. OBJECTIVE In this meta-analysis we sought to assess the diagnostic performance of plasma amyloid (Aβ40, Aβ42, Aβ42/40 ratio), tangle (p-tau181), and neurodegeneration (total tau [t-tau], neurofilament light [NfL]) biomarkers. METHODS Electronic databases were screened for studies reporting biomarker concentrations for AD and control cohorts. Biomarker performance was examined by random-effect meta-analyses based on the ratio between biomarker concentrations in patients and controls. RESULTS 83 studies published between 1996 and 2020 were included in the analyses. Aβ42/40 ratio as well as Aβ42 discriminated AD patients from controls when using novel platforms such as immunomagnetic reduction (IMR). We found significant differences in ptau-181 concentration for studies based on single molecule array (Simoa), but not for studies based on IMR or ELISA. T-tau was significantly different between AD patients and control in IMR and Simoa but not in ELISA-based studies. In contrast, NfL differentiated between groups across platforms. Exosome studies showed strong separation between patients and controls for Aβ42, t-tau, and p-tau181. CONCLUSION Currently available assays for sampling plasma ATN biomarkers appear to differentiate between AD patients and controls. Novel assay methodologies have given the field a significant boost for testing these biomarkers, such as IMR for Aβ, Simoa for p-tau181. Enriching samples through extracellular vesicles shows promise but requires further validation.
Collapse
Affiliation(s)
- Ivan Koychev
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Katrin Jansen
- Department of Psychology, University of Münster, Münster, Germany
| | - Alina Dette
- Department of Psychology, University of Münster, Münster, Germany
| | - Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Heinz Holling
- Department of Psychology, University of Münster, Münster, Germany
| |
Collapse
|
69
|
Ledreux A, Thomas S, Hamlett ED, Trautman C, Gilmore A, Rickman Hager E, Paredes DA, Margittai M, Fortea J, Granholm AC. Small Neuron-Derived Extracellular Vesicles from Individuals with Down Syndrome Propagate Tau Pathology in the Wildtype Mouse Brain. J Clin Med 2021; 10:3931. [PMID: 34501378 PMCID: PMC8432237 DOI: 10.3390/jcm10173931] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/24/2021] [Accepted: 08/28/2021] [Indexed: 12/11/2022] Open
Abstract
Individuals with Down syndrome (DS) exhibit Alzheimer's disease (AD) pathology at a young age, including amyloid plaques and neurofibrillary tangles (NFTs). Tau pathology can spread via extracellular vesicles, such as exosomes. The cargo of neuron-derived small extracellular vesicles (NDEVs) from individuals with DS contains p-Tau at an early age. The goal of the study was to investigate whether NDEVs isolated from the blood of individuals with DS can spread Tau pathology in the brain of wildtype mice. We purified NDEVs from the plasma of patients with DS-AD and controls and injected small quantities using stereotaxic surgery into the dorsal hippocampus of adult wildtype mice. Seeding competent Tau conformers were amplified in vitro from DS-AD NDEVs but not NDEVs from controls. One month or 4 months post-injection, we examined Tau pathology in mouse brains. We found abundant p-Tau immunostaining in the hippocampus of the mice injected with DS-AD NDEVs compared to injections of age-matched control NDEVs. Double labeling with neuronal and glial markers showed that p-Tau staining was largely found in neurons and, to a lesser extent, in glial cells and that p-Tau immunostaining was spreading along the corpus callosum and the medio-lateral axis of the hippocampus. These studies demonstrate that NDEVs from DS-AD patients exhibit Tau seeding capacity and give rise to tangle-like intracellular inclusions.
Collapse
Affiliation(s)
- Aurélie Ledreux
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO 80208, USA; (S.T.); (C.T.); (A.G.); (D.A.P.); (A.-C.G.)
| | - Sarah Thomas
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO 80208, USA; (S.T.); (C.T.); (A.G.); (D.A.P.); (A.-C.G.)
| | - Eric D. Hamlett
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Camille Trautman
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO 80208, USA; (S.T.); (C.T.); (A.G.); (D.A.P.); (A.-C.G.)
| | - Anah Gilmore
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO 80208, USA; (S.T.); (C.T.); (A.G.); (D.A.P.); (A.-C.G.)
| | - Emily Rickman Hager
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO 80208, USA; (E.R.H.); (M.M.)
| | - Daniel A. Paredes
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO 80208, USA; (S.T.); (C.T.); (A.G.); (D.A.P.); (A.-C.G.)
| | - Martin Margittai
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO 80208, USA; (E.R.H.); (M.M.)
| | - Juan Fortea
- Hospital de la Santa Creu i Sant Pau and Catalan Down Syndrome Foundation, 08041 Barcelona, Spain;
| | - Ann-Charlotte Granholm
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO 80208, USA; (S.T.); (C.T.); (A.G.); (D.A.P.); (A.-C.G.)
| |
Collapse
|
70
|
Mosquera-Heredia MI, Morales LC, Vidal OM, Barceló E, Silvera-Redondo C, Vélez JI, Garavito-Galofre P. Exosomes: Potential Disease Biomarkers and New Therapeutic Targets. Biomedicines 2021; 9:1061. [PMID: 34440265 PMCID: PMC8393483 DOI: 10.3390/biomedicines9081061] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023] Open
Abstract
Exosomes are extracellular vesicles released by cells, both constitutively and after cell activation, and are present in different types of biological fluid. Exosomes are involved in the pathogenesis of diseases, such as cancer, neurodegenerative diseases, pregnancy disorders and cardiovascular diseases, and have emerged as potential non-invasive biomarkers for the detection, prognosis and therapeutics of a myriad of diseases. In this review, we describe recent advances related to the regulatory mechanisms of exosome biogenesis, release and molecular composition, as well as their role in health and disease, and their potential use as disease biomarkers and therapeutic targets. In addition, the advantages and disadvantages of their main isolation methods, characterization and cargo analysis, as well as the experimental methods used for exosome-mediated drug delivery, are discussed. Finally, we present potential perspectives for the use of exosomes in future clinical practice.
Collapse
Affiliation(s)
- Maria I. Mosquera-Heredia
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| | - Luis C. Morales
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| | - Oscar M. Vidal
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| | - Ernesto Barceló
- Instituto Colombiano de Neuropedagogía, Barranquilla 080020, Colombia;
| | - Carlos Silvera-Redondo
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| | - Jorge I. Vélez
- Department of Industrial Engineering, Universidad del Norte, Barranquilla 081007, Colombia;
| | - Pilar Garavito-Galofre
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| |
Collapse
|
71
|
Mittal R, Bencie N, Langlie J, Mittal J, Eshraghi AA. Exosomes as drug delivery vehicles and biomarkers for neurological and auditory systems. J Cell Physiol 2021; 236:8035-8049. [PMID: 34224589 DOI: 10.1002/jcp.30484] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/19/2021] [Accepted: 06/14/2021] [Indexed: 12/14/2022]
Abstract
Exosomes are small extracellular membrane particles that play a crucial role in intracellular signaling. Research shows that exosomes have the potential to be used as biomarkers or drug delivery systems in specific organs, such as the neurological system and the inner ear. Exosomes in neurological and auditory systems release different molecules when under stress versus in healthy states, highlighting their potential use as biomarkers in the identification of diseased states. Studies have suggested that exosomes can be harnessed for drug delivery to hard-to-reach organs, such as cochlear sensory hair cells and the brain due to their ability to cross the blood-labyrinth and blood-brain barriers. In this article, we describe the biogenesis, classification, and characterization methods of exosomes. We then discuss recent studies that indicate their potential usage as biomarkers and drug delivery systems to help treat inner ear and neurological disorders.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Nicole Bencie
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jake Langlie
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jeenu Mittal
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Adrien A Eshraghi
- Department of Otolaryngology, Cochlear Implant and Hearing Research Laboratory, University of Miami Miller School of Medicine, Miami, Florida, USA.,Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA.,Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
72
|
Kim KY, Shin KY, Chang KA. Brain-Derived Exosomal Proteins as Effective Biomarkers for Alzheimer's Disease: A Systematic Review and Meta-Analysis. Biomolecules 2021; 11:biom11070980. [PMID: 34356604 PMCID: PMC8301985 DOI: 10.3390/biom11070980] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/16/2021] [Accepted: 06/29/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disease, affects approximately 50 million people worldwide, which warrants the search for reliable new biomarkers for early diagnosis of AD. Brain-derived exosomal (BDE) proteins, which are extracellular nanovesicles released by all cell lineages of the central nervous system, have been focused as biomarkers for diagnosis, screening, prognosis prediction, and monitoring in AD. This review focused on the possibility of BDE proteins as AD biomarkers. The articles published prior to 26 January 2021 were searched in PubMed, EMBASE, Web of Science, and Cochrane Library to identify all relevant studies that reported exosome biomarkers in blood samples of patients with AD. From 342 articles, 20 studies were selected for analysis. We conducted a meta-analysis of six BDE proteins and found that levels of amyloid-β42 (standardized mean difference (SMD) = 1.534, 95% confidence interval [CI]: 0.595-2.474), total-tau (SMD = 1.224, 95% CI: 0.534-1.915), tau phosphorylated at threonine 181 (SMD = 4.038, 95% CI: 2.312-5.764), and tau phosphorylated at serine 396 (SMD = 2.511, 95% CI: 0.795-4.227) were significantly different in patients with AD compared to those in control. Whereas, those of p-tyrosine-insulin receptor substrate-1 and heat shock protein 70 did not show significant differences. This review suggested that Aβ42, t-tau, p-T181-tau, and p-S396-tau could be effective in diagnosing AD as blood biomarkers, despite the limitation in the meta-analysis based on the availability of data. Therefore, certain BDE proteins could be used as effective biomarkers for AD.
Collapse
Affiliation(s)
- Ka Young Kim
- Department of Nursing, College of Nursing, Gachon University, Incheon 21936, Korea;
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea
| | - Ki Young Shin
- Bio-MAX Institute, Seoul National University, Seoul 08826, Korea
- Correspondence: (K.Y.S.); (K.-AC.); Tel.: +82-2-880-1737 (K.Y.S.); +82-32-899-6411 (K.-AC.)
| | - Keun-A Chang
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea
- Department of Pharmacology, College of Medicine, Gachon University, Incheon 21936, Korea
- Neuroscience of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Incheon 21936, Korea
- Correspondence: (K.Y.S.); (K.-AC.); Tel.: +82-2-880-1737 (K.Y.S.); +82-32-899-6411 (K.-AC.)
| |
Collapse
|
73
|
Guix FX, Capitán AM, Casadomé-Perales Á, Palomares-Pérez I, López Del Castillo I, Miguel V, Goedeke L, Martín MG, Lamas S, Peinado H, Fernández-Hernando C, Dotti CG. Increased exosome secretion in neurons aging in vitro by NPC1-mediated endosomal cholesterol buildup. Life Sci Alliance 2021; 4:4/8/e202101055. [PMID: 34183444 PMCID: PMC8321659 DOI: 10.26508/lsa.202101055] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 11/24/2022] Open
Abstract
The present study unveils the molecular mechanism through which neurons in vitro compensate age-associated proteostasis defects by increasing exosome release. As neurons age, they show a decrease in their ability to degrade proteins and membranes. Because undegraded material is a source of toxic products, defects in degradation are associated with reduced cell function and survival. However, there are very few dead neurons in the aging brain, suggesting the action of compensatory mechanisms. We show in this work that ageing neurons in culture show large multivesicular bodies (MVBs) filled with intralumenal vesicles (ILVs) and secrete more small extracellular vesicles than younger neurons. We also show that the high number of ILVs is the consequence of the accumulation of cholesterol in MVBs, which in turn is due to decreased levels of the cholesterol extruding protein NPC1. NPC1 down-regulation is the consequence of a combination of upregulation of the NPC1 repressor microRNA 33, and increased degradation, due to Akt-mTOR targeting of NPC1 to the phagosome. Although releasing more exosomes can be beneficial to old neurons, other cells, neighbouring and distant, can be negatively affected by the waste material they contain.
Collapse
Affiliation(s)
- Francesc X Guix
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Ana Marrero Capitán
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Álvaro Casadomé-Perales
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Irene Palomares-Pérez
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Inés López Del Castillo
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Verónica Miguel
- Molecular Pathophysiology of Fibrosis, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Leigh Goedeke
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA.,Integrative Cell Signalling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Mauricio G Martín
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| | - Santiago Lamas
- Molecular Pathophysiology of Fibrosis, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Héctor Peinado
- Microenvironment and Metastasis Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA.,Integrative Cell Signalling and Neurobiology of Metabolism Program, Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Carlos G Dotti
- Molecular Neuropathology Unit, Physiological and Pathological Processes Program, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma de Madrid (UAM), Madrid, Spain
| |
Collapse
|
74
|
Shim KH, Go HG, Bae H, Jeong DE, Kim D, Youn YC, Kim S, An SSA, Kang MJ. Decreased Exosomal Acetylcholinesterase Activity in the Plasma of Patients With Parkinson's Disease. Front Aging Neurosci 2021; 13:665400. [PMID: 34122043 PMCID: PMC8193230 DOI: 10.3389/fnagi.2021.665400] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
Exosomes, which are small extracellular vesicles produced from various cell types, contain a variety of molecular constituents, such as proteins, lipids, and RNA. Recently, exosomal biomarkers have been investigated to probe the understanding and diagnosis of neurodegenerative disorders. Previous reports have demonstrated increased exosomal α-synuclein (α-syn) in patients with Parkinson’s disease (PD) in comparison to healthy controls (HC). Interestingly, the cholinergic loss was revealed in the central and peripheral nervous systems in histopathology and molecular neuroimaging. Thereby, we simultaneously examined acetylcholinesterase (AChE) with α-syn as exosomal markers. Exosomes were isolated from the plasma of 34 FP-CIT PET proven patients with PD and 29 HC. Exosomal α-syn and AChE activity were quantified andthe relationship with clinical parameters was analyzed. Remarkably, exosomal AChE activity was significantly decreased in PD compared to HC (P = 0.002). Moreover, exosomal AChE activity in PD revealed a strong negative correlation with disease severity, including H&Y (P = 0.007) and UPDRS part III (P = 0.047) scores. By contrast, no significant difference in exosomal α-syn concentration was observed between groups. These results support the occurrence of cholinergic dysfunction in PD, and they could be implicated with disease progression, especially motor deficits. Exosomal AChE activity with advanced exosome isolation techniques may be a reliable biomarker for the early diagnosis and prognosis of PD.
Collapse
Affiliation(s)
- Kyu Hwan Shim
- Department of Neurology, Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, South Korea
| | - Han Gyeol Go
- Department of Neurology, Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, South Korea.,Department of Bionano Technology, Gachon University, Seongnam-si, South Korea
| | - Heewon Bae
- Department of Neurology, Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, South Korea
| | - Da-Eun Jeong
- Department of Neurology, Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, South Korea
| | - Danyeong Kim
- Department of Bionano Technology, Gachon University, Seongnam-si, South Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University Hospital, Seoul, South Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University Bundang Hospital and Seoul National University College of Medicine, Seongnam-si, South Korea
| | - Seong Soo A An
- Department of Bionano Technology, Gachon University, Seongnam-si, South Korea
| | - Min Ju Kang
- Department of Neurology, Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, South Korea
| |
Collapse
|
75
|
Wysocka A, Palasz E, Steczkowska M, Niewiadomska G. Dangerous Liaisons: Tau Interaction with Muscarinic Receptors. Curr Alzheimer Res 2021; 17:224-237. [PMID: 32329686 PMCID: PMC7509759 DOI: 10.2174/1567205017666200424134311] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 02/05/2020] [Accepted: 03/25/2020] [Indexed: 11/22/2022]
Abstract
The molecular processes underlying neurodegenerative diseases (such as Alzheimer's Disease - AD) remain poorly understood. There is also an imperative need for disease-modifying therapies in AD since the present treatments, acetylcholinesterase inhibitors and NMDA antagonists, do not halt its progression. AD and other dementias present unique pathological features such as that of microtubule associated protein tau metabolic regulation. Tau has numerous binding partners, including signaling molecules, cytoskeletal elements and lipids, which suggests that it is a multifunctional protein. AD has also been associated with severe loss of cholinergic markers in the brain and such loss may be due to the toxic interaction of tau with cholinergic muscarinic receptors. By using specific antagonists of muscarinic receptors it was found in vitro that extracellular tau binds to M1 and M3 receptors and which the increase of intracellular calcium found in neuronal cells upon tau-binding. However, so far, the significance of tau signaling through muscarinic receptor in vivo in tauopathic models remains uncertain. The data reviewed in the present paper highlight the significant effect of M1 receptor/tau interaction in exacerbating tauopathy related pathological features and suggest that selective M1 agonists may serve as a prototype for future therapeutic development toward modification of currently intractable neurodegenerative diseases, such as tauopathies.
Collapse
Affiliation(s)
- Adrianna Wysocka
- Neurobiology Center, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Ewelina Palasz
- Department of Applied Physiology, Mossakowski Medical Research Center, 02-093 Warsaw, Poland
| | - Marta Steczkowska
- Department of Applied Physiology, Mossakowski Medical Research Center, 02-093 Warsaw, Poland
| | - Grazyna Niewiadomska
- Neurobiology Center, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| |
Collapse
|
76
|
Beatriz M, Vilaça R, Lopes C. Exosomes: Innocent Bystanders or Critical Culprits in Neurodegenerative Diseases. Front Cell Dev Biol 2021; 9:635104. [PMID: 34055771 PMCID: PMC8155522 DOI: 10.3389/fcell.2021.635104] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are nano-sized membrane-enclosed particles released by cells that participate in intercellular communication through the transfer of biologic material. EVs include exosomes that are small vesicles that were initially associated with the disposal of cellular garbage; however, recent findings point toward a function as natural carriers of a wide variety of genetic material and proteins. Indeed, exosomes are vesicle mediators of intercellular communication and maintenance of cellular homeostasis. The role of exosomes in health and age-associated diseases is far from being understood, but recent evidence implicates exosomes as causative players in the spread of neurodegenerative diseases. Cells from the central nervous system (CNS) use exosomes as a strategy not only to eliminate membranes, toxic proteins, and RNA species but also to mediate short and long cell-to-cell communication as carriers of important messengers and signals. The accumulation of protein aggregates is a common pathological hallmark in many neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and prion diseases. Protein aggregates can be removed and delivered to degradation by the endo-lysosomal pathway or can be incorporated in multivesicular bodies (MVBs) that are further released to the extracellular space as exosomes. Because exosome transport damaged cellular material, this eventually contributes to the spread of pathological misfolded proteins within the brain, thus promoting the neurodegeneration process. In this review, we focus on the role of exosomes in CNS homeostasis, their possible contribution to the development of neurodegenerative diseases, the usefulness of exosome cargo as biomarkers of disease, and the potential benefits of plasma circulating CNS-derived exosomes.
Collapse
Affiliation(s)
- Margarida Beatriz
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Rita Vilaça
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Carla Lopes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
77
|
Counil H, Krantic S. Synaptic Activity and (Neuro)Inflammation in Alzheimer's Disease: Could Exosomes be an Additional Link? J Alzheimers Dis 2021; 74:1029-1043. [PMID: 32176642 DOI: 10.3233/jad-191237] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Nanosized extracellular vesicles, known as exosomes, are produced by all cell types in mammalian organisms and have been recently involved in neurodegeneration. In the brain, both glia and neurons give rise to exosomes, which contribute to their intercellular communication. In addition, brain-derived exosomes have a remarkable property to cross the blood-brain-barrier bi-directionally. In this line, exosomes of central origin have been identified in peripheral circulation and already considered as putative blood biomarkers of neurodegenerative diseases, including Alzheimer's disease (AD). Moreover, tentative use of exosomes as vehicle for the clearance of brain-born toxic proteins or, conversely, neuroprotective drug delivery, was also envisaged. However, little is known about the precise role of exosomes in the control and regulation of neuronal functions. Based on the presence of subunits of glutamate receptors in neuron-derived exosomes on one hand, and complement proteins in astrocyte-derived exosomes on the other hand, we hypothesize that exosomes may participate in the control of neuronal excitability via inflammatory-like mechanisms both at the central level and from the periphery. In this review, we will focus on AD and discuss the mechanisms by which exosomes of neuronal, glial, and/or peripheral origin could impact on neuronal excitability either directly or indirectly.
Collapse
Affiliation(s)
- Hermine Counil
- Sorbonne Université, Inserm UMRS 938, St. Antoine Res. Ctr. (CRSA), F-75012, Paris, France
| | - Slavica Krantic
- Sorbonne Université, Inserm UMRS 938, St. Antoine Res. Ctr. (CRSA), F-75012, Paris, France
| |
Collapse
|
78
|
Liang Y, Lehrich BM, Zheng S, Lu M. Emerging methods in biomarker identification for extracellular vesicle-based liquid biopsy. J Extracell Vesicles 2021; 10:e12090. [PMID: 34012517 PMCID: PMC8114032 DOI: 10.1002/jev2.12090] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 03/17/2021] [Accepted: 04/13/2021] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) are released by many cell types and distributed within various biofluids. EVs have a lipid membrane-confined structure that allows for carrying unique molecular information originating from their parent cells. The species and quantity of EV cargo molecules, including nucleic acids, proteins, lipids, and metabolites, may vary largely owing to their parent cell types and the pathophysiologic status. Such heterogeneity in EV populations provides immense challenges to researchers, yet allows for the possibility to prognosticate the pathogenesis of a particular tissue from unique molecular signatures of dispersing EVs within biofluids. However, the inherent nature of EV's small size requires advanced methods for EV purification and evaluation from the complex biofluid. Recently, the interdisciplinary significance of EV research has attracted growing interests, and the EV analytical platforms for their diagnostic prospect have markedly progressed. This review summarizes the recent advances in these EV detection techniques and methods with the intention of translating an EV-based liquid biopsy into clinical practice. This article aims to present an overview of current EV assessment techniques, with a focus on their progress and limitations, as well as an outlook on the clinical translation of an EV-based liquid biopsy that may augment current paradigms for the diagnosis, prognosis, and monitoring the response to therapy in a variety of disease settings.
Collapse
Affiliation(s)
- Yaxuan Liang
- Center for Biological Science and Technology, Advanced Institute of Natural SciencesBeijing Normal University at ZhuhaiZhuhaiChina
| | - Brandon M. Lehrich
- Medical Scientist Training ProgramUniversity of Pittsburgh School of Medicine and Carnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Siyang Zheng
- Department Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
- Department of Electrical and Computer EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Mengrou Lu
- Department Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| |
Collapse
|
79
|
Bok E, Leem E, Lee BR, Lee JM, Yoo CJ, Lee EM, Kim J. Role of the Lipid Membrane and Membrane Proteins in Tau Pathology. Front Cell Dev Biol 2021; 9:653815. [PMID: 33996814 PMCID: PMC8119898 DOI: 10.3389/fcell.2021.653815] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Abnormal accumulation of misfolded tau aggregates is a pathological hallmark of various tauopathies including Alzheimer’s disease (AD). Although tau is a cytosolic microtubule-associated protein enriched in neurons, it is also found in extracellular milieu, such as interstitial fluid, cerebrospinal fluid, and blood. Accumulating evidence showed that pathological tau spreads along anatomically connected areas in the brain through intercellular transmission and templated misfolding, thereby inducing neurodegeneration and cognitive dysfunction. In line with this, the spatiotemporal spreading of tau pathology is closely correlated with cognitive decline in AD patients. Although the secretion and uptake of tau involve multiple different pathways depending on tau species and cell types, a growing body of evidence suggested that tau is largely secreted in a vesicle-free forms. In this regard, the interaction of vesicle-free tau with membrane is gaining growing attention due to its importance for both of tau secretion and uptake as well as aggregation. Here, we review the recent literature on the mechanisms of the tau-membrane interaction and highlights the roles of lipids and proteins at the membrane in the tau-membrane interaction as well as tau aggregation.
Collapse
Affiliation(s)
- Eugene Bok
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Eunju Leem
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Bo-Ram Lee
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Ji Min Lee
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea.,School of Life Sciences, Kyungpook National University, Daegu, South Korea
| | - Chang Jae Yoo
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea.,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Eun Mi Lee
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea.,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Jaekwang Kim
- Dementia Research Group, Korea Brain Research Institute, Daegu, South Korea
| |
Collapse
|
80
|
Xiao L, Hareendran S, Loh YP. Function of exosomes in neurological disorders and brain tumors. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2021; 2:55-79. [PMID: 34368812 PMCID: PMC8341051 DOI: 10.20517/evcna.2021.04] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/15/2021] [Accepted: 03/19/2021] [Indexed: 12/11/2022]
Abstract
Exosomes are a subtype of extracellular vesicles released from different cell types including those in the nervous system, and are enriched in a variety of bioactive molecules such as RNAs, proteins and lipids. Numerous studies have indicated that exosomes play a critical role in many physiological and pathological activities by facilitating intercellular communication and modulating cells' responses to external environments. Particularly in the central nervous system, exosomes have been implicated to play a role in many neurological disorders such as abnormal neuronal development, neurodegenerative diseases, epilepsy, mental disorders, stroke, brain injury and brain cancer. Since exosomes recapitulate the characteristics of the parental cells and have the capacity to cross the blood-brain barrier, their cargo can serve as potential biomarkers for early diagnosis and clinical assessment of disease treatment. In this review, we describe the latest findings and current knowledge of the roles exosomes play in various neurological disorders and brain cancer, as well as their application as promising biomarkers. The potential use of exosomes to deliver therapeutic molecules to treat diseases of the central nervous system is also discussed.
Collapse
Affiliation(s)
| | | | - Y. Peng Loh
- Section on Cellular Neurobiology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
81
|
Secreted Extracellular Vesicle Molecular Cargo as a Novel Liquid Biopsy Diagnostics of Central Nervous System Diseases. Int J Mol Sci 2021; 22:ijms22063267. [PMID: 33806874 PMCID: PMC8004928 DOI: 10.3390/ijms22063267] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/18/2021] [Accepted: 03/20/2021] [Indexed: 12/11/2022] Open
Abstract
Secreted extracellular vesicles (EVs) are heterogeneous cell-derived membranous granules which carry a large diversity of molecules and participate in intercellular communication by transferring these molecules to target cells by endocytosis. In the last decade, EVs’ role in several pathological conditions, from etiology to disease progression or therapy evasion, has been consolidated, including in central nervous system (CNS)-related disorders. For this review, we performed a systematic search of original works published, reporting the presence of molecular components expressed in the CNS via EVs, which have been purified from plasma, serum or cerebrospinal fluid. Our aim is to provide a list of molecular EV components that have been identified from both nonpathological conditions and the most common CNS-related disorders. We discuss the methods used to isolate and enrich EVs from specific CNS-cells and the relevance of its components in each disease context.
Collapse
|
82
|
Extracellular Vesicles: Novel Roles in Neurological Disorders. Stem Cells Int 2021; 2021:6640836. [PMID: 33679989 PMCID: PMC7904361 DOI: 10.1155/2021/6640836] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/13/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022] Open
Abstract
Exosomes are small extracellular vesicles (EVs) secreted by almost all cells, which have been recognized as a novel platform for intercellular communication in the central nervous system (CNS). Exosomes are capable of transferring proteins, nucleic acids, lipids, and metabolites between neurons and glial cells, contributing to CNS development and maintenance of homeostasis. Evidence shows that exosomes originating from CNS cells act as suppressors or promoters in the initiation and progression of neurological disorders. Moreover, these exosomes have been shown to transfer molecules associated with diseases through the blood-brain barrier (BBB) and thus can be detected in blood. This unique feature enables exosomes to act as potential diagnostic biomarkers for neurological disorders. In addition, a substantial number of researches have indicated that exosomes derived from mesenchymal stem cells (MSCs) have repair effects on neurological disorders. Herein, we briefly introduce the roles of exosomes under physiological and pathological conditions. In particular, novel roles of exosomes as potential diagnostic biomarkers and therapeutic tools for neurological disorders are highlighted.
Collapse
|
83
|
Pampuscenko K, Morkuniene R, Krasauskas L, Smirnovas V, Tomita T, Borutaite V. Distinct Neurotoxic Effects of Extracellular Tau Species in Primary Neuronal-Glial Cultures. Mol Neurobiol 2021; 58:658-667. [PMID: 33001416 DOI: 10.1007/s12035-020-02150-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/23/2020] [Indexed: 12/27/2022]
Abstract
Recent data from various experimental models support the link between extracellular tau and neurodegeneration; however, the exact mechanisms by which extracellular tau or its modified forms or aggregates cause neuronal death remain unclear. We have previously shown that exogenously applied monomers and oligomers of the longest tau isoform (2N4R) at micromolar concentrations induced microglial phagocytosis of stressed-but-viable neurons in vitro. In this study, we investigated whether extracellular phosphorylated tau2N4R (p-tau2N4R), isoform 1N4R (tau1N4R) and K18 peptide can induce neuronal death or loss in primary neuronal-glial cell cultures. We found that p-tau2N4R at 30 nM concentration induced loss of viable neurons; however, 700 nM p-tau2N4R caused necrosis of both neurons and microglia, and this neuronal death was partially glial cell-dependent. We also found that extracellular tau1N4R oligomers, but not monomers, at 3 μM concentration caused neuronal death in mixed cell cultures: self-assembly tau1N4R dimers-tetramers induced neuronal necrosis and apoptosis, whereas Aβ-promoted tau1N4R oligomers caused glial cell-dependent loss of neurons without signs of increased cell death. Monomeric and pre-aggregated tau peptide containing 4R repeats (K18) had no effect in mixed cultures, suggesting that tau neurotoxicity might be dependent on N-terminal part of the protein. Taken together, our results show that extracellular p-tau2N4R is the most toxic form among investigated tau species inducing loss of neurons at low nanomolar concentrations and that neurotoxicity of tau1N4R is dependent on its aggregation state.
Collapse
Affiliation(s)
- Katryna Pampuscenko
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania.
| | - Ramune Morkuniene
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Lukas Krasauskas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Vytautas Smirnovas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Vilmante Borutaite
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
84
|
Pinnell JR, Cui M, Tieu K. Exosomes in Parkinson disease. J Neurochem 2021; 157:413-428. [PMID: 33372290 DOI: 10.1111/jnc.15288] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/17/2020] [Accepted: 12/23/2020] [Indexed: 12/18/2022]
Abstract
Parkinson disease (PD) is a prevalent neurodegenerative disease, in which the formation of misfolded and aggregated α-synuclein is a key neuropathological hallmark. Recent studies reveal that extracellular vesicles such as exosomes present a potential mechanism for propagation of pathological α-synuclein throughout the brain. The ability of exosomes to transport proteins and genetic material between cells, including mRNA and microRNAs which have been implicated in PD pathology, provides critical insights as to how exosomes may contribute to pathological progression in PD. Advances have also been made in the investigation of exosomes as potential tools for the modulation of Parkinson's pathology; their detection extracellularly may facilitate their use as biomarkers, while their small size could be utilised as vectors for the delivery of therapeutics. The aim of this review was to highlight our current knowledge of the role of exosomes in PD and potential clinical application.
Collapse
Affiliation(s)
- Jennifer R Pinnell
- Department of Environmental Health Sciences, Florida International University, Miami, FL, USA.,Peninsula Schools of Medicine and Dentistry, Plymouth University, Plymouth, UK
| | - Mei Cui
- Department of Neurology, Huashan hospital, Fudan University, Shanghai, China
| | - Kim Tieu
- Department of Environmental Health Sciences, Florida International University, Miami, FL, USA.,Biomolecular Sciences Institute, Florida International University, Miami, FL, USA
| |
Collapse
|
85
|
Yu Z, Shi M, Stewart T, Fernagut PO, Huang Y, Tian C, Dehay B, Atik A, Yang D, De Giorgi F, Ichas F, Canron MH, Ceravolo R, Frosini D, Kim HJ, Feng T, Meissner WG, Zhang J. Reduced oligodendrocyte exosome secretion in multiple system atrophy involves SNARE dysfunction. Brain 2021; 143:1780-1797. [PMID: 32428221 DOI: 10.1093/brain/awaa110] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 01/24/2020] [Accepted: 02/23/2020] [Indexed: 12/13/2022] Open
Abstract
Transportation of key proteins via extracellular vesicles has been recently implicated in various neurodegenerative disorders, including Parkinson's disease, as a new mechanism of disease spreading and a new source of biomarkers. Extracellular vesicles likely to be derived from the brain can be isolated from peripheral blood and have been reported to contain higher levels of α-synuclein (α-syn) in Parkinson's disease patients. However, very little is known about extracellular vesicles in multiple system atrophy, a disease that, like Parkinson's disease, involves pathological α-syn aggregation, though the process is centred around oligodendrocytes in multiple system atrophy. In this study, a novel immunocapture technology was developed to isolate blood CNPase-positive, oligodendrocyte-derived enriched microvesicles (OEMVs), followed by fluorescent nanoparticle tracking analysis and assessment of α-syn levels contained within the OEMVs. The results demonstrated that the concentrations of OEMVs were significantly lower in multiple system atrophy patients, compared to Parkinson's disease patients and healthy control subjects. It is also noted that the population of OEMVs involved was mainly in the size range closer to that of exosomes, and that the average α-syn concentrations (per vesicle) contained in these OEMVs were not significantly different among the three groups. The phenomenon of reduced OEMVs was again observed in a transgenic mouse model of multiple system atrophy and in primary oligodendrocyte cultures, and the mechanism involved was likely related, at least in part, to an α-syn-mediated interference in the interaction between syntaxin 4 and VAMP2, leading to the dysfunction of the SNARE complex. These results suggest that reduced OEMVs could be an important mechanism related to pathological α-syn aggregation in oligodendrocytes, and the OEMVs found in peripheral blood could be further explored for their potential as multiple system atrophy biomarkers.
Collapse
Affiliation(s)
- Zhenwei Yu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100050, China
| | - Min Shi
- Department of Pathology, University of Washington School of Medicine, 325 9th Ave, HMC Box 359635, Seattle, WA 98104, USA
| | - Tessandra Stewart
- Department of Pathology, University of Washington School of Medicine, 325 9th Ave, HMC Box 359635, Seattle, WA 98104, USA
| | - Pierre-Olivier Fernagut
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France.,Université de Poitiers, Laboratoire de Neurosciences Expérimentales et Cliniques, UMR_S 1084, F-86000 Poitiers, France.,INSERM, Laboratoire de Neurosciences Expérimentales et Cliniques, UMR_S 1084, F-86000 Poitiers, France
| | - Yang Huang
- Department of Pathology, Peking University Health Science Centre and Third Hospital, Beijing, China
| | - Chen Tian
- Department of Pathology, University of Washington School of Medicine, 325 9th Ave, HMC Box 359635, Seattle, WA 98104, USA.,Department of Pathology, Peking University Health Science Centre and Third Hospital, Beijing, China
| | - Benjamin Dehay
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Anzari Atik
- Department of Pathology, University of Washington School of Medicine, 325 9th Ave, HMC Box 359635, Seattle, WA 98104, USA
| | - Dishun Yang
- Department of Pathology, University of Washington School of Medicine, 325 9th Ave, HMC Box 359635, Seattle, WA 98104, USA
| | - Francesca De Giorgi
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France.,Université de Poitiers, Laboratoire de Neurosciences Expérimentales et Cliniques, UMR_S 1084, F-86000 Poitiers, France.,INSERM, Laboratoire de Neurosciences Expérimentales et Cliniques, UMR_S 1084, F-86000 Poitiers, France
| | - François Ichas
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France.,Université de Poitiers, Laboratoire de Neurosciences Expérimentales et Cliniques, UMR_S 1084, F-86000 Poitiers, France.,INSERM, Laboratoire de Neurosciences Expérimentales et Cliniques, UMR_S 1084, F-86000 Poitiers, France
| | - Marie-Hélène Canron
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Daniela Frosini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Han-Joon Kim
- Department of Neurology and Movement Disorder Center, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Tao Feng
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Wassilios G Meissner
- Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France.,Service de Neurologie, CRMR Atrophie Multisystématisée, CHU Bordeaux, F-33000 Bordeaux, France.,Department of Medicine, University of Otago, Christchurch, New Zealand.,New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Jing Zhang
- Department of Pathology, University of Washington School of Medicine, 325 9th Ave, HMC Box 359635, Seattle, WA 98104, USA.,Department of Pathology, Peking University Health Science Centre and Third Hospital, Beijing, China.,Advanced Innovation Center for Human Brain Protection, TianTan Hospital, Capital Medical University, Beijing 100050, China.,Department of Pathology, the First Affiliated Hospital and School of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
86
|
Pedrioli G, Paganetti P. Hijacking Endocytosis and Autophagy in Extracellular Vesicle Communication: Where the Inside Meets the Outside. Front Cell Dev Biol 2021; 8:595515. [PMID: 33490063 PMCID: PMC7817780 DOI: 10.3389/fcell.2020.595515] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 11/18/2020] [Indexed: 12/25/2022] Open
Abstract
Extracellular vesicles, phospholipid bilayer-membrane vesicles of cellular origin, are emerging as nanocarriers of biological information between cells. Extracellular vesicles transport virtually all biologically active macromolecules (e.g., nucleotides, lipids, and proteins), thus eliciting phenotypic changes in recipient cells. However, we only partially understand the cellular mechanisms driving the encounter of a soluble ligand transported in the lumen of extracellular vesicles with its cytosolic receptor: a step required to evoke a biologically relevant response. In this context, we review herein current evidence supporting the role of two well-described cellular transport pathways: the endocytic pathway as the main entry route for extracellular vesicles and the autophagic pathway driving lysosomal degradation of cytosolic proteins. The interplay between these pathways may result in the target engagement between an extracellular vesicle cargo protein and its cytosolic target within the acidic compartments of the cell. This mechanism of cell-to-cell communication may well own possible implications in the pathogenesis of neurodegenerative disorders.
Collapse
Affiliation(s)
- Giona Pedrioli
- Neurodegeneration Research Group, Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Torricella-Taverne, Switzerland
- Member of the International Ph.D. Program of the Biozentrum, University of Basel, Basel, Switzerland
| | - Paolo Paganetti
- Neurodegeneration Research Group, Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Torricella-Taverne, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
87
|
Rastogi S, Sharma V, Bharti PS, Rani K, Modi GP, Nikolajeff F, Kumar S. The Evolving Landscape of Exosomes in Neurodegenerative Diseases: Exosomes Characteristics and a Promising Role in Early Diagnosis. Int J Mol Sci 2021; 22:E440. [PMID: 33406804 PMCID: PMC7795439 DOI: 10.3390/ijms22010440] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases (ND) remains to be one of the biggest burdens on healthcare systems and serves as a leading cause of disability and death. Alzheimer's disease (AD) is among the most common of such disorders, followed by Parkinson's disease (PD). The basic molecular details of disease initiation and pathology are still under research. Only recently, the role of exosomes has been linked to the initiation and progression of these neurodegenerative diseases. Exosomes are small bilipid layer enclosed extracellular vesicles, which were once considered as a cellular waste and functionless. These nano-vesicles of 30-150 nm in diameter carry specific proteins, lipids, functional mRNAs, and high amounts of non-coding RNAs (miRNAs, lncRNAs, and circRNAs). As the exosomes content is known to vary as per their originating and recipient cells, these vesicles can be utilized as a diagnostic biomarker for early disease detection. Here we review exosomes, their biogenesis, composition, and role in neurodegenerative diseases. We have also provided details for their characterization through an array of available techniques. Their updated role in neurodegenerative disease pathology is also discussed. Finally, we have shed light on a novel field of salivary exosomes as a potential candidate for early diagnosis in neurodegenerative diseases and compared the biomarkers of salivary exosomes with other blood/cerebrospinal fluid (CSF) based exosomes within these neurological ailments.
Collapse
Affiliation(s)
- Simran Rastogi
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India; (S.R.); (V.S.); (P.S.B.)
| | - Vaibhav Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India; (S.R.); (V.S.); (P.S.B.)
| | - Prahalad Singh Bharti
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India; (S.R.); (V.S.); (P.S.B.)
| | - Komal Rani
- Department of Biotechnology, Amity University, Mumbai 410206, India;
| | - Gyan P. Modi
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India;
| | - Fredrik Nikolajeff
- Department of Health Science, Lulea Technical University, 97187 Lulea, Sweden
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India; (S.R.); (V.S.); (P.S.B.)
| |
Collapse
|
88
|
Exosomal tau with seeding activity is released from Alzheimer's disease synapses, and seeding potential is associated with amyloid beta. J Transl Med 2021; 101:1605-1617. [PMID: 34462532 PMCID: PMC8590975 DOI: 10.1038/s41374-021-00644-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 01/23/2023] Open
Abstract
Synaptic transfer of tau has long been hypothesized from the human pathology pattern and has been demonstrated in vitro and in vivo, but the precise mechanisms remain unclear. Extracellular vesicles such as exosomes have been suggested as a mechanism, but not all tau is exosomal. The present experiments use a novel flow cytometry assay to quantify depolarization of synaptosomes by KCl after loading with FM2-10, which induces a fluorescence reduction associated with synaptic vesicle release; the degree of reduction in cryopreserved human samples equaled that seen in fresh mouse synaptosomes. Depolarization induced the release of vesicles in the size range of exosomes, along with tetraspanin markers of extracellular vesicles. A number of tau peptides were released, including tau oligomers; released tau was primarily unphosphorylated and C-terminal truncated, with Aβ release just above background. When exosomes were immunopurified from release supernatants, a prominent tau band showed a dark smeared appearance of SDS-stable oligomers along with the exosomal marker syntenin-1, and these exosomes induced aggregation in the HEK tau biosensor assay. However, the flow-through did not seed aggregation. Size exclusion chromatography of purified released exosomes shows faint signals from tau in the same fractions that show a CD63 band, an exosomal size signal, and seeding activity. Crude synaptosomes from control, tauopathy, and AD cases demonstrated lower seeding in tauopathy compared to AD that is correlated with the measured Aβ42 level. These results show that AD synapses release exosomal tau that is C-terminal-truncated, oligomeric, and with seeding activity that is enhanced by Aβ. Taken together with previous findings, these results are consistent with a direct prion-like heterotypic seeding of tau by Aβ within synaptic terminals, with subsequent loading of aggregated tau onto exosomes that are released and competent for tau seeding activity.
Collapse
|
89
|
Current and future applications of induced pluripotent stem cell-based models to study pathological proteins in neurodegenerative disorders. Mol Psychiatry 2021; 26:2685-2706. [PMID: 33495544 PMCID: PMC8505258 DOI: 10.1038/s41380-020-00999-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/02/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
Neurodegenerative disorders emerge from the failure of intricate cellular mechanisms, which ultimately lead to the loss of vulnerable neuronal populations. Research conducted across several laboratories has now provided compelling evidence that pathogenic proteins can also contribute to non-cell autonomous toxicity in several neurodegenerative contexts, including Alzheimer's, Parkinson's, and Huntington's diseases as well as Amyotrophic Lateral Sclerosis. Given the nearly ubiquitous nature of abnormal protein accumulation in such disorders, elucidating the mechanisms and routes underlying these processes is essential to the development of effective treatments. To this end, physiologically relevant human in vitro models are critical to understand the processes surrounding uptake, release and nucleation under physiological or pathological conditions. This review explores the use of human-induced pluripotent stem cells (iPSCs) to study prion-like protein propagation in neurodegenerative diseases, discusses advantages and limitations of this model, and presents emerging technologies that, combined with the use of iPSC-based models, will provide powerful model systems to propel fundamental research forward.
Collapse
|
90
|
Exosomes: Their Role in Pathogenesis, Diagnosis and Treatment of Diseases. Cancers (Basel) 2020; 13:cancers13010084. [PMID: 33396739 PMCID: PMC7795854 DOI: 10.3390/cancers13010084] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The aim of this review is to provide an overview of the current scientific evidence concerning the role played by exosomes in the pathogenesis, diagnosis and treatment of diseases. The potential use of exosomes as delivery vectors for small-molecule therapeutic agents will be discussed. In addition, a special emphasis will be placed on the involvement of exosomes in oncological diseases, as well as to their potential therapeutic application as liquid biopsy tools mainly in cancer diagnosis. A better understanding of exosome biology could improve the results of clinical interventions using exosomes as therapeutic agents. Abstract Exosomes are lipid bilayer particles released from cells into their surrounding environment. These vesicles are mediators of near and long-distance intercellular communication and affect various aspects of cell biology. In addition to their biological function, they play an increasingly important role both in diagnosis and as therapeutic agents. In this paper, we review recent literature related to the molecular composition of exosomes, paying special attention to their role in pathogenesis, along with their application as biomarkers and as therapeutic tools. In this context, we analyze the potential use of exosomes in biomedicine, as well as the limitations that preclude their wider application.
Collapse
|
91
|
Quiroz-Baez R, Hernández-Ortega K, Martínez-Martínez E. Insights Into the Proteomic Profiling of Extracellular Vesicles for the Identification of Early Biomarkers of Neurodegeneration. Front Neurol 2020; 11:580030. [PMID: 33362690 PMCID: PMC7759525 DOI: 10.3389/fneur.2020.580030] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are involved in the development and progression of neurodegenerative diseases, including Alzheimer's and Parkinson's disease. Moreover, EVs have the capacity to modify the physiology of neuronal circuits by transferring proteins, RNA, lipids, and metabolites. The proteomic characterization of EVs (exosomes and microvesicles) from preclinical models and patient samples has the potential to reveal new proteins and molecular networks that affect the normal physiology prior to the appearance of traditional biomarkers of neurodegeneration. Noteworthy, many of the genetic risks associated to the development of Alzheimer's and Parkinson's disease affect the crosstalk between mitochondria, endosomes, and lysosomes. Recent research has focused on determining the role of endolysosomal trafficking in the onset of neurodegenerative diseases. Proteomic studies indicate an alteration of biogenesis and molecular content of EVs as a result of endolysosomal and autophagic dysfunction. In this review, we discuss the status of EV proteomic characterization and their usefulness in discovering new biomarkers for the differential diagnosis of neurodegenerative diseases. Despite the challenges related to the failure to follow a standard isolation protocol and their implementation for a clinical setting, the analysis of EV proteomes has revealed the presence of key proteins with post-translational modifications that can be measured in peripheral fluids.
Collapse
Affiliation(s)
- Ricardo Quiroz-Baez
- Departamento de Investigación Básica, Dirección de Investigación, Instituto Nacional de Geriatría, Ciudad de México, Mexico
| | - Karina Hernández-Ortega
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Eduardo Martínez-Martínez
- Laboratory of Cell Communication & Extracellular Vesicles, Division of Basic Science, Instituto Nacional de Medicina Genómica, Ciudad de México, Mexico
| |
Collapse
|
92
|
Characterization of tau binding by gosuranemab. Neurobiol Dis 2020; 146:105120. [DOI: 10.1016/j.nbd.2020.105120] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/11/2020] [Accepted: 09/24/2020] [Indexed: 12/15/2022] Open
|
93
|
Aulston B, Liu Q, Mante M, Florio J, Rissman RA, Yuan SH. Extracellular Vesicles Isolated from Familial Alzheimer's Disease Neuronal Cultures Induce Aberrant Tau Phosphorylation in the Wild-Type Mouse Brain. J Alzheimers Dis 2020; 72:575-585. [PMID: 31594233 DOI: 10.3233/jad-190656] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of secreted particles consisting of microvesicles, which are released by budding of the cellular membrane, and exosomes, which are secreted through exocytosis from multivesicular bodies. EV cargo consists of a wide range of proteins and nucleic acids that can be transferred between cells. Importantly, EVs may be pathogenically involved in neurodegenerative diseases such as Alzheimer's disease (AD). While EVs derived from AD neurons have been found to be neurotoxic in vitro, little is known about the pathological consequences of AD EVs in vivo. Furthermore, although all known familial AD (fAD) mutations involve either amyloid-β protein precursor (AβPP) or the machinery that processes AβPP, hyperphosphorylation of the microtubule associated protein tau appears to play a critical role in fAD-associated neurodegeneration, and previous reports suggest EVs may propagate tau pathology in the AD brain. Therefore, we hypothesized that fAD EVs may have a mechanistic involvement in the development of fAD-associated tau pathology. To test this, we isolated EVs from iPSC-derived neuronal cultures generated from an fAD patient harboring a A246E mutation to presenilin-1 and stereotactically injected these EVs into the hippocampi of wild-type C57BL/6 mice. Five weeks after injection, mice were euthanized and pathology evaluated. Mice injected with fAD EVs displayed increased tau phosphorylation at multiple sites relative to PBS and non-disease control EV injected groups. Moreover, fAD EV injected hippocampi contained significantly more tau inclusions in the CA1 hippocampal neuronal field than controls. In total, these findings identify EVs as a potential mediator of fAD-associated tau dysregulation and warrant future studies to investigate the therapeutic potential of EV-targeted treatments for fAD.
Collapse
Affiliation(s)
- Brent Aulston
- University of California, San Diego, Department of Neurosciences, La Jolla, CA, USA
| | - Qing Liu
- University of California, San Diego, Department of Neurosciences, La Jolla, CA, USA
| | - Michael Mante
- University of California, San Diego, Department of Neurosciences, La Jolla, CA, USA
| | - Jazmin Florio
- University of California, San Diego, Department of Neurosciences, La Jolla, CA, USA
| | - Robert A Rissman
- University of California, San Diego, Department of Neurosciences, La Jolla, CA, USA.,Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA
| | - Shauna H Yuan
- University of California, San Diego, Department of Neurosciences, La Jolla, CA, USA
| |
Collapse
|
94
|
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: 6.2] [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
|
95
|
Bell BJ, Malvankar MM, Tallon C, Slusher BS. Sowing the Seeds of Discovery: Tau-Propagation Models of Alzheimer's Disease. ACS Chem Neurosci 2020; 11:3499-3509. [PMID: 33050700 DOI: 10.1021/acschemneuro.0c00531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The propagation of pathological proteins throughout the brain is the primary physiological hallmark of the progression of Alzheimer's Disease (AD). A growing body of evidence indicates that hyperphosphorylated Tau proteins are spread transcellularly between neurons in a prionlike fashion, inducing misfolding and aggregation into neurofibrillary tangles which accumulate along specific connectivity pathways. Earlier transgenic rodent AD models did not capture this disease-relevant spread, and therefore, seeded Tau-propagation models have been developed. Here, mutant human Tau (as isolated protein or packaged into an adeno-associated virus (AAV) viral vector) is stereotaxically injected into select brain regions and its histopathological propagation to downstream neurons quantified. These models offer a faster and more direct mechanism to evaluate genetic components and therapeutic approaches which attenuate Tau spreading in vivo. Recently, these Tau-seeding models have revealed several new targets for AD drug discovery, including nSMase2, SIRT1, p300/CBP, LRP1, and TYROBP, as well as the potential therapeutics based on melatonin and chondroitinase ABC. Importantly, these Tau-propagation rodent models more closely phenocopy the progression of AD in humans and are therefore likely to improve preclinical studies and derisk future moves into clinical trials.
Collapse
Affiliation(s)
- Benjamin J. Bell
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Medhinee M. Malvankar
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Carolyn Tallon
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
96
|
The Potential of Liquid Biopsy of the Brain Using Blood Extracellular Vesicles: The First Step Toward Effective Neuroprotection Against Neurodegenerative Diseases. Mol Diagn Ther 2020; 24:703-713. [PMID: 32975732 DOI: 10.1007/s40291-020-00493-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2020] [Indexed: 10/23/2022]
Abstract
Early diagnosis and biomarker-based ante-mortem tests are essential in efforts against the development of neurodegenerative diseases and can be considered primary neuroprotective measures. Blood is the ideal biofluid for a routine ante-mortem screening test. However, biomarker discovery in the blood is particularly difficult because of interference from factors both intrinsic and extrinsic to blood with the detection of hallmark neurodegenerative biomarkers, such as the pathological prion protein, amyloid-β, and others. Blood extracellular vesicles (EVs), such as exosomes, are cell-derived vesicles released into the blood from all parts of the body (including the brain and spinal cord). They are an enriched source of neural-derived EVs containing neurodegenerative biomarkers that mirror (in the blood) the condition present in the brain. The feasibility of using, and the reliability of, neural-derived blood EVs (NDBEVs) as a method of diagnosing Alzheimer disease and other neurodegenerative diseases has been assessed in strong proof-of-concept studies. Results from these studies strongly suggest that NDBEVs might represent the right strategy for specific, reliable, and early diagnosis of neurodegenerative diseases. Based on these results, NDBEVs might enable the creation of an ante-mortem blood test (liquid biopsy of the brain) for neurodegenerative diseases. This would enormously accelerate the therapy of neurodegenerative diseases. This review highlights the powerful potential of liquid biopsy of the brain using NDBEVs for early diagnosis and treatment of neurodegenerative diseases, and the challenges and limitations related to the identification of clinically applicable EV (exosomal) biomarkers using blood are discussed.
Collapse
|
97
|
Merezhko M, Uronen RL, Huttunen HJ. The Cell Biology of Tau Secretion. Front Mol Neurosci 2020; 13:569818. [PMID: 33071756 PMCID: PMC7539664 DOI: 10.3389/fnmol.2020.569818] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/26/2020] [Indexed: 12/27/2022] Open
Abstract
The progressive accumulation and spread of misfolded tau protein in the nervous system is the hallmark of tauopathies, progressive neurodegenerative diseases with only symptomatic treatments available. A growing body of evidence suggests that spreading of tau pathology can occur via cell-to-cell transfer involving secretion and internalization of pathological forms of tau protein followed by templated misfolding of normal tau in recipient cells. Several studies have addressed the cell biological mechanisms of tau secretion. It now appears that instead of a single mechanism, cells can secrete tau via three coexisting pathways: (1) translocation through the plasma membrane; (2) membranous organelles-based secretion; and (3) ectosomal shedding. The relative importance of these pathways in the secretion of normal and pathological tau is still elusive, though. Moreover, glial cells contribute to tau propagation, and the involvement of different cell types, as well as different secretion pathways, complicates the understanding of prion-like propagation of tauopathy. One of the important regulators of tau secretion in neuronal activity, but its mechanistic connection to tau secretion remains unclear and may involve all three secretion pathways of tau. This review article summarizes recent advancements in the field of tau secretion with an emphasis on cell biological aspects of the secretion process and discusses the role of neuronal activity and glial cells in the spread of pathological forms of tau.
Collapse
Affiliation(s)
- Maria Merezhko
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | - Henri J Huttunen
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
98
|
Walsh DM, Selkoe DJ. Amyloid β-protein and beyond: the path forward in Alzheimer's disease. Curr Opin Neurobiol 2020; 61:116-124. [PMID: 32197217 DOI: 10.1016/j.conb.2020.02.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 12/13/2022]
Abstract
Basic research on the biological mechanism of Alzheimer's disease has focused for decades on the age-related aggregation of the amyloid β-protein and its apparent downstream effects on microglia, astrocytes and neurons, including the posttranslational modification of the tau protein that seems necessary for symptom expression. Here, we discuss the highly challenging process of developing disease-modifying therapies and highlight several key areas of current research that are progressing in exciting directions. We conclude that further deep molecular analyses of the disease, including the mechanisms of β-amyloidosis, will enable more effective clinical trials and ultimately achieve the progress that our patients so deserve.
Collapse
Affiliation(s)
- Dominic M Walsh
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States; Alzheimer's Disease and Dementia Research Unit, Biogen Inc., 115 Broadway, Cambridge, MA 02142, United States.
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
99
|
Ausó E, Gómez-Vicente V, Esquiva G. Biomarkers for Alzheimer's Disease Early Diagnosis. J Pers Med 2020; 10:E114. [PMID: 32899797 PMCID: PMC7563965 DOI: 10.3390/jpm10030114] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, affecting the central nervous system (CNS) through the accumulation of intraneuronal neurofibrillary tau tangles (NFTs) and β-amyloid plaques. By the time AD is clinically diagnosed, neuronal loss has already occurred in many brain and retinal regions. Therefore, the availability of early and reliable diagnosis markers of the disease would allow its detection and taking preventive measures to avoid neuronal loss. Current diagnostic tools in the brain, such as magnetic resonance imaging (MRI), positron emission tomography (PET) imaging, and cerebrospinal fluid (CSF) biomarkers (Aβ and tau) detection are invasive and expensive. Brain-secreted extracellular vesicles (BEVs) isolated from peripheral blood have emerged as novel strategies in the study of AD, with enormous potential as a diagnostic evaluation of therapeutics and treatment tools. In addition; similar mechanisms of neurodegeneration have been demonstrated in the brain and the eyes of AD patients. Since the eyes are more accessible than the brain, several eye tests that detect cellular and vascular changes in the retina have also been proposed as potential screening biomarkers. The aim of this study is to summarize and discuss several potential markers in the brain, eye, blood, and other accessible biofluids like saliva and urine, and correlate them with earlier diagnosis and prognosis to identify individuals with mild symptoms prior to dementia.
Collapse
Affiliation(s)
| | | | - Gema Esquiva
- Department of Optics, Pharmacology and Anatomy, University of Alicante, 03690 Alicante, Spain; (E.A.); (V.G.-V.)
| |
Collapse
|
100
|
De La-Rocque S, Moretto E, Butnaru I, Schiavo G. Knockin' on heaven's door: Molecular mechanisms of neuronal tau uptake. J Neurochem 2020; 156:563-588. [PMID: 32770783 PMCID: PMC8432157 DOI: 10.1111/jnc.15144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/18/2022]
Abstract
Since aggregates of the microtubule‐binding protein tau were found to be the main component of neurofibrillary tangles more than 30 years ago, their contribution to neurodegeneration in Alzheimer's disease (AD) and tauopathies has become well established. Recent work shows that both tau load and its distribution in the brain of AD patients correlate with cognitive decline more closely compared to amyloid plaque deposition. In addition, the amyloid cascade hypothesis has been recently challenged because of disappointing results of clinical trials designed to treat AD by reducing beta‐amyloid levels, thus fuelling a renewed interest in tau. There is now robust evidence to indicate that tau pathology can spread within the central nervous system via a prion‐like mechanism following a stereotypical pattern, which can be explained by the trans‐synaptic inter‐neuronal transfer of pathological tau. In the receiving neuron, tau has been shown to take multiple routes of internalisation, which are partially dependent on its conformation and aggregation status. Here, we review the emerging mechanisms proposed for the uptake of extracellular tau in neurons and the requirements for the propagation of its pathological conformers, addressing how they gain access to physiological tau monomers in the cytosol. Furthermore, we highlight some of the key mechanistic gaps of the field, which urgently need to be addressed to expand our understanding of tau propagation and lead to the identification of new therapeutic strategies for tauopathies.
Collapse
Affiliation(s)
- Samantha De La-Rocque
- UK Dementia Research Institute, University College London, London, UK.,Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Edoardo Moretto
- UK Dementia Research Institute, University College London, London, UK
| | - Ioana Butnaru
- UK Dementia Research Institute, University College London, London, UK
| | - Giampietro Schiavo
- UK Dementia Research Institute, University College London, London, UK.,Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|