1
|
Xing X, Liu H, Zhang M, Li Y. Mapping the current trends and hotspots of extracellular vesicles in Alzheimer's disease: a bibliometric analysis. Front Aging Neurosci 2024; 16:1485750. [PMID: 39759397 PMCID: PMC11697149 DOI: 10.3389/fnagi.2024.1485750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 12/06/2024] [Indexed: 01/07/2025] Open
Abstract
Background Extracellular vesicles (EVs) have garnered significant attention in Alzheimer's disease (AD) research over the past decade, largely due to their potential in diagnostics and therapeutics. Although the investigation of EVs in AD is a relatively recent endeavor, a comprehensive bibliometric analysis of this rapidly growing field has yet to be conducted. Methods This study aims to elucidate and synthesize the relationship between EVs and AD, offering critical insights to guide future research and expand therapeutic possibilities. Over the past 10-15 years, substantial progress has been made in this domain. Through bibliometric techniques, this analysis assesses research performance by examining scientific publications and metrics, including productivity indicators, impact measurements, data mining, and visualization tools. Results A total of 602 publications were analyzed using various online platforms for bibliometric analysis. Notably, the number of publications began to increase rapidly in 2018, with China and the United States emerging as leaders in this research area. The National Institute on Aging produced the highest number of publications among institutions. The Journal of Molecular Sciences and the Journal of Biological Chemistry were the most prolific and most frequently cited journals, respectively. Among individual contributors, Dimitrios Kapogiannis was identified as the most productive author, while Edward J. Goetzl was the most co-cited. The most prevalent keywords included "neurodegenerative diseases," "exosomes," "blood biomarkers," "amyloid beta," "microglia," and "tau protein." Current research hotspots involve microRNA dysregulation, oxidative stress, carboxyl-terminal fragments, small EVs, and mesenchymal stem cell-derived EVs, indicating key areas for future research. Conclusion Research on microRNA dysregulation, oxidative stress, carboxyl-terminal fragments, small EVs, and mesenchymal stem cell-derived EVs represents a critical frontier in the study of Alzheimer's disease. The role of EV-mediated neuroinflammation in AD is a focal point of ongoing investigation and will likely shape future developments in the field.
Collapse
Affiliation(s)
- Xiaolian Xing
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Neurology, Taiyuan Central Hospital, Taiyuan, Shanxi, China
| | - Hongwei Liu
- Department of Neurology, Taiyuan Central Hospital, Taiyuan, Shanxi, China
| | - Minheng Zhang
- Department of Gerontology, The First People's Hospital of Jinzhong, Yuci, Shanxi, China
| | - Yang Li
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
2
|
Li J, Song J, Jia L, Wang M, Ji X, Meng R, Zhou D. Exosomes in Central Nervous System Diseases: A Comprehensive Review of Emerging Research and Clinical Frontiers. Biomolecules 2024; 14:1519. [PMID: 39766226 PMCID: PMC11673277 DOI: 10.3390/biom14121519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
Exosomes, nano-sized lipid bilayer vesicles, have garnered significant attention as mediators of cell communication, particularly within the central nervous system (CNS). Their unique properties, including high stability, low immunogenicity, and the ability to traverse the blood-brain barrier (BBB), position them as promising tools for understanding and addressing CNS diseases. This comprehensive review delves into the biogenesis, properties, composition, functions, and isolation of exosomes, with a particular focus on their roles in cerebrovascular diseases, neurodegenerative disorders, and CNS tumors. Exosomes are involved in key pathophysiological processes in the CNS, including angiogenesis, inflammation, apoptosis, and cellular microenvironment modification. They demonstrate promise in mitigating ischemic injury, regulating inflammatory responses, and providing neuroprotection across various CNS conditions. Furthermore, exosomes carry distinct biomolecules, offering a novel method for the early diagnosis and monitoring of CNS diseases. Despite their potential, challenges such as complex extraction processes, the heterogeneity of exosomal contents, and targeted delivery limitations hinder their clinical application. Nevertheless, exosomes hold significant promise for advancing our understanding of CNS diseases and developing novel therapeutic strategies. This manuscript significantly contributes to the field by highlighting exosomes' potential in advancing our understanding of CNS diseases, underscoring their unique value in developing novel therapeutic strategies and mediating cellular communication.
Collapse
Affiliation(s)
- Jingrun Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jiahao Song
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Lina Jia
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Mengqi Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xunming Ji
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Ran Meng
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Da Zhou
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Advanced Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China
- National Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
3
|
Myers C, Cornwall GA. Host defense amyloids: Biosensors of the immune system? Andrology 2024; 12:973-980. [PMID: 37963844 DOI: 10.1111/andr.13555] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 11/16/2023]
Abstract
There is considerable evidence showing that highly ordered aggregate structures known as amyloids carry out essential biological roles in species ranging from bacteria to humans. Indeed, many antimicrobial peptides/proteins form amyloids to carry out their host defense functions and many amyloids are antimicrobial. The similarity of host defense amyloids from bacterial biofilms to the mammalian epididymal amyloid matrix implies highly conserved host defense structures/functions. With an emphasis on the epididymal amyloid matrix, here we review the common properties of host defense amyloids including unique traits that would allow them to function as powerful biosensors of the immune system.
Collapse
Affiliation(s)
- Caitlyn Myers
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Gail A Cornwall
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| |
Collapse
|
4
|
Peng KY, Liemisa B, Pasato J, D'Acunzo P, Pawlik M, Heguy A, Penikalapati SC, Labuza A, Pidikiti H, Alldred MJ, Ginsberg SD, Levy E, Mathews PM. Apolipoprotein E2 Expression Alters Endosomal Pathways in a Mouse Model With Increased Brain Exosome Levels During Aging. Traffic 2024; 25:e12937. [PMID: 38777335 PMCID: PMC11141728 DOI: 10.1111/tra.12937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024]
Abstract
The polymorphic APOE gene is the greatest genetic determinant of sporadic Alzheimer's disease risk: the APOE4 allele increases risk, while the APOE2 allele is neuroprotective compared with the risk-neutral APOE3 allele. The neuronal endosomal system is inherently vulnerable during aging, and APOE4 exacerbates this vulnerability by driving an enlargement of early endosomes and reducing exosome release in the brain of humans and mice. We hypothesized that the protective effects of APOE2 are, in part, mediated through the endosomal pathway. Messenger RNA analyses showed that APOE2 leads to an enrichment of endosomal pathways in the brain when compared with both APOE3 and APOE4. Moreover, we show age-dependent alterations in the recruitment of key endosomal regulatory proteins to vesicle compartments when comparing APOE2 to APOE3. In contrast to the early endosome enlargement previously shown in Alzheimer's disease and APOE4 models, we detected similar morphology and abundance of early endosomes and retromer-associated vesicles within cortical neurons of aged APOE2 targeted-replacement mice compared with APOE3. Additionally, we observed increased brain extracellular levels of endosome-derived exosomes in APOE2 compared with APOE3 mice during aging, consistent with enhanced endosomal cargo clearance by exosomes to the extracellular space. Our findings thus demonstrate that APOE2 enhances an endosomal clearance pathway, which has been shown to be impaired by APOE4 and which may be protective due to APOE2 expression during brain aging.
Collapse
Affiliation(s)
- Katherine Y Peng
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Braison Liemisa
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Jonathan Pasato
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Pasquale D'Acunzo
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Monika Pawlik
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Adriana Heguy
- Genome Technology Center, New York University Grossman School of Medicine, New York, New York, USA
- Department of Pathology, New York University Grossman School of Medicine, New York, New York, USA
| | - Sai C Penikalapati
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Amanda Labuza
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Harshitha Pidikiti
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Melissa J Alldred
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Stephen D Ginsberg
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York, USA
- Department of Neuroscience & Physiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Efrat Levy
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York, USA
- Department of Biochemistry & Molecular Pharmacology, New York University Grossman School of Medicine, New York, New York, USA
| | - Paul M Mathews
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
5
|
D'Acunzo P, Argyrousi EK, Ungania JM, Kim Y, DeRosa S, Pawlik M, Goulbourne CN, Arancio O, Levy E. Mitovesicles secreted into the extracellular space of brains with mitochondrial dysfunction impair synaptic plasticity. Mol Neurodegener 2024; 19:34. [PMID: 38616258 PMCID: PMC11017499 DOI: 10.1186/s13024-024-00721-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 03/18/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND Hypometabolism tied to mitochondrial dysfunction occurs in the aging brain and in neurodegenerative disorders, including in Alzheimer's disease, in Down syndrome, and in mouse models of these conditions. We have previously shown that mitovesicles, small extracellular vesicles (EVs) of mitochondrial origin, are altered in content and abundance in multiple brain conditions characterized by mitochondrial dysfunction. However, given their recent discovery, it is yet to be explored what mitovesicles regulate and modify, both under physiological conditions and in the diseased brain. In this study, we investigated the effects of mitovesicles on synaptic function, and the molecular players involved. METHODS Hippocampal slices from wild-type mice were perfused with the three known types of EVs, mitovesicles, microvesicles, or exosomes, isolated from the brain of a mouse model of Down syndrome or of a diploid control and long-term potentiation (LTP) recorded. The role of the monoamine oxidases type B (MAO-B) and type A (MAO-A) in mitovesicle-driven LTP impairments was addressed by treatment of mitovesicles with the irreversible MAO inhibitors pargyline and clorgiline prior to perfusion of the hippocampal slices. RESULTS Mitovesicles from the brain of the Down syndrome model reduced LTP within minutes of mitovesicle addition. Mitovesicles isolated from control brains did not trigger electrophysiological effects, nor did other types of brain EVs (microvesicles and exosomes) from any genotype tested. Depleting mitovesicles of their MAO-B, but not MAO-A, activity eliminated their ability to alter LTP. CONCLUSIONS Mitovesicle impairment of LTP is a previously undescribed paracrine-like mechanism by which EVs modulate synaptic activity, demonstrating that mitovesicles are active participants in the propagation of cellular and functional homeostatic changes in the context of neurodegenerative disorders.
Collapse
Affiliation(s)
- Pasquale D'Acunzo
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 10962, Orangeburg, NY, USA
- Department of Psychiatry, New York University Grossman School of Medicine, 10016, New York, NY, USA
| | - Elentina K Argyrousi
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, 10027, New York, NY, USA
- Department of Medicine, Columbia University, 10027, New York, NY, USA
| | - Jonathan M Ungania
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 10962, Orangeburg, NY, USA
| | - Yohan Kim
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 10962, Orangeburg, NY, USA
- Department of Psychiatry, New York University Grossman School of Medicine, 10016, New York, NY, USA
| | - Steven DeRosa
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 10962, Orangeburg, NY, USA
| | - Monika Pawlik
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 10962, Orangeburg, NY, USA
| | - Chris N Goulbourne
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 10962, Orangeburg, NY, USA
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, 10027, New York, NY, USA
- Department of Medicine, Columbia University, 10027, New York, NY, USA
| | - Efrat Levy
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 10962, Orangeburg, NY, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, 10016, New York, NY, USA.
- Department of Biochemistry & Molecular Pharmacology, New York University Grossman School of Medicine, 10027, New York, NY, USA.
- NYU Neuroscience Institute, New York University Grossman School of Medicine, 10016, New York, NY, USA.
| |
Collapse
|
6
|
Chemparathy DT, Ray S, Ochs C, Ferguson N, Gawande DY, Dravid SM, Callen S, Sil S, Buch S. Neuropathogenic role of astrocyte-derived extracellular vesicles in HIV-associated neurocognitive disorders. J Extracell Vesicles 2024; 13:e12439. [PMID: 38647111 PMCID: PMC11034007 DOI: 10.1002/jev2.12439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/22/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
Our previous findings demonstrated that astrocytic HIF-1α plays a major role in HIV-1 Tat-mediated amyloidosis which can lead to Alzheimer's-like pathology-a comorbidity of HIV-Associated Neurocognitive Disorders (HAND). These amyloids can be shuttled in extracellular vesicles, and we sought to assess whether HIV-1 Tat stimulated astrocyte-derived EVs (ADEVs) containing the toxic amyloids could result in neuronal injury in vitro and in vivo. We thus hypothesized that blocking HIF-1α could likely mitigate HIV-1 Tat-ADEV-mediated neuronal injury. Rat hippocampal neurons when exposed to HIV-1 Tat-ADEVs carrying the toxic amyloids exhibited amyloid accumulation and synaptodendritic injury, leading to functional loss as evidenced by alterations in miniature excitatory post synaptic currents. The silencing of astrocytic HIF-1α not only reduced the biogenesis of ADEVs, as well as amyloid cargos, but also ameliorated neuronal synaptodegeneration. Next, we determined the effect of HIV-1 Tat-ADEVs carrying amyloids in the hippocampus of naive mice brains. Naive mice receiving the HIV-1 Tat-ADEVs, exhibited behavioural changes, and Alzheimer's 's-like pathology accompanied by synaptodegeneration. This impairment(s) was not observed in mice injected with HIF-1α silenced ADEVs. This is the first report demonstrating the role of amyloid-carrying ADEVs in mediating synaptodegeneration leading to behavioural changes associated with HAND and highlights the protective role of HIF-1α.
Collapse
Affiliation(s)
- Divya T. Chemparathy
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Sudipta Ray
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Chase Ochs
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Natasha Ferguson
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Dinesh Y. Gawande
- Department of Pharmacology and NeuroscienceCreighton UniversityOmahaNebraskaUSA
| | - Shashank M. Dravid
- Department of Pharmacology and NeuroscienceCreighton UniversityOmahaNebraskaUSA
| | - Shannon Callen
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Susmita Sil
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Shilpa Buch
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| |
Collapse
|
7
|
Melachroinou K, Divolis G, Tsafaras G, Karampetsou M, Fortis S, Stratoulias Y, Papadopoulou G, Kriebardis AG, Samiotaki M, Vekrellis K. Endogenous Alpha-Synuclein is Essential for the Transfer of Pathology by Exosome-Enriched Extracellular Vesicles, Following Inoculation with Preformed Fibrils in vivo. Aging Dis 2024; 15:869-892. [PMID: 37548944 PMCID: PMC10917543 DOI: 10.14336/ad.2023.0614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/14/2023] [Indexed: 08/08/2023] Open
Abstract
The main pathological hallmark of Parkinson's disease (PD) and related synucleinopathies is the presence of intracellular proteinaceous aggregates, enriched in the presynaptic protein alpha-Synuclein (α-Syn). α-Syn association with exosomes has been previously documented both as a physiological process of secretion and as a pathological process of disease transmission, however, critical information about the mechanisms governing this interplay is still lacking. To address this, we utilized the α-Syn preformed fibril (PFF) mouse model of PD, as a source of brain-derived exosome-enriched extracellular vesicles (ExE-EVs) and assessed their pathogenic capacity following intrastriatal injections in host wild type (WT) mouse brain. We further investigated the impact of the fibrillar α-Syn on the exosomal cargo independent of the endogenous α-Syn, by isolating ExE-EVs from PFF-injected α-Syn knockout mice. Although PFF inoculation does not alter the morphology, size distribution, and quantity of brain-derived ExE-EVs, it triggers changes in the exosomal proteome related to synaptic and mitochondrial function, as well as metabolic processes. Importantly, we showed that the presence of the endogenous α-Syn is essential for the ExE-EVs to acquire a pathogenic capacity, allowing them to mediate disease transmission by inducing phosphorylated-α-Syn pathology. Notably, misfolded α-Syn containing ExE-EVs when injected in WT mice were able to induce astrogliosis and synaptic alterations in the host brain, at very early stages of α-Syn pathology, preceding the formation of the insoluble α-Syn accumulations. Collectively, our data suggest that exosomal cargo defines their ability to spread α-Syn pathology.
Collapse
Affiliation(s)
- Katerina Melachroinou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Georgios Divolis
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - George Tsafaras
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Mantia Karampetsou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Sotirios Fortis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica (UniWA), Egaleo, Greece.
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece.
| | - Yannis Stratoulias
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Gina Papadopoulou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| | - Anastasios G Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica (UniWA), Egaleo, Greece.
| | - Martina Samiotaki
- Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.
| | - Kostas Vekrellis
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
8
|
Bodart-Santos V, Pinheiro LS, da Silva-Junior AJ, Froza RL, Ahrens R, Gonçalves RA, Andrade MM, Chen Y, Alcantara CDL, Grinberg LT, Leite REP, Ferreira ST, Fraser PE, De Felice FG. Alzheimer's disease brain-derived extracellular vesicles reveal altered synapse-related proteome and induce cognitive impairment in mice. Alzheimers Dement 2023; 19:5418-5436. [PMID: 37204850 DOI: 10.1002/alz.13134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/15/2023] [Accepted: 04/17/2023] [Indexed: 05/20/2023]
Abstract
INTRODUCTION Extracellular vesicles (EVs) have been implicated in the spread of neuropathology in Alzheimer's disease (AD), but their involvement in behavioral outcomes linked to AD remains to be determined. METHODS EVs isolated from post mortem brain tissue from control, AD, or frontotemporal dementia (FTD) donors, as well as from APP/PS1 mice, were injected into the hippocampi of wild-type (WT) or a humanized Tau mouse model (hTau/mTauKO). Memory tests were carried out. Differentially expressed proteins in EVs were assessed by proteomics. RESULTS Both AD-EVs and APP/PS1-EVs trigger memory impairment in WT mice. We further demonstrate that AD-EVs and FTD-EVs carry Tau protein, present altered protein composition associated with synapse regulation and transmission, and trigger memory impairment in hTau/mTauKO mice. DISCUSSION Results demonstrate that AD-EVs and FTD-EVs have negative impacts on memory in mice and suggest that, in addition to spreading pathology, EVs may contribute to memory impairment in AD and FTD. HIGHLIGHTS Aβ was detected in EVs from post mortem AD brain tissue and APP/PS1 mice. Tau was enriched in EVs from post mortem AD, PSP and FTD brain tissue. AD-derived EVs and APP/PS1-EVs induce cognitive impairment in wild-type (WT) mice. AD- and FTD-derived EVs induce cognitive impairment in humanized Tau mice. Proteomics findings associate EVs with synapse dysregulation in tauopathies.
Collapse
Affiliation(s)
- Victor Bodart-Santos
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Lisandra S Pinheiro
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Almir J da Silva-Junior
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rudimar L Froza
- Oswaldo Cruz Institute, Oswaldo Cruz Foundation, FIOCRUZ, Rio de Janeiro, Brazil
| | - Rosemary Ahrens
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Rafaella A Gonçalves
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences and Department of Psychiatry, Queen's University, Kingston, Canada
| | - Mayara M Andrade
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Yan Chen
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Carolina de Lima Alcantara
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lea T Grinberg
- Department of Pathology, University of São Paulo Medical School, Sao Paulo, Brazil
- Memory and Aging Center, Department of Neurology and Pathology, University of California San Francisco, San Francisco, California, USA
| | - Renata E P Leite
- Department of Pathology, University of São Paulo Medical School, Sao Paulo, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences and Department of Psychiatry, Queen's University, Kingston, Canada
- D'OR Institute for Research and Education, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Kim B, Kang Y, Mendelson FE, Hayes JM, Savelieff MG, Nagrath S, Feldman EL. Palmitate and glucose increase amyloid precursor protein in extracellular vesicles: Missing link between metabolic syndrome and Alzheimer's disease. J Extracell Vesicles 2023; 12:e12340. [PMID: 37898562 PMCID: PMC10613125 DOI: 10.1002/jev2.12340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 06/05/2023] [Accepted: 06/11/2023] [Indexed: 10/30/2023] Open
Abstract
The metabolic syndrome (MetS) and Alzheimer's disease share several pathological features, including insulin resistance, abnormal protein processing, mitochondrial dysfunction and elevated inflammation and oxidative stress. The MetS constitutes elevated fasting glucose, obesity, dyslipidaemia and hypertension and increases the risk of developing Alzheimer's disease, but the precise mechanism remains elusive. Insulin resistance, which develops from a diet rich in sugars and saturated fatty acids, such as palmitate, is shared by the MetS and Alzheimer's disease. Extracellular vesicles (EVs) are also a point of convergence, with altered dynamics in both the MetS and Alzheimer's disease. However, the role of palmitate- and glucose-induced insulin resistance in the brain and its potential link through EVs to Alzheimer's disease is unknown. We demonstrate that palmitate and high glucose induce insulin resistance and amyloid precursor protein phosphorylation in primary rat embryonic cortical neurons and human cortical stem cells. Palmitate also triggers insulin resistance in oligodendrocytes, the supportive glia of the brain. Palmitate and glucose enhance amyloid precursor protein secretion from cortical neurons via EVs, which induce tau phosphorylation when added to naïve neurons. Additionally, EVs from palmitate-treated oligodendrocytes enhance insulin resistance in recipient neurons. Overall, our findings suggest a novel theory underlying the increased risk of Alzheimer's disease in MetS mediated by EVs, which spread Alzheimer's pathology and insulin resistance.
Collapse
Affiliation(s)
- Bhumsoo Kim
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichiganUSA
| | - Yoon‐Tae Kang
- Department of Chemical Engineering and Biointerfaces InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - Faye E. Mendelson
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichiganUSA
| | - John M. Hayes
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichiganUSA
| | - Masha G. Savelieff
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichiganUSA
| | - Sunitha Nagrath
- Department of Chemical Engineering and Biointerfaces InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - Eva L. Feldman
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- NeuroNetwork for Emerging TherapiesUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
10
|
Ran Q, Tian H, Lin J, Wang H, Wang B, Chen Z, Song D, Gong C. Mesenchymal Stem Cell-Derived Exosomes: A Novel Approach to Diabetes-Associated Cognitive Impairment. J Inflamm Res 2023; 16:4213-4228. [PMID: 37753267 PMCID: PMC10519429 DOI: 10.2147/jir.s429532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/12/2023] [Indexed: 09/28/2023] Open
Abstract
The progression of diabetes frequently results in a myriad of neurological disorders, including ischemic stroke, depression, blood-brain barrier impairment, and cognitive dysfunction. Notably, diabetes-associated cognitive impairment, a prevalent comorbidity during the course of diabetes, progressively affects patients' cognitive abilities and may reciprocally influence diabetes management, thereby severely impacting patients' quality of life. Extracellular vesicles, particularly nanoscale exosomes, have garnered considerable attention in recent years. These exosomes carry and transfer various functional molecules, such as proteins, lipids, and diverse non-coding RNAs, serving as novel regulators and communicators in intercellular interactions. Of particular interest, mesenchymal stem cell-derived exosomes (MSC-Exos) have been reported to traverse the blood-brain barrier and ameliorate intracerebral pathologies. This review elucidates the role of MSC-Exos in diabetes-related cognitive impairment, with a focus on their applications as biomarkers, modulation of neuronal regeneration and synaptic plasticity, anti-inflammatory properties, antioxidative effects, and their involvement in regulating the functionality of β-amyloid proteins during the course of cognitive impairment. The immense therapeutic potential of MSC-Exos in the treatment of diabetes-induced cognitive dysfunction is emphasized.
Collapse
Affiliation(s)
- Qingsen Ran
- Department of Science and Education, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangzhou Province, 518118, People’s Republic of China
| | - He Tian
- Department of Science and Education, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangzhou Province, 518118, People’s Republic of China
| | - Jian Lin
- Department of Science and Education, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangzhou Province, 518118, People’s Republic of China
| | - Han Wang
- Department of Gastroenterology, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, 130021, People’s Republic of China
| | - Bo Wang
- Department of Science and Education, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangzhou Province, 518118, People’s Republic of China
| | - Zhixin Chen
- Department of Science and Education, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangzhou Province, 518118, People’s Republic of China
| | - Da Song
- Department of Science and Education, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangzhou Province, 518118, People’s Republic of China
| | - Chunzhu Gong
- Department of Science and Education, Shenzhen Pingle Orthopedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, Guangzhou Province, 518118, People’s Republic of China
| |
Collapse
|
11
|
Tohumeken S, Deme P, Yoo SW, Gupta S, Rais R, Slusher BS, Haughey NJ. Neuronal deletion of nSMase2 reduces the production of Aβ and directly protects neurons. Neurobiol Dis 2023; 177:105987. [PMID: 36603748 DOI: 10.1016/j.nbd.2023.105987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/28/2022] [Accepted: 01/01/2023] [Indexed: 01/03/2023] Open
Abstract
Extracellular vesicles (EVs) have been proposed to regulate the deposition of Aβ. Multiple publications have shown that APP, amyloid processing enzymes and Aβ peptides are associated with EVs. However, very little Aβ is associated with EVs compared with the total amount Aβ present in human plasma, CSF, or supernatants from cultured neurons. The involvement of EVs has largely been inferred by pharmacological inhibition or whole body deletion of the sphingomyelin hydrolase neutral sphingomyelinase-2 (nSMase2) that is a key regulator for the biogenesis of at-least one population of EVs. Here we used a Cre-Lox system to selectively delete nSMase2 from pyramidal neurons in APP/PS1 mice (APP/PS1-SMPD3-Nex1) and found a ∼ 70% reduction in Aβ deposition at 6 months of age and ∼ 35% reduction at 12 months of age in both cortex and hippocampus. Brain ceramides were increased in APP/PS1 compared with Wt mice, but were similar to Wt in APP/PS1-SMPD3-Nex1 mice suggesting that elevated brain ceramides in this model involves neuronally expressed nSMase2. Reduced levels of PSD95 and deficits of long-term potentiation in APP/PS1 mice were normalized in APP/PS1-SMPD3-Nex1 mice. In contrast, elevated levels of IL-1β, IL-8 and TNFα in APP/PS1 mice were not normalized in APP/PS1-SMPD3-Nex1 mice compared with APP/PS1 mice. Mechanistic studies showed that the size of liquid ordered membrane microdomains was increased in APP/PS1 mice, as were the amounts of APP and BACE1 localized to these microdomains. Pharmacological inhibition of nSMase2 activity with PDDC reduced the size of the liquid ordered membrane microdomains, reduced the localization of APP with BACE1 and reduced the production of Aβ1-40 and Aβ1-42. Although inhibition of nSMase2 reduced the release and increased the size of EVs, very little Aβ was associated with EVs in all conditions tested. We also found that nSMase2 directly protected neurons from the toxic effects of oligomerized Aβ and preserved neural network connectivity despite considerable Aβ deposition. These data demonstrate that nSMase2 plays a role in the production of Aβ by stabilizing the interaction of APP with BACE1 in liquid ordered membrane microdomains, and directly protects neurons from the toxic effects of Aβ. The effects of inhibiting nSMase2 on EV biogenesis may be independent from effects on Aβ production and neuronal protection.
Collapse
Affiliation(s)
- Sehmus Tohumeken
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Pragney Deme
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Seung Wan Yoo
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Sujasha Gupta
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America
| | - Rana Rais
- The Johns Hopkins University School of Medicine, Departments of Psychiatry, United States of America
| | - Barbara S Slusher
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America; The Johns Hopkins University School of Medicine, Departments of Johns Hopkins Drug Discovery, United States of America; The Johns Hopkins University School of Medicine, Departments of Psychiatry, United States of America; The Johns Hopkins University School of Medicine, Departments of Pharmacology and Molecular Sciences, United States of America; The Johns Hopkins University School of Medicine, Departments of Department of Oncology, United States of America; The Johns Hopkins University School of Medicine, Departments of Department of Neuroscience, United States of America; The Johns Hopkins University School of Medicine, Departments of Department of Medicine, Baltimore, MD, United States of America
| | - Norman J Haughey
- The Johns Hopkins University School of Medicine, Departments of Neurology, United States of America; The Johns Hopkins University School of Medicine, Departments of Johns Hopkins Drug Discovery, United States of America.
| |
Collapse
|
12
|
Emerging Roles of Extracellular Vesicles in Alzheimer's Disease: Focus on Synaptic Dysfunction and Vesicle-Neuron Interaction. Cells 2022; 12:cells12010063. [PMID: 36611856 PMCID: PMC9818402 DOI: 10.3390/cells12010063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Alzheimer's disease (AD) is considered by many to be a synaptic failure. Synaptic function is in fact deeply affected in the very early disease phases and recognized as the main cause of AD-related cognitive impairment. While the reciprocal involvement of amyloid beta (Aβ) and tau peptides in these processes is under intense investigation, the crucial role of extracellular vesicles (EVs) released by different brain cells as vehicles for these molecules and as mediators of early synaptic alterations is gaining more and more ground in the field. In this review, we will summarize the current literature on the contribution of EVs derived from distinct brain cells to neuronal alterations and build a working model for EV-mediated propagation of synaptic dysfunction in early AD. A deeper understanding of EV-neuron interaction will provide useful targets for the development of novel therapeutic approaches aimed at hampering AD progression.
Collapse
|
13
|
Vaz M, Soares Martins T, Henriques AG. Extracellular vesicles in the study of Alzheimer's and Parkinson's diseases: Methodologies applied from cells to biofluids. J Neurochem 2022; 163:266-309. [PMID: 36156258 PMCID: PMC9828694 DOI: 10.1111/jnc.15697] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 01/12/2023]
Abstract
Extracellular vesicles (EVs) are gaining increased importance in fundamental research as key players in disease pathogenic mechanisms, but also in translational and clinical research due to their value in biomarker discovery, either for diagnostics and/or therapeutics. In the first research scenario, the study of EVs isolated from neuronal models mimicking neurodegenerative diseases can open new avenues to better understand the pathological mechanisms underlying these conditions or to identify novel molecular targets for diagnosis and/or therapeutics. In the second research scenario, the easy availability of EVs in body fluids and the specificity of their cargo, which can reflect the cell of origin or disease profiles, turn these into attractive diagnostic tools. EVs with exosome-like characteristics, circulating in the bloodstream and other peripheral biofluids, constitute a non-invasive and rapid alternative to study several conditions, including brain-related disorders. In both cases, several EVs isolation methods are already available, but each neuronal model or biofluid presents its own challenges. Herein, a literature overview on EVs isolation methodologies from distinct neuronal models (cellular culture and brain tissue) and body fluids (serum, plasma, cerebrospinal fluid, urine and saliva) was carried out. Focus was given to approaches employed in the context of Alzheimer's and Parkinson's diseases, and the main research findings discussed. The topics here revised will facilitate the choice of EVs isolation methodologies and potentially prompt new discoveries in EVs research and in the neurodegenerative diseases field.
Collapse
Affiliation(s)
- Margarida Vaz
- Biomarker Discovery TeamNeuroscience and Signalling GroupInstitute of Biomedicine (iBiMED)Department of Medical SciencesUniversity of AveiroAveiroPortugal
| | - Tânia Soares Martins
- Biomarker Discovery TeamNeuroscience and Signalling GroupInstitute of Biomedicine (iBiMED)Department of Medical SciencesUniversity of AveiroAveiroPortugal
| | - Ana Gabriela Henriques
- Biomarker Discovery TeamNeuroscience and Signalling GroupInstitute of Biomedicine (iBiMED)Department of Medical SciencesUniversity of AveiroAveiroPortugal
| |
Collapse
|
14
|
Huang Y, Driedonks TAP, Cheng L, Rajapaksha H, Turchinovich A, Routenberg DA, Nagaraj R, Redding-Ochoa J, Arab T, Powell BH, Pletnikova O, Troncoso JC, Zheng L, Hill AF, Mahairaki V, Witwer KW. Relationships of APOE Genotypes With Small RNA and Protein Cargo of Brain Tissue Extracellular Vesicles From Patients With Late-Stage AD. Neurol Genet 2022; 8:e200026. [PMID: 36405397 PMCID: PMC9667865 DOI: 10.1212/nxg.0000000000200026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/20/2022] [Indexed: 11/24/2022]
Abstract
Background and Objectives Variants of the apolipoprotein E (APOE) gene are the greatest known risk factors for sporadic Alzheimer disease (AD). Three major APOE isoform alleles, ε2, ε3, and ε4, encode and produce proteins that differ by only 1-2 amino acids but have different binding partner interactions. Whereas APOE ε2 is protective against AD relative to ε3, ε4 is associated with an increased risk for AD development. However, the role of APOE in gene regulation in AD pathogenesis has remained largely undetermined. Extracellular vesicles (EVs) are lipid bilayer-delimited particles released by cells to dispose of unwanted materials and mediate intercellular communication, and they are implicated in AD pathophysiology. Brain-derived EVs (bdEVs) could act locally in the tissue and reflect cellular changes. To reveal whether APOE genotype affects EV components in AD brains, bdEVs were separated from patients with AD with different APOE genotypes for parallel small RNA and protein profile. Methods bdEVs from late-stage AD brains (BRAAK stages 5-6) from patients with APOE genotypes ε2/3 (n = 5), ε3/3 (n = 5), ε3/4 (n = 6), and ε4/4 (n = 6) were separated using our published protocol into a 10,000g pelleted extracellular fraction (10K) and a further purified EV fraction. Counting, sizing, and multiomic characterization by small RNA sequencing and proteomic analysis were performed for 10K, EVs, and source tissue. Results Comparing APOE genotypes, no significant differences in bdEV total particle concentration or morphology were observed. Overall small RNA and protein profiles of 10K, EVs, and source tissue also did not differ substantially between different APOE genotypes. However, several differences in individual RNAs (including miRNAs and tRNAs) and proteins in 10K and EVs were observed when comparing the highest and lowest risk groups (ε4/4 and ε2/3). Bioinformatic analysis and previous publications indicate a potential regulatory role of these molecules in AD. Discussion For patients with late-stage AD in this study, only a few moderate differences were observed for small RNA and protein profiles between APOE genotypes. Among these, several newly identified 10K and EV-associated molecules may play roles in AD progression. Possibly, larger genotype-related differences exist and are more apparent in or before earlier disease stages.
Collapse
Affiliation(s)
- Yiyao Huang
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Tom A P Driedonks
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lesley Cheng
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Harinda Rajapaksha
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andrey Turchinovich
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - David A Routenberg
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Rajini Nagaraj
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Javier Redding-Ochoa
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Tanina Arab
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Bonita H Powell
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Olga Pletnikova
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Juan C Troncoso
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lei Zheng
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andrew F Hill
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Vasiliki Mahairaki
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology (Y.H., T.A.P.D., T.A., B.H.P., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Biochemistry and Chemistry (L.C., H.R., A.F.H.), La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia; Molecular Epidemiology (A.T.), German Cancer Research Center DKFZ, Heidelberg, Germany; SciBerg e.Kfm (A.T.), Mannheim, Germany; Meso Scale Diagnostics (D.A.R., R.N.), LLC, Rockville, MD; Department of Pathology (J.R.-O., O.P., J.C.T.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pathology and Anatomical Sciences (O.P.), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY; Department of Neurology (J.C.T., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Laboratory Medicine (L.Z.), Institute of Health and Sport (A.F.H.), Victoria University, Melbourne, Australia; Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Genetic Medicine (V.M.); and Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease (V.M., K.W.W.), Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
15
|
Cechinel LR, Batabyal RA, Freishtat RJ, Zohn IE. Parental obesity-induced changes in developmental programming. Front Cell Dev Biol 2022; 10:918080. [PMID: 36274855 PMCID: PMC9585252 DOI: 10.3389/fcell.2022.918080] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Many studies support the link between parental obesity and the predisposition to develop adult-onset metabolic syndromes that include obesity, high blood pressure, dyslipidemia, insulin resistance, and diabetes in the offspring. As the prevalence of obesity increases in persons of childbearing age, so does metabolic syndrome in their descendants. Understanding how parental obesity alters metabolic programs in the progeny, predisposing them to adult-onset metabolic syndrome, is key to breaking this cycle. This review explores the basis for altered metabolism of offspring exposed to overnutrition by focusing on critical developmental processes influenced by parental obesity. We draw from human and animal model studies, highlighting the adaptations in metabolism that occur during normal pregnancy that become maladaptive with obesity. We describe essential phases of development impacted by parental obesity that contribute to long-term alterations in metabolism in the offspring. These encompass gamete formation, placentation, adipogenesis, pancreas development, and development of brain appetite control circuits. Parental obesity alters the developmental programming of these organs in part by inducing epigenetic changes with long-term consequences on metabolism. While exposure to parental obesity during any of these phases is sufficient to alter long-term metabolism, offspring often experience multiple exposures throughout their development. These insults accumulate to increase further the susceptibility of the offspring to the obesogenic environments of modern society.
Collapse
|
16
|
Thellung S, Corsaro A, Dellacasagrande I, Nizzari M, Zambito M, Florio T. Proteostasis unbalance in prion diseases: Mechanisms of neurodegeneration and therapeutic targets. Front Neurosci 2022; 16:966019. [PMID: 36148145 PMCID: PMC9485628 DOI: 10.3389/fnins.2022.966019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/05/2022] [Indexed: 01/18/2023] Open
Abstract
Transmissible spongiform encephalopathies (TSEs), or prion diseases, are progressive neurodegenerative disorders of the central nervous system that affect humans and animals as sporadic, inherited, and infectious forms. Similarly to Alzheimer's disease and other neurodegenerative disorders, any attempt to reduce TSEs' lethality or increase the life expectancy of affected individuals has been unsuccessful. Typically, the onset of symptoms anticipates the fatal outcome of less than 1 year, although it is believed to be the consequence of a decades-long process of neuronal death. The duration of the symptoms-free period represents by itself a major obstacle to carry out effective neuroprotective therapies. Prions, the infectious entities of TSEs, are composed of a protease-resistant protein named prion protein scrapie (PrPSc) from the prototypical TSE form that afflicts ovines. PrPSc misfolding from its physiological counterpart, cellular prion protein (PrPC), is the unifying pathogenic trait of all TSEs. PrPSc is resistant to intracellular turnover and undergoes amyloid-like fibrillation passing through the formation of soluble dimers and oligomers, which are likely the effective neurotoxic entities. The failure of PrPSc removal is a key pathogenic event that defines TSEs as proteopathies, likewise other neurodegenerative disorders, including Alzheimer's, Parkinson's, and Huntington's disease, characterized by alteration of proteostasis. Under physiological conditions, protein quality control, led by the ubiquitin-proteasome system, and macroautophagy clears cytoplasm from improperly folded, redundant, or aggregation-prone proteins. There is evidence that both of these crucial homeostatic pathways are impaired during the development of TSEs, although it is still unclear whether proteostasis alteration facilitates prion protein misfolding or, rather, PrPSc protease resistance hampers cytoplasmic protein quality control. This review is aimed to critically analyze the most recent advancements in the cause-effect correlation between PrPC misfolding and proteostasis alterations and to discuss the possibility that pharmacological restoring of ubiquitin-proteasomal competence and stimulation of autophagy could reduce the intracellular burden of PrPSc and ameliorate the severity of prion-associated neurodegeneration.
Collapse
Affiliation(s)
- Stefano Thellung
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Alessandro Corsaro
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Irene Dellacasagrande
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Mario Nizzari
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Martina Zambito
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Tullio Florio
- Section of Pharmacology, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- *Correspondence: Tullio Florio
| |
Collapse
|
17
|
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: 54] [Impact Index Per Article: 18.0] [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
|
18
|
Sex Differentially Alters Secretion of Brain Extracellular Vesicles During Aging: A Potential Mechanism for Maintaining Brain Homeostasis. Neurochem Res 2022; 47:3428-3439. [PMID: 35904699 PMCID: PMC9546961 DOI: 10.1007/s11064-022-03701-1] [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: 05/10/2022] [Revised: 06/27/2022] [Accepted: 07/15/2022] [Indexed: 11/06/2022]
Abstract
Extracellular vesicles (EVs) in the brain play a role in neuronal homeostasis by removing intracellular material and regulating cell-to-cell communication. Given that sex and aging differentially modulate brain networks, we investigated sex-dependent differences in EV levels and content in the brain during aging. EVs were isolated from the brains of 3, 6, 12, 18, and 24 month-old female and male C57BL/6 J mice, and the levels of different EV species determined. While the number of plasma membrane-derived microvesicles and a subset of late endosomes-derived exosomes increased with age in the brain of female mice, no significant changes were seen in males. Mitochondria-derived mitovesicles in the brain increased during aging in both sexes, a change that may reflect aging-dependent alterations in mitochondrial function. These findings reveal enhanced turnover during aging in female brains, suggesting a mechanism for advantageous successful female brain aging and sex-depending different susceptibility to age-related neurodegenerative diseases.
Collapse
|
19
|
Soudy R, Kimura R, Fu W, Patel A, Jhamandas J. Extracellular vesicles enriched with amylin receptor are cytoprotective against the Aß toxicity in vitro. PLoS One 2022; 17:e0267164. [PMID: 35421203 PMCID: PMC9009604 DOI: 10.1371/journal.pone.0267164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 04/03/2022] [Indexed: 11/19/2022] Open
Abstract
Extracellular vesicles (EVs) are double membrane structures released by all cell types with identified roles in the generation, transportation, and degradation of amyloid-β protein (Aβ) oligomers in Alzheimer’s disease (AD). EVs are thus increasingly recognized to play a neuroprotective role in AD, through their ability to counteract the neurotoxic effects of Aβ, possibly through interactions with specific receptors on cell membranes. Our previous studies have identified the amylin receptor (AMY), particularly AMY3 subtype, as a mediator of the deleterious actions of Aβ in vitro and in vivo experimental paradigms. In the present study, we demonstrate that AMY3 enriched EVs can bind soluble oligomers of Aß and protect N2a cells against toxic effects of this peptide. The effect was specific to amylin receptor as it was blocked in the presence of amylin receptor antagonist AC253. This notion was supported by reduced Aβ binding to EVs from AMY depleted mice compared to those from wild type (Wt) mice. Finally, application of AMY3, but not Wt derived, EVs to hippocampal brain slices improved Aβ-induced reduction of long-term potentiation, a cellular surrogate of memory. Collectively, our observations support the role of AMY receptors, particularly AMY3, in EVs as a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Rania Soudy
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ryoichi Kimura
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Center for Liberal Arts and Sciences, Sanyo-Onoda City University, Yamaguchi, Japan
| | - Wen Fu
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Aarti Patel
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jack Jhamandas
- Department of Medicine (Neurology), Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- * E-mail:
| |
Collapse
|
20
|
Yamaguchi H, Kawahara H, Kodera N, Kumaki A, Tada Y, Tang Z, Sakai K, Ono K, Yamada M, Hanayama R. Extracellular Vesicles Contribute to the Metabolism of Transthyretin Amyloid in Hereditary Transthyretin Amyloidosis. Front Mol Biosci 2022; 9:839917. [PMID: 35402512 PMCID: PMC8983912 DOI: 10.3389/fmolb.2022.839917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/22/2022] [Indexed: 11/13/2022] Open
Abstract
Hereditary (variant) transthyretin amyloidosis (ATTRv amyloidosis), which is caused by variants in the transthyretin (TTR) gene, leads to TTR amyloid deposits in multiple organs and various symptoms such as limb ataxia, muscle weakness, and cardiac failure. Interaction between amyloid proteins and extracellular vesicles (EVs), which are secreted by various cells, is known to promote the clearance of the proteins, but it is unclear whether EVs are involved in the formation and deposition of TTR amyloid in ATTRv amyloidosis. To clarify the relationship between ATTRv amyloidosis and EVs, serum-derived EVs were analyzed. In this study, we showed that cell-derived EVs are involved in the formation of TTR amyloid deposits on the membrane of small EVs, as well as the deposition of TTR amyloid in cells. Human serum-derived small EVs also altered the degree of aggregation and deposition of TTR. Furthermore, the amount of TTR aggregates in serum-derived small EVs in patients with ATTRv amyloidosis was lower than that in healthy controls. These results indicate that EVs contribute to the metabolism of TTR amyloid, and suggest that TTR in serum-derived small EVs is a potential target for future ATTRv amyloidosis diagnosis and therapy.
Collapse
Affiliation(s)
- Hiroki Yamaguchi
- Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
- Department of Neurology and Neurobiology of Aging, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hironori Kawahara
- Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
- WPI Nano Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Japan
- *Correspondence: Hironori Kawahara, ; Rikinari Hanayama,
| | - Noriyuki Kodera
- WPI Nano Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Ayanori Kumaki
- Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yasutake Tada
- Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
- Department of Neurology and Neurobiology of Aging, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Zixin Tang
- Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kenji Sakai
- Department of Neurology and Neurobiology of Aging, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kenjiro Ono
- Department of Neurology and Neurobiology of Aging, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Masahito Yamada
- Department of Neurology and Neurobiology of Aging, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
- Department of Internal Medicine, Division of Neurology, Kudanzaka Hospital, Tokyo, Japan
| | - Rikinari Hanayama
- Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
- WPI Nano Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Japan
- *Correspondence: Hironori Kawahara, ; Rikinari Hanayama,
| |
Collapse
|
21
|
Picca A, Guerra F, Calvani R, Coelho-Junior HJ, Bucci C, Marzetti E. Circulating extracellular vesicles: friends and foes in neurodegeneration. Neural Regen Res 2022; 17:534-542. [PMID: 34380883 PMCID: PMC8504375 DOI: 10.4103/1673-5374.320972] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/19/2021] [Accepted: 04/21/2021] [Indexed: 12/01/2022] Open
Abstract
Extracellular vesicles have been identified as pivotal mediators of intercellular communication with critical roles in physiological and pathological conditions. Via this route, several molecules (e.g., nucleic acids, proteins, metabolites) can be transferred to proximal and distant targets to convey specific information. Extracellular vesicle-associated cargo molecules have been proposed as markers of several disease conditions for their potential of tracking down the generating cell. Indeed, circulating extracellular vesicles may represent biomarkers of dysfunctional cellular quality control systems especially in conditions characterized by the accrual of intracellular misfolded proteins. Furthermore, the identification of extracellular vesicles as tools for the delivery of nucleic acids or other cargo molecules to diseased tissues makes these circulating shuttles possible targets for therapeutic development. The increasing interest in the study of extracellular vesicles as biomarkers resides mainly in the fact that the identification of peripheral levels of extracellular vesicle-associated proteins might reflect molecular events occurring in hardly accessible tissues, such as the brain, thereby serving as a "brain liquid biopsy". The exploitation of extracellular vesicles for diagnostic and therapeutic purposed might offer unprecedented opportunities to develop personalized approaches. Here, we discuss the bright and dark sides of extracellular vesicles in the setting of two main neurodegenerative diseases (i.e., Parkinson's and Alzheimer's diseases). A special focus will be placed on the possibility of using extracellular vesicles as biomarkers for the two conditions to enable disease tracking and treatment monitoring.
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, Lecce, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Hélio José Coelho-Junior
- Università Cattolica del Sacro Cuore, Institute of Internal Medicine and Geriatrics, Rome, Italy
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, Lecce, Italy
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Institute of Internal Medicine and Geriatrics, Rome, Italy
| |
Collapse
|
22
|
Impact of endolysosomal dysfunction upon exosomes in neurodegenerative diseases. Neurobiol Dis 2022; 166:105651. [DOI: 10.1016/j.nbd.2022.105651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/11/2022] [Accepted: 01/25/2022] [Indexed: 11/22/2022] Open
|
23
|
Mohammadi B, Song F, Matamoros-Angles A, Shafiq M, Damme M, Puig B, Glatzel M, Altmeppen HC. Anchorless risk or released benefit? An updated view on the ADAM10-mediated shedding of the prion protein. Cell Tissue Res 2022; 392:215-234. [PMID: 35084572 PMCID: PMC10113312 DOI: 10.1007/s00441-022-03582-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/12/2022] [Indexed: 11/24/2022]
Abstract
The prion protein (PrP) is a broadly expressed glycoprotein linked with a multitude of (suggested) biological and pathological implications. Some of these roles seem to be due to constitutively generated proteolytic fragments of the protein. Among them is a soluble PrP form, which is released from the surface of neurons and other cell types by action of the metalloprotease ADAM10 in a process termed 'shedding'. The latter aspect is the focus of this review, which aims to provide a comprehensive overview on (i) the relevance of proteolytic processing in regulating cellular PrP functions, (ii) currently described involvement of shed PrP in neurodegenerative diseases (including prion diseases and Alzheimer's disease), (iii) shed PrP's expected roles in intercellular communication in many more (patho)physiological conditions (such as stroke, cancer or immune responses), (iv) and the need for improved research tools in respective (future) studies. Deeper mechanistic insight into roles played by PrP shedding and its resulting fragment may pave the way for improved diagnostics and future therapeutic approaches in diseases of the brain and beyond.
Collapse
Affiliation(s)
- Behnam Mohammadi
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- Working Group for Interdisciplinary Neurobiology and Immunology (INI Research), Hamburg, Germany
| | - Feizhi Song
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Andreu Matamoros-Angles
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Mohsin Shafiq
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Markus Damme
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Berta Puig
- Department of Neurology, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | | |
Collapse
|
24
|
Rabaneda-Bueno R, Mena-Montes B, Torres-Castro S, Torres-Carrillo N, Torres-Carrillo NM. Advances in Genetics and Epigenetic Alterations in Alzheimer's Disease: A Notion for Therapeutic Treatment. Genes (Basel) 2021; 12:1959. [PMID: 34946908 PMCID: PMC8700838 DOI: 10.3390/genes12121959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is a disabling neurodegenerative disorder that leads to long-term functional and cognitive impairment and greatly reduces life expectancy. Early genetic studies focused on tracking variations in genome-wide DNA sequences discovered several polymorphisms and novel susceptibility genes associated with AD. However, despite the numerous risk factors already identified, there is still no fully satisfactory explanation for the mechanisms underlying the onset of the disease. Also, as with other complex human diseases, the causes of low heritability are unclear. Epigenetic mechanisms, in which changes in gene expression do not depend on changes in genotype, have attracted considerable attention in recent years and are key to understanding the processes that influence age-related changes and various neurological diseases. With the recent use of massive sequencing techniques, methods for studying epigenome variations in AD have also evolved tremendously, allowing the discovery of differentially expressed disease traits under different conditions and experimental settings. This is important for understanding disease development and for unlocking new potential AD therapies. In this work, we outline the genomic and epigenomic components involved in the initiation and development of AD and identify potentially effective therapeutic targets for disease control.
Collapse
Affiliation(s)
- Rubén Rabaneda-Bueno
- Biology Centre of the Czech Academy of Sciences, Institute of Hydrobiology, 37005 České Budějovice, Czech Republic
- School of Biological Sciences, James Clerk Maxwell Building, The King’s Buildings Campus, University of Edinburgh, Edinburgh EH9 3FD, UK
| | - Beatriz Mena-Montes
- Laboratorio de Biología del Envejecimiento, Departamento de Investigación Básica, Instituto Nacional de Geriatría, Mexico City 10200, Mexico;
| | - Sara Torres-Castro
- Departamento de Epidemiología Demográfica y Determinantes Sociales, Instituto Nacional de Geriatría, Mexico City 10200, Mexico;
| | - Norma Torres-Carrillo
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (N.T.-C.); (N.M.T.-C.)
| | - Nora Magdalena Torres-Carrillo
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (N.T.-C.); (N.M.T.-C.)
| |
Collapse
|
25
|
Zhang N, He F, Li T, Chen J, Jiang L, Ouyang XP, Zuo L. Role of Exosomes in Brain Diseases. Front Cell Neurosci 2021; 15:743353. [PMID: 34588957 PMCID: PMC8473913 DOI: 10.3389/fncel.2021.743353] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 08/10/2021] [Indexed: 12/16/2022] Open
Abstract
Exosomes are a subset of extracellular vesicles that act as messengers to facilitate communication between cells. Non-coding RNAs, proteins, lipids, and microRNAs are delivered by the exosomes to target molecules (such as proteins, mRNAs, or DNA) of host cells, thereby playing a key role in the maintenance of normal brain function. However, exosomes are also involved in the occurrence, prognosis, and clinical treatment of brain diseases, such as Alzheimer's disease, Parkinson's disease, stroke, and traumatic brain injury. In this review, we have summarized novel findings that elucidate the role of exosomes in the occurrence, prognosis, and treatment of brain diseases.
Collapse
Affiliation(s)
- Nan Zhang
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Hengyang Medical School, Institute of Neuroscience Research, University of South China, Hengyang, China
| | - Fengling He
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Hengyang Medical School, Institute of Neuroscience Research, University of South China, Hengyang, China
| | - Ting Li
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Hengyang Medical School, Institute of Neuroscience Research, University of South China, Hengyang, China
| | - Jinzhi Chen
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Hengyang Medical School, Institute of Neuroscience Research, University of South China, Hengyang, China
| | - Liping Jiang
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Hengyang Medical School, Institute of Neuroscience Research, University of South China, Hengyang, China.,Hunan Taihe Hospital, Changsha, China
| | - Xin-Ping Ouyang
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Hengyang Medical School, Institute of Neuroscience Research, University of South China, Hengyang, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Lielian Zuo
- Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Department of Physiology, Hengyang Medical School, Institute of Neuroscience Research, University of South China, Hengyang, China
| |
Collapse
|
26
|
Wu Q, Cortez L, Kamali-Jamil R, Sim V, Wille H, Kar S. Implications of exosomes derived from cholesterol-accumulated astrocytes in Alzheimer's disease pathology. Dis Model Mech 2021; 14:dmm048929. [PMID: 34524402 PMCID: PMC8560497 DOI: 10.1242/dmm.048929] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 09/06/2021] [Indexed: 12/25/2022] Open
Abstract
Amyloid β (Aβ) peptides generated from the amyloid precursor protein (APP) play a critical role in the development of Alzheimer's disease (AD) pathology. Aβ-containing neuronal exosomes, which represent a novel form of intercellular communication, have been shown to influence the function/vulnerability of neurons in AD. Unlike neurons, the significance of exosomes derived from astrocytes remains unclear. In this study, we evaluated the significance of exosomes derived from U18666A-induced cholesterol-accumulated astrocytes in the development of AD pathology. Our results show that cholesterol accumulation decreases exosome secretion, whereas lowering cholesterol increases exosome secretion, from cultured astrocytes. Interestingly, exosomes secreted from U18666A-treated astrocytes contain higher levels of APP, APP-C-terminal fragments, soluble APP, APP secretases and Aβ1-40 than exosomes secreted from control astrocytes. Furthermore, we show that exosomes derived from U18666A-treated astrocytes can lead to neurodegeneration, which is attenuated by decreasing Aβ production or by neutralizing exosomal Aβ peptide with an anti-Aβ antibody. These results, taken together, suggest that exosomes derived from cholesterol-accumulated astrocytes can play an important role in trafficking APP/Aβ peptides and influencing neuronal viability in the affected regions of the AD brain.
Collapse
Affiliation(s)
- Qi Wu
- Department of Medicine (Neurology), University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Leonardo Cortez
- Department of Medicine (Neurology), University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Razieh Kamali-Jamil
- Department of Biochemistry, Center for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Valerie Sim
- Department of Medicine (Neurology), University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Holger Wille
- Department of Biochemistry, Center for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2G3, Canada
| | - Satyabrata Kar
- Department of Medicine (Neurology), University of Alberta, Edmonton, AB T6G 2G3, Canada
- Department of Biochemistry, Center for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2G3, Canada
| |
Collapse
|
27
|
Landfield Q, Saito M, Hashim A, Canals-Baker S, Sershen H, Levy E, Saito M. Cocaine Induces Sex-Associated Changes in Lipid Profiles of Brain Extracellular Vesicles. Neurochem Res 2021; 46:2909-2922. [PMID: 34245421 PMCID: PMC8490334 DOI: 10.1007/s11064-021-03395-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/01/2021] [Accepted: 07/03/2021] [Indexed: 11/27/2022]
Abstract
Cocaine is a highly addictive stimulant with diverse effects on physiology. Recent studies indicate the involvement of extracellular vesicles (EVs) secreted by neural cells in the cocaine addiction process. It is hypothesized that cocaine affects secretion levels of EVs and their cargos, resulting in modulation of synaptic transmission and plasticity related to addiction physiology and pathology. Lipids present in EVs are important for EV formation and for intercellular lipid exchange that may trigger physiological and pathological responses, including neuroplasticity, neurotoxicity, and neuroinflammation. Specific lipids are highly enriched in EVs compared to parent cells, and recent studies suggest the involvement of various lipids in drug-induced synaptic plasticity during the development and maintenance of addiction processes. Therefore, we examined interstitial small EVs isolated from the brain of mice treated with either saline or cocaine, focusing on the effects of cocaine on the lipid composition of EVs. We demonstrate that 12 days of noncontingent repeated cocaine (10 mg/kg) injections to mice, which induce locomotor sensitization, cause lipid composition changes in brain EVs of male mice as compared with saline-injected controls. The most prominent change is the elevation of GD1a ganglioside in brain EVs of males. However, cocaine does not affect the EV lipid profiles of the brain in female mice. Understanding the relationship between lipid composition in EVs and vulnerability to cocaine addiction may provide insight into novel targets for therapies for addiction.
Collapse
Affiliation(s)
- Qwynn Landfield
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
| | - Mitsuo Saito
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
| | - Audrey Hashim
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
| | - Stefanie Canals-Baker
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
| | - Henry Sershen
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Efrat Levy
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Mariko Saito
- Division of Neurochemistry, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Orangeburg, NY, 10962, USA.
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
28
|
Chen XQ, Xing Z, Chen QD, Salvi RJ, Zhang X, Tycko B, Mobley WC, Yu YE. Mechanistic Analysis of Age-Related Clinical Manifestations in Down Syndrome. Front Aging Neurosci 2021; 13:700280. [PMID: 34276349 PMCID: PMC8281234 DOI: 10.3389/fnagi.2021.700280] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/09/2021] [Indexed: 12/15/2022] Open
Abstract
Down syndrome (DS) is the most common genetic cause of Alzheimer's disease (AD) due to trisomy for all or part of human chromosome 21 (Hsa21). It is also associated with other phenotypes including distinctive facial features, cardiac defects, growth delay, intellectual disability, immune system abnormalities, and hearing loss. All adults with DS demonstrate AD-like brain pathology, including amyloid plaques and neurofibrillary tangles, by age 40 and dementia typically by age 60. There is compelling evidence that increased APP gene dose is necessary for AD in DS, and the mechanism for this effect has begun to emerge, implicating the C-terminal APP fragment of 99 amino acid (β-CTF). The products of other triplicated genes on Hsa21 might act to modify the impact of APP triplication by altering the overall rate of biological aging. Another important age-related DS phenotype is hearing loss, and while its mechanism is unknown, we describe its characteristics here. Moreover, immune system abnormalities in DS, involving interferon pathway genes and aging, predispose to diverse infections and might modify the severity of COVID-19. All these considerations suggest human trisomy 21 impacts several diseases in an age-dependent manner. Thus, understanding the possible aging-related mechanisms associated with these clinical manifestations of DS will facilitate therapeutic interventions in mid-to-late adulthood, while at the same time shedding light on basic mechanisms of aging.
Collapse
Affiliation(s)
- Xu-Qiao Chen
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States
| | - Zhuo Xing
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Quang-Di Chen
- Department of Communicative Disorders and Sciences and Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, United States
| | - Richard J Salvi
- Department of Communicative Disorders and Sciences and Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, United States
| | - Xuming Zhang
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Benjamin Tycko
- Hackensack-Meridian Health Center for Discovery and Innovation, Nutley, NJ, United States.,Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, United States
| | - William C Mobley
- Department of Neurosciences, University of California San Diego, La Jolla, CA, United States
| | - Y Eugene Yu
- The Children's Guild Foundation Down Syndrome Research Program, Genetics and Genomics Program and Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States.,Genetics, Genomics and Bioinformatics Program, State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
29
|
Meldolesi J. Extracellular vesicles (exosomes and ectosomes) play key roles in the pathology of brain diseases. MOLECULAR BIOMEDICINE 2021; 2:18. [PMID: 35006460 PMCID: PMC8607397 DOI: 10.1186/s43556-021-00040-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Last century, neurons and glial cells were mostly believed to play distinct functions, relevant for the brain. Progressively, however, it became clear that neurons, astrocytes and microglia co-operate intensely with each other by release/binding of signaling factors, direct surface binding and generation/release of extracellular vesicles, the exosomes and ectosomes, called together vesicles in this abstract. The present review is focused on these vesicles, fundamental in various brain diseases. Their properties are extraordinary. The specificity of their membrane governs their fusion with distinct target cells, variable depending on the state and specificity of their cells of origin and target. Result of vesicle fusion is the discharge of their cargos into the cytoplasm of target cells. Cargos are composed of critical molecules, from proteins (various nature and function) to nucleotides (especially miRNAs), playing critical roles in immune and neurodegenerative diseases. Among immune diseases is multiple sclerosis, affected by extensive dysregulation of co-trafficking neural and glial vesicles, with distinct miRNAs inducing severe or reducing effects. The vesicle-dependent differences between progressive and relapsing-remitting forms of the disease are relevant for clinical developments. In Alzheimer’s disease the vesicles can affect the brain by changing their generation and inducing co-release of effective proteins, such Aβ and tau, from neurons and astrocytes. Specific miRNAs can delay the long-term development of the disease. Upon their traffic through the blood-brainbarrier, vesicles of various origin reach fluids where they are essential for the identification of biomarkers, important for diagnostic and therapeutic innovations, critical for the future of many brain patients.
Collapse
Affiliation(s)
- Jacopo Meldolesi
- Division of Neuroscience, San Raffaele Institute and Vita-Salute San Raffaele University, via Olgettina 58, 20132, Milan, Italy.
| |
Collapse
|
30
|
Brain-Derived Extracellular Vesicles in Health and Disease: A Methodological Perspective. Int J Mol Sci 2021; 22:ijms22031365. [PMID: 33573018 PMCID: PMC7866382 DOI: 10.3390/ijms22031365] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are double membrane structures released by presumably all cell types that transport and deliver lipids, proteins, and genetic material to near or distant recipient cells, thereby affecting their phenotype. The basic knowledge of their functions in healthy and diseased brain is still murky and many questions about their biology are unsolved. In neurological diseases, EVs are regarded as attractive biomarkers and as therapeutic tools due to their ability to cross the blood–brain barrier (BBB). EVs have been successfully isolated from conditioned media of primary brain cells and cerebrospinal fluid (CSF), but protocols allowing for the direct study of pathophysiological events mediated or influenced by EVs isolated from brain have only recently been published. This review aims to give a brief overview of the current knowledge of EVs’ functions in the central nervous system (CNS) and the current protocols to isolate brain-derived EVs (BDEVs) used in different publications. By comparing the proteomic analysis of some of these publications, we also assess the influence of the isolation method on the protein content of BDEVs.
Collapse
|
31
|
Gomes AR, Sangani NB, Fernandes TG, Diogo MM, Curfs LMG, Reutelingsperger CP. Extracellular Vesicles in CNS Developmental Disorders. Int J Mol Sci 2020; 21:E9428. [PMID: 33322331 PMCID: PMC7763819 DOI: 10.3390/ijms21249428] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022] Open
Abstract
The central nervous system (CNS) is the most complex structure in the body, consisting of multiple cell types with distinct morphology and function. Development of the neuronal circuit and its function rely on a continuous crosstalk between neurons and non-neural cells. It has been widely accepted that extracellular vesicles (EVs), mainly exosomes, are effective entities responsible for intercellular CNS communication. They contain membrane and cytoplasmic proteins, lipids, non-coding RNAs, microRNAs and mRNAs. Their cargo modulates gene and protein expression in recipient cells. Several lines of evidence indicate that EVs play a role in modifying signal transduction with subsequent physiological changes in neurogenesis, gliogenesis, synaptogenesis and network circuit formation and activity, as well as synaptic pruning and myelination. Several studies demonstrate that neural and non-neural EVs play an important role in physiological and pathological neurodevelopment. The present review discusses the role of EVs in various neurodevelopmental disorders and the prospects of using EVs as disease biomarkers and therapeutics.
Collapse
Affiliation(s)
- Ana Rita Gomes
- Department of Bioengineering and IBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (A.R.G.); (T.G.F.); (M.M.D.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Department of Biochemistry, Maastricht University, Cardiovascular Research Institute Maastricht, 6200 MD Maastricht, The Netherlands;
- GKC-Rett Expertise Centre, Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands;
| | - Nasim Bahram Sangani
- Department of Biochemistry, Maastricht University, Cardiovascular Research Institute Maastricht, 6200 MD Maastricht, The Netherlands;
- GKC-Rett Expertise Centre, Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands;
| | - Tiago G. Fernandes
- Department of Bioengineering and IBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (A.R.G.); (T.G.F.); (M.M.D.)
| | - M. Margarida Diogo
- Department of Bioengineering and IBB—Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal; (A.R.G.); (T.G.F.); (M.M.D.)
| | - Leopold M. G. Curfs
- GKC-Rett Expertise Centre, Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands;
| | - Chris P. Reutelingsperger
- Department of Biochemistry, Maastricht University, Cardiovascular Research Institute Maastricht, 6200 MD Maastricht, The Netherlands;
- GKC-Rett Expertise Centre, Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands;
| |
Collapse
|
32
|
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
|
33
|
Pérez-González R, Kim Y, Miller C, Pacheco-Quinto J, Eckman EA, Levy E. Extracellular vesicles: where the amyloid precursor protein carboxyl-terminal fragments accumulate and amyloid-β oligomerizes. FASEB J 2020; 34:12922-12931. [PMID: 32772431 PMCID: PMC7496786 DOI: 10.1096/fj.202000823r] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/11/2020] [Accepted: 07/20/2020] [Indexed: 12/20/2022]
Abstract
Pleiotropic roles are proposed for brain extracellular vesicles (EVs) in the development of Alzheimer's disease (AD). Our previous studies have suggested a beneficial role for EVs in AD, where the endosomal system in vulnerable neurons is compromised, contributing to the removal of accumulated material from neurons. However, the involvement of EVs in propagating AD amyloidosis throughout the brain has been considered because the amyloid‐β precursor protein (APP), APP metabolites, and key APP cleaving enzymes were identified in association with EVs. Here, we undertook to determine whether the secretase machinery is actively processing APP in EVs isolated from the brains of wild‐type and APP overexpressing Tg2576 mice. We found that full‐length APP is cleaved in EVs incubated in the absence of cells. The resulting metabolites, both α‐ and β‐APP carboxyl‐terminal fragments and APP intracellular domain accumulate in EVs over time and amyloid‐β dimerizes. Thus, EVs contribute to the removal from neurons and transport of APP‐derived neurotoxic peptides. While this is potentially a venue for propagation of the pathology throughout the brain, it may contribute to efficient removal of neurotoxic peptides from the brain.
Collapse
Affiliation(s)
- Rocío Pérez-González
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Yohan Kim
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Chelsea Miller
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, USA
| | - Javier Pacheco-Quinto
- Biomedical Research Institute of New Jersey, Cedar Knolls, and Atlantic Health Systems, Morristown, NJ, USA
| | - Elizabeth A Eckman
- Biomedical Research Institute of New Jersey, Cedar Knolls, and Atlantic Health Systems, Morristown, NJ, USA
| | - Efrat Levy
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University School of Medicine, New York, NY, USA.,Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA.,Neuroscience Institute, New York University School of Medicine, New York, NY, USA
| |
Collapse
|