1
|
Manolopoulos A, Yao PJ, Kapogiannis D. Extracellular vesicles: translational research and applications in neurology. Nat Rev Neurol 2025; 21:265-282. [PMID: 40181198 DOI: 10.1038/s41582-025-01080-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2025] [Indexed: 04/05/2025]
Abstract
Over the past few decades, extensive basic, translational and clinical research has been devoted to deciphering the physiological and pathogenic roles of extracellular vesicles (EVs) in the nervous system. The presence of brain cell-derived EVs in the blood, carrying diverse cargoes, has enabled the development of predictive, diagnostic, prognostic, disease-monitoring and treatment-response biomarkers for various neurological disorders. In this Review, we consider how EV biomarkers can bring us closer to understanding the complex pathogenesis of neurological disorders such as Alzheimer disease, Parkinson disease, stroke, traumatic brain injury, amyotrophic lateral sclerosis and multiple sclerosis. We describe how translational research on EVs might unfold bidirectionally, proceeding from basic to clinical studies but also in the opposite direction, with biomarker findings in the clinic leading to novel hypotheses that can be tested in the laboratory. We demonstrate the potential value of EVs across all stages of the therapeutic development pipeline, from identifying therapeutic targets to the use of EVs as reporters in model systems and biomarkers in clinical research. Finally, we discuss how the cargo and physicochemical properties of naturally occurring and custom-engineered EVs can be leveraged as novel treatments and vehicles for drug delivery, potentially revolutionizing neurotherapeutics.
Collapse
Affiliation(s)
- Apostolos Manolopoulos
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, Baltimore, MD, USA
| | - Pamela J Yao
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, Baltimore, MD, USA
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, Baltimore, MD, USA.
| |
Collapse
|
2
|
Chen J, Tian C, Xiong X, Yang Y, Zhang J. Extracellular vesicles: new horizons in neurodegeneration. EBioMedicine 2025; 113:105605. [PMID: 40037089 PMCID: PMC11925178 DOI: 10.1016/j.ebiom.2025.105605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/28/2025] [Accepted: 02/05/2025] [Indexed: 03/06/2025] Open
Abstract
Extracellular vesicles (EVs) are lipid-enclosed nanovesicles secreted by diverse cell types that orchestrate intercellular communication through cargo delivery. Their pivotal roles span from supporting the development of normal central nervous system (CNS) to contributing to the pathogenesis of neurological diseases. Particularly noteworthy is their involvement in the propagation of pathogenic proteins, such as those involved in neurodegenerative disorders, and nucleic acids, closely linking them to disease onset and progression. Moreover, EVs have emerged as promising diagnostic biomarkers for neurological disorders and as tools for disease staging, owing to their ability to traverse the blood-brain barrier and their specific, stable, and accessible properties. This review comprehensively explores the realm of CNS-derived EVs found in peripheral blood, encompassing their detection methods, transport mechanisms, and diverse roles in various neurodegenerative diseases. Furthermore, we evaluate the potentials and limitations of EVs in clinical applications and highlight prospective research directions in this rapidly evolving field.
Collapse
Affiliation(s)
- Jun Chen
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310002, China
| | - Chen Tian
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310002, China
| | - Xiao Xiong
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310002, China
| | - Ying Yang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310002, China.
| | - Jing Zhang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310002, China; National Human Brain Bank for Health and Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310002, China.
| |
Collapse
|
3
|
Wang M, Liu Z, Wang W, Chopp M, Millman M, Li Y, Cepparulo P, Kemper A, Li C, Zhang L, Zhang Y, Zhang ZG. Enhanced Small Extracellular Vesicle Uptake by Activated Interneurons Improves Stroke Recovery in Mice. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70036. [PMID: 40134760 PMCID: PMC11934211 DOI: 10.1002/jex2.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/07/2025] [Accepted: 01/24/2025] [Indexed: 03/27/2025]
Abstract
Neuronal circuitry remodelling, which comprises excitatory and inhibitory neurons, is critical for improving neurological outcomes after a stroke. Preclinical studies have shown that small extracellular vesicles (sEVs) have a therapeutic effect on stroke recovery. However, it is highly challenging to use sEVs to specifically target individual neuronal populations to enhance neuronal circuitry remodelling after stroke. In the present study, using a chemogenetic approach to specifically activate peri-infarct cortical interneurons in combination with the administration of sEVs derived from cerebral endothelial cells (CEC-sEVs), we showed that the CEC-sEVs were preferentially taken up by the activated neurons, leading to significant improvement of functional outcome after stroke, which was associated with augmentation of peri-infarct cortical axonal/dendritic outgrowth and of axonal remodelling of the corticospinal tract. The ultrastructural and Western blot analyses revealed that neurons with internalization of CEC-sEVs exhibited significantly reduced numbers of damaged mitochondria and proteins that mediate dysfunctional mitochondria, respectively. Together, these data indicate that the augmented uptake of CEC-sEVs by activated peri-infarct cortical interneurons facilitates neuronal circuitry remodelling and functional recovery after stroke, which has the potential to be a novel therapy for improving stroke recovery.
Collapse
Affiliation(s)
- Mingjin Wang
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | - Zhongwu Liu
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | - Weida Wang
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | - Michael Chopp
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
- Department of PhysicsOakland UniversityRochesterMichiganUSA
| | - Michael Millman
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | - Yanfeng Li
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | | | - Amy Kemper
- Department of PathologyHenry Ford HospitalDetroitMichiganUSA
| | - Chao Li
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | - Li Zhang
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | - Yi Zhang
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | | |
Collapse
|
4
|
Counil H, Silva RO, Rabanel J, Zaouter C, Haddad M, Ben Khedher MR, Brambilla D, Fülöp T, Patten SA, Ramassamy C. Brain penetration of peripheral extracellular vesicles from Alzheimer's patients and induction of microglia activation. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70027. [PMID: 39830834 PMCID: PMC11740088 DOI: 10.1002/jex2.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/13/2024] [Accepted: 12/08/2024] [Indexed: 01/22/2025]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative pathology. Brain-derived extracellular vesicles (EVs) have been demonstrated to be implicated in AD pathogenesis by facilitating the propagation of Tau, amyloid-β and inflammatory cytokines. However, the impact of peripheral EVs (pEVs) in AD pathogenesis remains poorly investigated. The objective of our study was to compare the passage of pEVs from adults, cognitively healthy elderly, and AD patients through the blood-brain barrier (BBB), to evaluate their uptake in the brain and to assess their impact on the microglia activity using in vitro and in vivo models. To this end, pEVs were enriched, characterized, and fluorescently labelled. The passage of pEVs through the endothelial bEnd.3 cells was studied in a Transwell device with either neuronal or microglia cells seeded at the bottom of the well. Following the internalization of pEVs from AD patients, microglia adopted an amoeboid morphology and released a heightened level of pro-inflammatory cytokine IL-6. To further assess their in vivo transport across the BBB, pEVs were injected into the blood circulation of 2-days post-fertilization Tg(flk1:EGFP) zebrafish. The biodistribution of pEVs was monitored at 1 and 24 h post-injection using confocal microscopy. We demonstrated that pEVs traverse the BBB by transcytosis and subsequently diffuse progressively into the brain. pEVs were then internalized by neuronal and radial glial cells as seen in Tg(huc:EGFP) and Tg(gfap:EGFP) zebrafish, respectively. Additional experiments were performed with the intrahippocampal injection of pEVs in the mouse, indicating their spreading throughout the brain and their uptake by neuronal and glial cells. These findings contribute to novel insights into the fate of pEVs following their passage through the BBB in vitro and in vivo, and demonstrate for the first time that pEVs from AD patients affect microglia activity. This suggests a potential mechanism through which peripheral tissue cues may contribute to AD pathogenesis.
Collapse
Affiliation(s)
- Hermine Counil
- INRSCentre Armand‐Frappier Santé BiotechnologieLavalQuebecCanada
| | | | - Jean‐Michel Rabanel
- INRSCentre Armand‐Frappier Santé BiotechnologieLavalQuebecCanada
- School of Pharmaceutical Sciences, Faculty of MedecineUniversity of OttawaOttawaOntarioCanada
| | | | - Mohamed Haddad
- INRSCentre Armand‐Frappier Santé BiotechnologieLavalQuebecCanada
| | - Mohamed Raâfet Ben Khedher
- INRSCentre Armand‐Frappier Santé BiotechnologieLavalQuebecCanada
- Higher Institute of Biotechnology of BejaUniversity of JendoubaBejaTunisia
| | - Davide Brambilla
- Université de MontréalFaculté de Pharmacie, Pavillon Jean‐CoutuMontréalQuebecCanada
| | - Tamas Fülöp
- Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health SciencesUniversity of SherbrookeSherbrookeQuebecCanada
| | | | | |
Collapse
|
5
|
Pfaffl MW. Combining Cre-LoxP and single-cell sequencing technologies: insights into the extracellular vesicle cargo transfer. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:614-617. [PMID: 39811732 PMCID: PMC11725425 DOI: 10.20517/evcna.2024.58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 01/16/2025]
Abstract
The recent study from the Pogge von Strandmann group published in Cellular and Molecular Immunology, by Alashkar Alhamwe et al., combined for the first time the Cre-LoxP recombination system with single-cell sequencing. The group monitored the tumor-derived extracellular vesicle (EV) uptake and the EV functions in the recipient non-malignant cells in a pancreatic ductal adenocarcinoma mouse model. Recombination events and EV uptake, together with resulting gene expression changes in macrophages, neutrophils, and mast cells, were detected by single-cell sequencing technology of the tumor tissue. This new approach is highly specific, as it can identify single EV recipient cells without interfering with the EV biogenesis or the phenotype.
Collapse
Affiliation(s)
- Michael W. Pfaffl
- Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Freising 85354, Germany.
| |
Collapse
|
6
|
Buck AH, Nolte-'t Hoen ENM. The Nature and Nurture of Extracellular Vesicle-Mediated Signaling. Annu Rev Genet 2024; 58:409-432. [PMID: 39231450 DOI: 10.1146/annurev-genet-111523-102725] [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] [Indexed: 09/06/2024]
Abstract
In the last decade, it has become clear that extracellular vesicles (EVs) are a ubiquitous component of living systems. These small membrane-enclosed particles can confer diverse functions to the cells that release, capture, or coexist with them in an environment. We use examples across living systems to produce a conceptual framework that classifies three modes by which EVs exert functions: (a) EV release that serves a function for producing cells, (b) EV modification of the extracellular environment, and (c) EV interactions with, and alteration of, receiving cells. We provide an overview of the inherent properties of EVs (i.e., their nature) as well as factors in the environment and receiving cell (i.e., nurture) that determine whether transmission of EV cargo leads to functional cellular responses. This review broadens the context for ruminating on EV functions and highlights the emergent properties of EVs that define their role in biology and will shape their applications in medicine.
Collapse
Affiliation(s)
- Amy H Buck
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom;
| | - Esther N M Nolte-'t Hoen
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands;
| |
Collapse
|
7
|
Spanos M, Gokulnath P, Li G, Hutchins E, Meechoovet B, Sheng Q, Chatterjee E, Sharma R, Carnel-Amar N, Lin C, Azzam C, Ghaeli I, Amancherla KV, Victorino JF, Garcia-Mansfield K, Pfeffer R, Sahu P, Lindman BR, Elmariah S, Gamazon ER, Betti MJ, Bledsoe X, Lance ML, Absi T, Su YR, Do N, Contreras MG, Varrias D, Kladas M, Radulovic M, Tsiachris D, Spanos A, Tsioufis K, Ellinor PT, Tucker NR, Januzzi JL, Pirrotte P, Jovanovic-Talisman T, Van Keuren-Jensen K, Shah R, Das S. Cardiomyocyte-derived circulating extracellular vesicles allow a non-invasive liquid biopsy of myocardium in health and disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.19.24314009. [PMID: 39371135 PMCID: PMC11451713 DOI: 10.1101/2024.09.19.24314009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The ability to track disease without tissue biopsy in patients is a major goal in biology and medicine. Here, we identify and characterize cardiomyocyte-derived extracellular vesicles in circulation (EVs; "cardiovesicles") through comprehensive studies of induced pluripotent stem cell-derived cardiomyocytes, genetic mouse models, and state-of-the-art mass spectrometry and low-input transcriptomics. These studies identified two markers (POPDC2, CHRNE) enriched on cardiovesicles for biotinylated antibody-based immunocapture. Captured cardiovesicles were enriched in canonical cardiomyocyte transcripts/pathways with distinct profiles based on human disease type (heart failure, myocardial infarction). In paired myocardial tissue-plasma from patients, highly expressed genes in cardiovesicles were largely cardiac-enriched (vs. "bulk" EVs, which were more organ non-specific) with high expression in myocardial tissue by single nuclear RNA-seq, largely in cardiomyocytes. These results demonstrate the first "liquid" biopsy discovery platform to interrogate cardiomyocyte states noninvasively in model systems and in human disease, allowing non-invasive characterization of cardiomyocyte biology for discovery and therapeutic applications.
Collapse
Affiliation(s)
- Michail Spanos
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Albert Einstein College of Medicine/ North Central Bronx/Jacobi Medical Center, New York City Health and Hospitals, The Bronx, NY, USA
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | - Priyanka Gokulnath
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Elizabeth Hutchins
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, AZ, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Bessie Meechoovet
- Division of Neurogenomics, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Quanhu Sheng
- Department of Biostatistics (Q.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Emeli Chatterjee
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ritin Sharma
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Natacha Carnel-Amar
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Claire Lin
- Division of Cardiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Christopher Azzam
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ima Ghaeli
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Kaushik V Amancherla
- Vanderbilt Translational and Clinical Research Center (R.S.), Vanderbilt University Medical Center, Nashville, TN, USA
| | - José Fabian Victorino
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Krystine Garcia-Mansfield
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Ryan Pfeffer
- Masonic Medical Research Institute, Utica, NY, USA 13501
| | - Parul Sahu
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian R. Lindman
- Vanderbilt Translational and Clinical Research Center (R.S.), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sammy Elmariah
- Department of Medicine, Cardiovascular Division, University of California-San Francisco, San Francisco, CA, USA
| | - Eric R. Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Michael J. Betti
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Xavier Bledsoe
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | | | - Tarek Absi
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yan Ru Su
- Vanderbilt Translational and Clinical Research Center (R.S.), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ngoc Do
- Spectradyne LLC, Signal Hill, CA, USA
| | - Marta Garcia Contreras
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Dimitrios Varrias
- Albert Einstein College of Medicine/ Jacobi Medical Center, The Bronx, NY, USA
| | - Michail Kladas
- Albert Einstein College of Medicine/ North Central Bronx/Jacobi Medical Center, New York City Health and Hospitals, The Bronx, NY, USA
| | - Miroslav Radulovic
- Albert Einstein College of Medicine/ North Central Bronx/Jacobi Medical Center, New York City Health and Hospitals, The Bronx, NY, USA
| | - Dimitris Tsiachris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | | | - Konstantinos Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | - Patrick T. Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - James L. Januzzi
- Division of Cardiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Baim Institute for Clinical Research, Boston, Massachusetts, USA
| | - Patrick Pirrotte
- Collaborative Center for Translational Mass Spectrometry, Translational Genomics Research Institute, Phoenix, AZ, USA
- Cancer & Cell Biology Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Tijana Jovanovic-Talisman
- Department of Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Ravi Shah
- Vanderbilt Translational and Clinical Research Center (R.S.), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Oliveira Silva R, Counil H, Rabanel JM, Haddad M, Zaouter C, Ben Khedher MR, Patten SA, Ramassamy C. Donepezil-Loaded Nanocarriers for the Treatment of Alzheimer's Disease: Superior Efficacy of Extracellular Vesicles Over Polymeric Nanoparticles. Int J Nanomedicine 2024; 19:1077-1096. [PMID: 38317848 PMCID: PMC10843980 DOI: 10.2147/ijn.s449227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/07/2024] [Indexed: 02/07/2024] Open
Abstract
Introduction Drug delivery across the blood-brain barrier (BBB) is challenging and therefore severely restricts neurodegenerative diseases therapy such as Alzheimer's disease (AD). Donepezil (DNZ) is an acetylcholinesterase (AChE) inhibitor largely prescribed to AD patients, but its use is limited due to peripheral adverse events. Nanodelivery strategies with the polymer Poly (lactic acid)-poly(ethylene glycol)-based nanoparticles (NPs-PLA-PEG) and the extracellular vesicles (EVs) were developed with the aim to improve the ability of DNZ to cross the BBB, its brain targeting and efficacy. Methods EVs were isolated from human plasma and PLA-PEG NPs were synthesized by nanoprecipitation. The toxicity, brain targeting capacity and cholinergic activities of the formulations were evaluated both in vitro and in vivo. Results EVs and NPs-PLA-PEG were designed to be similar in size and charge, efficiently encapsulated DNZ and allowed sustained drug release. In vitro study showed that both formulations EVs-DNZ and NPs-PLA-PEG-DNZ were highly internalized by the endothelial cells bEnd.3. These cells cultured on the Transwell® model were used to analyze the transcytosis of both formulations after validation of the presence of tight junctions, the transendothelial electrical resistance (TEER) values and the permeability of the Dextran-FITC. In vivo study showed that both formulations were not toxic to zebrafish larvae (Danio rerio). However, hyperactivity was evidenced in the NPs-PLA-PEG-DNZ and free DNZ groups but not the EVs-DNZ formulations. Biodistribution analysis in zebrafish larvae showed that EVs were present in the brain parenchyma, while NPs-PLA-PEG remained mainly in the bloodstream. Conclusion The EVs-DNZ formulation was more efficient to inhibit the AChE enzyme activity in the zebrafish larvae head. Thus, the bioinspired delivery system (EVs) is a promising alternative strategy for brain-targeted delivery by substantially improving the activity of DNZ for the treatment of AD.
Collapse
Affiliation(s)
- Rummenigge Oliveira Silva
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Hermine Counil
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | | | - Mohamed Haddad
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Charlotte Zaouter
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Mohamed Raâfet Ben Khedher
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
- Higher Institute of Biotechnology of Beja, University of Jendouba, Beja, Tunisia
| | - Shunmoogum A Patten
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Charles Ramassamy
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| |
Collapse
|
9
|
Hermann DM, Peruzzotti-Jametti L, Giebel B, Pluchino S. Extracellular vesicles set the stage for brain plasticity and recovery by multimodal signalling. Brain 2024; 147:372-389. [PMID: 37768167 PMCID: PMC10834259 DOI: 10.1093/brain/awad332] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/07/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Extracellular vesicles (EVs) are extremely versatile naturally occurring membrane particles that convey complex signals between cells. EVs of different cellular sources are capable of inducing striking therapeutic responses in neurological disease models. Differently from pharmacological compounds that act by modulating defined signalling pathways, EV-based therapeutics possess multiple abilities via a variety of effectors, thus allowing the modulation of complex disease processes that may have very potent effects on brain tissue recovery. When applied in vivo in experimental models of neurological diseases, EV-based therapeutics have revealed remarkable effects on immune responses, cell metabolism and neuronal plasticity. This multimodal modulation of neuroimmune networks by EVs profoundly influences disease processes in a highly synergistic and context-dependent way. Ultimately, the EV-mediated restoration of cellular functions helps to set the stage for neurological recovery. With this review we first outline the current understanding of the mechanisms of action of EVs, describing how EVs released from various cellular sources identify their cellular targets and convey signals to recipient cells. Then, mechanisms of action applicable to key neurological conditions such as stroke, multiple sclerosis and neurodegenerative diseases are presented. Pathways that deserve attention in specific disease contexts are discussed. We subsequently showcase considerations about EV biodistribution and delineate genetic engineering strategies aiming at enhancing brain uptake and signalling. By sketching a broad view of EV-orchestrated brain plasticity and recovery, we finally define possible future clinical EV applications and propose necessary information to be provided ahead of clinical trials. Our goal is to provide a steppingstone that can be used to critically discuss EVs as next generation therapeutics for brain diseases.
Collapse
Affiliation(s)
- Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, D-45122 Essen, Germany
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0NN, UK
| | - Bernd Giebel
- Institute of Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Stefano Pluchino
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge CB2 0AH, UK
| |
Collapse
|
10
|
Lu D, Wu JP, Yang QW, Wang HY, Yang JJ, Zhang GG, Wang C, Yang YL, Zhu L, Sun XZ. Recent advances in lipid nanovesicles for targeted treatment of spinal cord injury. Front Bioeng Biotechnol 2023; 11:1261288. [PMID: 37691909 PMCID: PMC10486273 DOI: 10.3389/fbioe.2023.1261288] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
The effective regeneration and functional restoration of damaged spinal cord tissue have been a long-standing concern in regenerative medicine. Treatment of spinal cord injury (SCI) is challenging due to the obstruction of the blood-spinal cord barrier (BSCB), the lack of targeting of drugs, and the complex pathophysiology of injury sites. Lipid nanovesicles, including cell-derived nanovesicles and synthetic lipid nanovesicles, are highly biocompatible and can penetrate BSCB, and are therefore effective delivery systems for targeted treatment of SCI. We summarize the progress of lipid nanovesicles for the targeted treatment of SCI, discuss their advantages and challenges, and provide a perspective on the application of lipid nanovesicles for SCI treatment. Although most of the lipid nanovesicle-based therapy of SCI is still in preclinical studies, this low immunogenicity, low toxicity, and highly engineerable nanovesicles will hold great promise for future spinal cord injury treatments.
Collapse
Affiliation(s)
- Di Lu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nano-science and Technology, Beijing, China
| | - Jiu-Ping Wu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi-Wei Yang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nano-science and Technology, Beijing, China
| | - Hua-Yi Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nano-science and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jun-Jie Yang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gang-Gang Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chen Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nano-science and Technology, Beijing, China
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, China
| | - Yan-Lian Yang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nano-science and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ling Zhu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, National Center for Nano-science and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xin-Zhi Sun
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Ben Khedher MR, Haddad M, Fulop T, Laurin D, Ramassamy C. Implication of Circulating Extracellular Vesicles-Bound Amyloid-β42 Oligomers in the Progression of Alzheimer's Disease. J Alzheimers Dis 2023; 96:813-825. [PMID: 37840502 DOI: 10.3233/jad-230823] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
BACKGROUND The perplex interrelation between circulating extracellular vesicles (cEVs) and amyloid-β (Aβ) deposits in the context of Alzheimer's disease (AD) is poorly understood. OBJECTIVE This study aims to 1) analyze the possible cross-linkage of the neurotoxic amyloid-β oligomers (oAβ) to the human cEVs, 2) identify cEVs corona proteins associated with oAβ binding, and 3) analyze the distribution and expression of targeted cEVs proteins in preclinical participants converted to AD 5 years later (Pre-AD). METHODS cEVs were isolated from 15 Pre-AD participants and 15 healthy controls selected from the Canadian Study of Health and Aging. Biochemical, clinical, lipid, and inflammatory profiles were measured. oAβ and cEVs interaction was determined by nanoparticle tracking analysis and proteinase K digestion. cEVs bound proteins were determined by ELISA. RESULTS oAβ were trapped by cEVs and were topologically bound to their external surface. We identified surface-exposed proteins functionally able to conjugate oAβ including apolipoprotein J (apoJ), apoE and RAGE, with apoJ being 30- to 130-fold higher than RAGE and apoE, respectively. The expression of cEVs apoJ was significantly lower in Pre-AD up to 5 years before AD onset. CONCLUSION Our findings suggest that cEVs might participate in oAβ clearance and that early dysregulation of cEVs could increase the risk of conversion to AD.
Collapse
Affiliation(s)
- Mohamed Raâfet Ben Khedher
- INRS-Centre Armand-Frappier Santé-Biotechnologie, Laval, QC, Canada
- Institute of Nutrition and Functional Foods, Québec, QC, Canada
- Higher Institute of Biotechnology of Beja, University of Jendouba, Beja, Tunisia
| | - Mohamed Haddad
- INRS-Centre Armand-Frappier Santé-Biotechnologie, Laval, QC, Canada
- Institute of Nutrition and Functional Foods, Québec, QC, Canada
| | - Tamas Fulop
- Department of Medicine, Geriatric Division, Research Center on Aging, Sherbrooke University, Sherbrooke, QC, Canada
| | - Danielle Laurin
- Institute of Nutrition and Functional Foods, Québec, QC, Canada
- Centre d'Excellence Sur le Vieillissement de Québec, CHU de Québec-Université Laval Research Centre, VI-TAM-Centre de Recherche en Santé Durable, Québec, QC, Canada
- Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Charles Ramassamy
- INRS-Centre Armand-Frappier Santé-Biotechnologie, Laval, QC, Canada
- Institute of Nutrition and Functional Foods, Québec, QC, Canada
| |
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: 10] [Impact Index Per Article: 3.3] [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
|
Li W, Wang J, Yin X, Shi H, Sun B, Ji M, Song H, Liu J, Dou Y, Xu C, Jiang X, Li J, Li L, Zhang CY, Zhang Y. Construction of a mouse model that can be used for tissue-specific EV screening and tracing in vivo. Front Cell Dev Biol 2022; 10:1015841. [DOI: 10.3389/fcell.2022.1015841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/08/2022] [Indexed: 11/21/2022] Open
Abstract
Extracellular vesicles (EVs) play an important role in the communication between tissues and cells. However, it is difficult to screen and trace EVs secreted by specific tissues in vivo, which affects the functional study of EVs in certain tissues under pathophysiological conditions. In this study, a Cre-dependent CD63flag-EGFP co-expressed with mCherry protein system expressing mice was constructed, which can be used for the secretion, movement, and sorting of EVs from specific tissues in vivo. This mouse model is an ideal research tool for studying the secretion amount, target tissue, and functional molecule screening of EVs in specific tissues under different pathophysiological conditions. Moreover, it provides a new research method to clarify the mechanism of secreted EVs in the pathogenesis of the disease.
Collapse
|
14
|
Progress and gaps of extracellular vesicle-mediated intercellular cargo transfer in the central nervous system. Commun Biol 2022; 5:1223. [PMID: 36369335 PMCID: PMC9652383 DOI: 10.1038/s42003-022-04050-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
A fundamentally novel function proposed for extracellular vesicles (EVs) is to transfer bioactive molecules in intercellular signaling. In this minireview, we discuss recent progress on EV-mediated cargo transfer in the central nervous system (CNS) and major gaps in previous studies. We also suggest a set of experiments necessary for bridging the gaps and establishing the physiological roles of EV-mediated cargo transfer.
Collapse
|
15
|
Synovial Fluid-Derived Extracellular Vesicles of Patients with Arthritides Contribute to Hippocampal Synaptic Dysfunctions and Increase with Mood Disorders Severity in Humans. Cells 2022; 11:cells11152276. [PMID: 35892573 PMCID: PMC9331474 DOI: 10.3390/cells11152276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 01/27/2023] Open
Abstract
Arthritides are a highly heterogeneous group of disorders that include two major clinical entities, localized joint disorders such as osteoarthritis (OA) and systemic autoimmune-driven diseases such as rheumatoid arthritis (RA). Arthritides are characterized by chronic debilitating musculoskeletal conditions and systemic chronic inflammation. Poor mental health is also one of the most common comorbidities of arthritides. Depressive symptoms which are most prevalent, negatively impact patient global assessment diminishing the probability of achieving the target of clinical remission. Here, we investigated new insights into mechanisms that link different joint disorders to poor mental health, and to this issue, we explored the action of the synovial fluid-derived extracellular vesicles (EVs) on neuronal function. Our data show that the exposure of neurons to different concentrations of EVs derived from both RA and OA synovial fluids (RA-EVs and OA-EVs) leads to increased excitatory synaptic transmission but acts on specific modifications on excitatory or inhibitory synapses, as evidenced by electrophysiological and confocal experiments carried out in hippocampal cultures. The treatment of neurons with EVs membrane is also responsible for generating similar effects to those found with intact EVs suggesting that changes in neuronal ability arise upon EVs membrane molecules′ interactions with neurons. In humans with arthritides, we found that nearly half of patients (37.5%) showed clinically significant psychiatric symptoms (CGIs score ≥ 3), and at least mild anxiety (HAM-A ≥ 7) or depression (MADRS and HAM-D ≥ 7); interestingly, these individuals revealed an increased concentration of synovial EVs. In conclusion, our data showing opposite changes at the excitatory and inhibitory levels in neurons treated with OA- and RA-EVs, lay the scientific basis for personalized medicine in OA and RA patients, and identify EVs as new potential actionable biomarkers in patients with OA/RA with poor mental health.
Collapse
|
16
|
Ran N, Li W, Zhang R, Lin C, Zhang J, Wei Z, Li Z, Yuan Z, Wang M, Fan B, Shen W, Li X, Zhou H, Yao X, Kong X, Feng S. Autologous exosome facilitates load and target delivery of bioactive peptides to repair spinal cord injury. Bioact Mater 2022; 25:766-782. [PMID: 37056263 PMCID: PMC10086682 DOI: 10.1016/j.bioactmat.2022.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/03/2022] [Accepted: 07/03/2022] [Indexed: 11/30/2022] Open
Abstract
Spinal cord injury (SCI) causes motor, sensory and automatic impairment due to rarely axon regeneration. Developing effective treatment for SCI in the clinic is extremely challenging because of the restrictive axonal regenerative ability and disconnection of neural elements after injury, as well as the limited systemic drug delivery efficiency caused by blood spinal cord barrier. To develop an effective non-invasive treatment strategy for SCI in clinic, we generated an autologous plasma exosome (AP-EXO) based biological scaffold where AP-EXO was loaded with neuron targeting peptide (RVG) and growth-facilitating peptides (ILP and ISP). This scaffold can be targeted delivered to neurons in the injured area and elicit robust axon regrowth across the lesion core to the levels over 30-fold greater than naïve treatment, thus reestablish the intraspinal circuits and promote motor functional recovery after spinal cord injury in mice. More importantly, in ex vivo, human plasma exosomes (HP-EXO) loaded with combinatory peptides of RVG, ILP and ISP showed safety and no liver and kidney toxicity in the application to nude SCI mice. Combining the efficacy and safety, the AP-EXO-based personalized treatment confers functional recovery after SCI and showed immense promising in biomedical applications in treating SCI. It is helpful to expand the application of combinatory peptides and human plasma derived autologous exosomes in promoting regeneration and recovery upon SCI treatment.
Collapse
Affiliation(s)
- Ning Ran
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wenxiang Li
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Renjie Zhang
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Caorui Lin
- Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Jianping Zhang
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhijian Wei
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zonghao Li
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhongze Yuan
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Min Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Baoyou Fan
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenyuan Shen
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xueying Li
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hengxing Zhou
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xue Yao
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaohong Kong
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Medical Molecular Virology, School of Medicine, Nankai University, Tianjin, China
- Corresponding author. Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Shiqing Feng
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Corresponding author. Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
17
|
Extracellular Vesicles at CNS barriers: Mode of action. Curr Opin Neurobiol 2022; 75:102569. [PMID: 35667340 DOI: 10.1016/j.conb.2022.102569] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 01/08/2023]
Abstract
The exchange of molecules between the brain and periphery is limited by cellular barriers such as the blood-brain barrier (BBB) and the blood-CSF barrier (BCB). Extracellular vesicles (EVs) secreted by brain cells or circulating in the blood stream interact with these barriers and provide a pathway for brain-periphery communication. This review briefly summarizes the main current concepts of EVs signaling over the BBB/BCB. EVs can either be released by barrier cells upon stimulation, act on barrier cells modulating barrier properties, or cross the barrier transferring cargo between the circulation and the brain. The mechanisms of EV signaling and passage over the BBB are increasingly being explored, with inflammation being a main driver. EVs acting at or through the barriers possess wide-ranging effects on brain-periphery communication in both healthy and pathological states. A deeper understanding of the mechanisms of action is important for translation into biomedical applications for brain diseases.
Collapse
|
18
|
Nicholson S, Baccarelli A, Prada D. Role of brain extracellular vesicles in air pollution-related cognitive impairment and neurodegeneration. ENVIRONMENTAL RESEARCH 2022; 204:112316. [PMID: 34728237 PMCID: PMC8671239 DOI: 10.1016/j.envres.2021.112316] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 09/15/2021] [Accepted: 10/28/2021] [Indexed: 05/07/2023]
Abstract
A relationship between environmental exposure to air pollution and cognitive impairment and neurological disorders has been described. Previous literature has focused on the direct effects of the air pollution components on neuronal and glial cells, as well as on involvement of oxidative stress and neuroinflammation on microglia and astrocyte reactivity. However, other mechanisms involved in the air pollution effects on central nervous system (CNS) toxicity can be playing critical roles. Increasingly, extracellular vesicle's (EVs) mediated intercellular communication is being recognized as impacting the development of cognitive impairment and neurological disorders like Alzheimer's disease and others. Here we describe the available evidence about toxic air pollutants and its components on brain, an involvement of brain cells specific and EVs types (based in the origin or in the size of EVs) in the initiation, exacerbation, and propagation of the neurotoxic effects (inflammation, neurodegeneration, and accumulation of neurotoxic proteins) induced by air pollution in the CNS. Additionally, we discuss the identification and isolation of neural-derived EVs from human plasma, the most common markers for neural-derived EVs, and their potential for use as diagnostic or therapeutic molecules for air pollution-related cognitive impairment and neurodegeneration.
Collapse
Affiliation(s)
- Stacia Nicholson
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, 10032, USA
| | - Andrea Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, 10032, USA
| | - Diddier Prada
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, 10032, USA; Instituto Nacional de Cancerología, Mexico City, 14080, Mexico.
| |
Collapse
|
19
|
Exosomal microRNAs have great potential in the neurorestorative therapy for traumatic brain injury. Exp Neurol 2022; 352:114026. [DOI: 10.1016/j.expneurol.2022.114026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/09/2022] [Accepted: 02/22/2022] [Indexed: 11/19/2022]
|
20
|
Ghoroghi S, Mary B, Asokan N, Goetz JG, Hyenne V. Tumor extracellular vesicles drive metastasis (it's a long way from home). FASEB Bioadv 2021; 3:930-943. [PMID: 34761175 PMCID: PMC8565230 DOI: 10.1096/fba.2021-00079] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
Among a plethora of functions, extracellular vesicles released by primary tumors spread in the organism and reach distant organs where they can induce the formation of a premetastatic niche. This constitutes a favorable microenvironment for circulating tumor cells which facilitates their seeding and colonization. In this review, we describe the journey of extracellular vesicles (EVs) from the primary tumor to the future metastatic organ, with a focus on the mechanisms used by EVs to target organs with a specific tropism (i.e., organotropism). We then highlight important tumor EV cargos in the context of premetastatic niche formation and summarize their known effects on extracellular matrix remodeling, angiogenesis, vessel permeabilization, resident cell activation, recruitment of foreign cells, and ultimately the formation of a pro-inflammatory and immuno-tolerant microenvironment. Finally, we discuss current experimental limitations and remaining opened questions in light of metastatic diagnosis and potential therapies targeting PMN formation.
Collapse
Affiliation(s)
- Shima Ghoroghi
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
| | - Benjamin Mary
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
| | - Nandini Asokan
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
| | - Jacky G Goetz
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
| | - Vincent Hyenne
- Tumor Biomechanics INSERM UMR_S1109 Strasbourg France
- Université de Strasbourg Strasbourg France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg France
- Equipe Labellisée Ligue Contre le Cancer Strasbourg France
- CNRS SNC5055 Strasbourg France
| |
Collapse
|
21
|
Valkov N, Das A, Tucker NR, Li G, Salvador AM, Chaffin MD, Pereira De Oliveira Junior G, Kur I, Gokulnath P, Ziegler O, Yeri A, Lu S, Khamesra A, Xiao C, Rodosthenous R, Srinivasan S, Toxavidis V, Tigges J, Laurent LC, Momma S, Kitchen R, Ellinor P, Ghiran I, Das S. SnRNA sequencing defines signaling by RBC-derived extracellular vesicles in the murine heart. Life Sci Alliance 2021; 4:4/12/e202101048. [PMID: 34663679 PMCID: PMC8548207 DOI: 10.26508/lsa.202101048] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 12/21/2022] Open
Abstract
In a unique model of fluorescent based mapping of EV recipient cells, RBC-EVs were found to signal to cardiac cells and regulate gene expression in a model of ischemic heart failure. Extracellular vesicles (EVs) mediate intercellular signaling by transferring their cargo to recipient cells, but the functional consequences of signaling are not fully appreciated. RBC-derived EVs are abundant in circulation and have been implicated in regulating immune responses. Here, we use a transgenic mouse model for fluorescence-based mapping of RBC-EV recipient cells to assess the role of this intercellular signaling mechanism in heart disease. Using fluorescent-based mapping, we detected an increase in RBC-EV–targeted cardiomyocytes in a murine model of ischemic heart failure. Single cell nuclear RNA sequencing of the heart revealed a complex landscape of cardiac cells targeted by RBC-EVs, with enrichment of genes implicated in cell proliferation and stress signaling pathways compared with non-targeted cells. Correspondingly, cardiomyocytes targeted by RBC-EVs more frequently express cellular markers of DNA synthesis, suggesting the functional significance of EV-mediated signaling. In conclusion, our mouse model for mapping of EV-recipient cells reveals a complex cellular network of RBC-EV–mediated intercellular communication in ischemic heart failure and suggests a functional role for this mode of intercellular signaling.
Collapse
Affiliation(s)
- Nedyalka Valkov
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Avash Das
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Nathan R Tucker
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.,Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA.,Masonic Medical Research Institute, Utica, NY, USA
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Ane M Salvador
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Mark D Chaffin
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA
| | | | - Ivan Kur
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Priyanka Gokulnath
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Olivia Ziegler
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Ashish Yeri
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Shulin Lu
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Aushee Khamesra
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Chunyang Xiao
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Srimeenakshi Srinivasan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | | | - John Tigges
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Louise C Laurent
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Stefan Momma
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Robert Kitchen
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.,Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA
| | - Ionita Ghiran
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
22
|
Bittel M, Reichert P, Sarfati I, Dressel A, Leikam S, Uderhardt S, Stolzer I, Phu TA, Ng M, Vu NK, Tenzer S, Distler U, Wirtz S, Rothhammer V, Neurath MF, Raffai RL, Günther C, Momma S. Visualizing transfer of microbial biomolecules by outer membrane vesicles in microbe-host-communication in vivo. J Extracell Vesicles 2021; 10:e12159. [PMID: 34664784 PMCID: PMC8524437 DOI: 10.1002/jev2.12159] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/29/2022] Open
Abstract
The intestinal microbiota influences mammalian host physiology in health and disease locally in the gut but also in organs devoid of direct contact with bacteria such as the liver and brain. Extracellular vesicles (EVs) or outer membrane vesicles (OMVs) released by microbes are increasingly recognized for their potential role as biological shuttle systems for inter-kingdom communication. However, physiologically relevant evidence for the transfer of functional biomolecules from the intestinal microbiota to individual host cells by OMVs in vivo is scarce. By introducing Escherichia coli engineered to express Cre-recombinase (E. coliCre ) into mice with a Rosa26.tdTomato-reporter background, we leveraged the Cre-LoxP system to report the transfer of bacterial OMVs to recipient cells in vivo. Colonizing the intestine of these mice with E. coliCre , resulted in Cre-recombinase induced fluorescent reporter gene-expression in cells along the intestinal epithelium, including intestinal stem cells as well as mucosal immune cells such as macrophages. Furthermore, even far beyond the gut, bacterial-derived Cre induced extended marker gene expression in a wide range of host tissues, including the heart, liver, kidney, spleen, and brain. Together, our findings provide a method and proof of principle that OMVs can serve as a biological shuttle system for the horizontal transfer of functional biomolecules between bacteria and mammalian host cells.
Collapse
Affiliation(s)
- Miriam Bittel
- Department of Medicine 1Friedrich‐Alexander‐University Erlangen‐NürnbergErlangenGermany
| | - Patrick Reichert
- Department of Medicine 1Friedrich‐Alexander‐University Erlangen‐NürnbergErlangenGermany
| | - Ilann Sarfati
- Department of Medicine 1Friedrich‐Alexander‐University Erlangen‐NürnbergErlangenGermany
- Deutsches Zentrum ImmuntherapieFriedrich‐Alexander University Erlangen‐NürnbergErlangenGermany
| | - Anja Dressel
- Department of Medicine 1Friedrich‐Alexander‐University Erlangen‐NürnbergErlangenGermany
- Deutsches Zentrum ImmuntherapieFriedrich‐Alexander University Erlangen‐NürnbergErlangenGermany
| | - Stefanie Leikam
- Department of Medicine 1Friedrich‐Alexander‐University Erlangen‐NürnbergErlangenGermany
| | - Stefan Uderhardt
- Department of Internal Medicine 3University Hospital Erlangen and Friedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)ErlangenGermany
- Exploratory Research UnitOptical Imaging Centre ErlangenFriedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Iris Stolzer
- Department of Medicine 1Friedrich‐Alexander‐University Erlangen‐NürnbergErlangenGermany
- Deutsches Zentrum ImmuntherapieFriedrich‐Alexander University Erlangen‐NürnbergErlangenGermany
| | - Tuan Anh Phu
- Northern California Institute for Research and EducationSan FranciscoCaliforniaUSA
| | - Martin Ng
- Northern California Institute for Research and EducationSan FranciscoCaliforniaUSA
| | - Ngan K. Vu
- Northern California Institute for Research and EducationSan FranciscoCaliforniaUSA
| | - Stefan Tenzer
- Institute of ImmunologyUniversity Medical Centre of the Johannes‐Gutenberg University MainzMainzGermany
- Research Centre for Immunotherapy (FZI)University Medical Center of the Johannes‐Gutenberg University MainzMainzGermany
| | - Ute Distler
- Institute of ImmunologyUniversity Medical Centre of the Johannes‐Gutenberg University MainzMainzGermany
- Research Centre for Immunotherapy (FZI)University Medical Center of the Johannes‐Gutenberg University MainzMainzGermany
| | - Stefan Wirtz
- Department of Medicine 1Friedrich‐Alexander‐University Erlangen‐NürnbergErlangenGermany
- Deutsches Zentrum ImmuntherapieFriedrich‐Alexander University Erlangen‐NürnbergErlangenGermany
| | - Veit Rothhammer
- Neurology Department (Experimental Glia Biology)University Hospital Erlangen and Friedrich‐Alexander‐University Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Markus F. Neurath
- Department of Medicine 1Friedrich‐Alexander‐University Erlangen‐NürnbergErlangenGermany
- Deutsches Zentrum ImmuntherapieFriedrich‐Alexander University Erlangen‐NürnbergErlangenGermany
| | - Robert L. Raffai
- Department of SurgeryDivision of Vascular and Endovascular SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of Veterans AffairsSurgical Service (112G)San Francisco VA Medical CentreSan FranciscoCaliforniaUSA
| | - Claudia Günther
- Department of Medicine 1Friedrich‐Alexander‐University Erlangen‐NürnbergErlangenGermany
- Deutsches Zentrum ImmuntherapieFriedrich‐Alexander University Erlangen‐NürnbergErlangenGermany
| | - Stefan Momma
- Institute of Neurology (Edinger Institute)Goethe UniversityFrankfurt am MainGermany
| |
Collapse
|
23
|
Verweij FJ, Balaj L, Boulanger CM, Carter DRF, Compeer EB, D'Angelo G, El Andaloussi S, Goetz JG, Gross JC, Hyenne V, Krämer-Albers EM, Lai CP, Loyer X, Marki A, Momma S, Nolte-'t Hoen ENM, Pegtel DM, Peinado H, Raposo G, Rilla K, Tahara H, Théry C, van Royen ME, Vandenbroucke RE, Wehman AM, Witwer K, Wu Z, Wubbolts R, van Niel G. The power of imaging to understand extracellular vesicle biology in vivo. Nat Methods 2021; 18:1013-1026. [PMID: 34446922 PMCID: PMC8796660 DOI: 10.1038/s41592-021-01206-3] [Citation(s) in RCA: 204] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 05/20/2021] [Indexed: 01/08/2023]
Abstract
Extracellular vesicles (EVs) are nano-sized lipid bilayer vesicles released by virtually every cell type. EVs have diverse biological activities, ranging from roles in development and homeostasis to cancer progression, which has spurred the development of EVs as disease biomarkers and drug nanovehicles. Owing to the small size of EVs, however, most studies have relied on isolation and biochemical analysis of bulk EVs separated from biofluids. Although informative, these approaches do not capture the dynamics of EV release, biodistribution, and other contributions to pathophysiology. Recent advances in live and high-resolution microscopy techniques, combined with innovative EV labeling strategies and reporter systems, provide new tools to study EVs in vivo in their physiological environment and at the single-vesicle level. Here we critically review the latest advances and challenges in EV imaging, and identify urgent, outstanding questions in our quest to unravel EV biology and therapeutic applications.
Collapse
Affiliation(s)
- Frederik J Verweij
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France.
- GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France.
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - David R F Carter
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK
- Evox Therapeutics Limited, Oxford Science Park, Oxford, UK
| | - Ewoud B Compeer
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, UK
| | - Gisela D'Angelo
- Institut Curie, PSL Research University, CNRS, UMR144 Cell Biology and Cancer, Paris, France
| | - Samir El Andaloussi
- Evox Therapeutics Limited, Oxford Science Park, Oxford, UK
- Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jacky G Goetz
- INSERM UMR_S1109, Tumor Biomechanics Lab, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Equipe Labellisée Ligue contre le Cancer, Strasbourg, France
| | | | - Vincent Hyenne
- INSERM UMR_S1109, Tumor Biomechanics Lab, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Equipe Labellisée Ligue contre le Cancer, Strasbourg, France
- CNRS SNC5055, Strasbourg, France
| | - Eva-Maria Krämer-Albers
- Johannes Gutenberg-Universität Mainz, Institute of Developmental Biology and Neurobiology, Mainz, Germany
| | - Charles P Lai
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei, Taiwan
| | - Xavier Loyer
- Université de Paris, PARCC, INSERM, Paris, France
| | - Alex Marki
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Stefan Momma
- Institute of Neurology (Edinger Institute), Goethe-University, Frankfurt am Main, Germany
| | - Esther N M Nolte-'t Hoen
- Department of Biomolecular Health Sciences, Faculty of veterinary medicine, Utrecht University, Utrecht, the Netherlands
| | - D Michiel Pegtel
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Hector Peinado
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Graça Raposo
- Institut Curie, PSL Research University, CNRS, UMR144 Cell Biology and Cancer, Paris, France
| | - Kirsi Rilla
- University of Eastern Finland, Institute of Biomedicine, Kuopio, Finland
| | - Hidetoshi Tahara
- Department of Cellular and Molecular Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Clotilde Théry
- Institut Curie, PSL Research University, INSERM U932, Immunity and Cancer, Paris, France
| | | | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Ann M Wehman
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Kenneth Witwer
- Department of Molecular and Comparative Pathobiology and Neurology and the Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhiwei Wu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
- Medical School, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| | - Richard Wubbolts
- Department of Biomolecular Health Sciences, Faculty of veterinary medicine, Utrecht University, Utrecht, the Netherlands
| | - Guillaume van Niel
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France.
- GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France.
| |
Collapse
|
24
|
Schnatz A, Müller C, Brahmer A, Krämer‐Albers E. Extracellular Vesicles in neural cell interaction and CNS homeostasis. FASEB Bioadv 2021; 3:577-592. [PMID: 34377954 PMCID: PMC8332475 DOI: 10.1096/fba.2021-00035] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/15/2022] Open
Abstract
Central nervous system (CNS) homeostasis critically depends on the interaction between neurons and glia cells. Extracellular vesicles (EVs) recently emerged as versatile messengers in CNS cell communication. EVs are released by neurons and glia in activity-dependent manner and address multiple target cells within and outside the nervous system. Here, we summarize the recent advances in understanding the physiological roles of EVs in the nervous system and their ability to deliver signals across the CNS barriers. In addition to the disposal of cellular components via EVs and clearance by phagocytic cells, EVs are involved in plasticity-associated processes, mediate trophic support and neuroprotection, promote axonal maintenance, and modulate neuroinflammation. While individual functional components of the EV cargo are becoming progressively identified, the role of neural EVs as compound multimodal signaling entities remains to be elucidated. Novel transgenic models and imaging technologies allow EV tracking in vivo and provide further insight into EV targeting and their mode of action. Overall, EVs represent key players in the maintenance of CNS homeostasis essential for the lifelong performance of neural networks and thus provide a wide spectrum of biomedical applications.
Collapse
Affiliation(s)
- Andrea Schnatz
- Institute of Developmental Biology and NeurobiologyBiology of Extracellular VesiclesUniversity of MainzMainzGermany
| | - Christina Müller
- Institute of Developmental Biology and NeurobiologyBiology of Extracellular VesiclesUniversity of MainzMainzGermany
| | - Alexandra Brahmer
- Institute of Developmental Biology and NeurobiologyBiology of Extracellular VesiclesUniversity of MainzMainzGermany
| | - Eva‐Maria Krämer‐Albers
- Institute of Developmental Biology and NeurobiologyBiology of Extracellular VesiclesUniversity of MainzMainzGermany
| |
Collapse
|
25
|
Momma S. Extracellular vesicles for remote brain repair. Curr Opin Genet Dev 2021; 70:61-65. [PMID: 34153928 DOI: 10.1016/j.gde.2021.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/19/2021] [Accepted: 06/01/2021] [Indexed: 11/28/2022]
Abstract
Recovery of brain function lost to disease or in old age is a challenging task in regenerative medicine. In the last two decades, therapeutic strategies have undergone significant shifts by a succession of major discoveries from adult neural stem cells and neurogenesis to the development of induced pluripotent stem cells to technologies for reprogramming cells in vitro and in vivo. Now, extracellular vesicles, small membrane-bound vesicles released by all cells and containing lipids, proteins, and nucleic acids, emerge as the next major technological opportunity. While substantial progress has been made on their potential use in therapy and EVs have entered many clinical trials, major aspects of their physiological role, in particular regarding their influence on brain function, remain unknown. However, a better understanding of their actual in vivo function, scope of communication, and possibilities to alter cellular processes in target cells will be needed. This review places EVs in the developing landscape of strategies for cellular repair of the brain and highlights their potential by looking at some recent progress in our understanding of their function in vivo.
Collapse
Affiliation(s)
- Stefan Momma
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt am Main, Germany.
| |
Collapse
|
26
|
Dutta D, Khan N, Wu J, Jay SM. Extracellular Vesicles as an Emerging Frontier in Spinal Cord Injury Pathobiology and Therapy. Trends Neurosci 2021; 44:492-506. [PMID: 33581883 PMCID: PMC8159852 DOI: 10.1016/j.tins.2021.01.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/28/2020] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are membrane-delimited particles that are secreted by nearly all cell types. EVs mediate crucial physiological functions and pathophysiological processes in the CNS. As carriers of diverse bioactive cargoes (e.g., proteins, lipids, and nucleic acids) that can be modified in response to external stimuli, EVs have emerged as pathological mediators following neurotrauma such as spinal cord injury (SCI). We discuss the roles of endogenous EVs in the CNS as well as crosstalk with peripheral EVs in relation to neurotrauma, with a particular focus on SCI. We then summarize the status of EV-based therapeutic advances in preclinical animal models for these conditions. Finally, we discuss new bioengineering strategies that are poised to enhance CNS-specific therapeutic capabilities of EVs.
Collapse
Affiliation(s)
- Dipankar Dutta
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Niaz Khan
- Department of Anesthesiology, and Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Junfang Wu
- Department of Anesthesiology, and Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA.
| | - Steven M Jay
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; Program in Molecular and Cell Biology, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
27
|
Spinal cord injury alters microRNA and CD81+ exosome levels in plasma extracellular nanoparticles with neuroinflammatory potential. Brain Behav Immun 2021; 92:165-183. [PMID: 33307173 PMCID: PMC7897251 DOI: 10.1016/j.bbi.2020.12.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/26/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) have been implicated mechanistically in the pathobiology of neurodegenerative disorders, including central nervous system injury. However, the role of EVs in spinal cord injury (SCI) has received limited attention to date. Moreover, technical limitations related to EV isolation and characterization methods can lead to misleading or contradictory findings. Here, we examined changes in plasma EVs after mouse SCI at multiple timepoints (1d, 3d, 7d, 14d) using complementary measurement techniques. Plasma EVs isolated by ultracentrifugation (UC) were decreased at 1d post-injury, as shown by nanoparticle tracking analysis (NTA), and paralleled an overall reduction in total plasma extracellular nanoparticles. Western blot (WB) analysis of UC-derived plasma EVs revealed increased expression of the tetraspanin exosome marker, CD81, between 1d and 7d post-injury. To substantiate these findings, we performed interferometric and fluorescence imaging of single, tetraspanin EVs captured directly from plasma with ExoView®. Consistent with WB, we observed significantly increased plasma CD81+ EV count and cargo at 1d post-injury. The majority of these tetraspanin EVs were smaller than 50 nm based on interferometry and were insufficiently resolved by flow cytometry-based detection. At the injury site, there was enhanced expression of EV biogenesis proteins that were also detected in EVs directly isolated from spinal cord tissue by WB. Surface expression of tetraspanins CD9 and CD63 increased in multiple cell types at the injury site; however, astrocyte CD81 expression uniquely decreased, as demonstrated by flow cytometry. UC-isolated plasma EV microRNA cargo was also significantly altered at 1d post-injury with changes similar to that reported in EVs released by astrocytes after inflammatory stimulation. When injected into the lateral ventricle, plasma EVs from SCI mice increased both pro- and anti-inflammatory gene as well as reactive astrocyte gene expression in the brain cortex. These studies provide the first detailed characterization of plasma EV dynamics after SCI and suggest that plasma EVs may be involved in posttraumatic brain inflammation.
Collapse
|
28
|
Li J, Salvador AM, Li G, Valkov N, Ziegler O, Yeri A, Xiao CY, Meechoovet B, Alsop E, Rodosthenous RS, Kundu P, Huan T, Levy D, Tigges J, Pico AR, Ghiran I, Silverman MG, Meng X, Kitchen R, Xu J, Keuren-Jensen KV, Shah R, Xiao J, Das S. Mir-30d Regulates Cardiac Remodeling by Intracellular and Paracrine Signaling. Circ Res 2021; 128:e1-e23. [PMID: 33092465 PMCID: PMC7790887 DOI: 10.1161/circresaha.120.317244] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 10/21/2020] [Indexed: 01/05/2023]
Abstract
RATIONALE Previous translational studies implicate plasma extracellular microRNA-30d (miR-30d) as a biomarker in left ventricular remodeling and clinical outcome in heart failure (HF) patients, although precise mechanisms remain obscure. OBJECTIVE To investigate the mechanism of miR-30d-mediated cardioprotection in HF. METHODS AND RESULTS In rat and mouse models of ischemic HF, we show that miR-30d gain of function (genetic, lentivirus, or agomiR-mediated) improves cardiac function, decreases myocardial fibrosis, and attenuates cardiomyocyte (CM) apoptosis. Genetic or locked nucleic acid-based knock-down of miR-30d expression potentiates pathological left ventricular remodeling, with increased dysfunction, fibrosis, and cardiomyocyte death. RNA sequencing of in vitro miR-30d gain and loss of function, together with bioinformatic prediction and experimental validation in cardiac myocytes and fibroblasts, were used to identify and validate direct targets of miR-30d. miR-30d expression is selectively enriched in cardiomyocytes, induced by hypoxic stress and is acutely protective, targeting MAP4K4 (mitogen-associate protein kinase 4) to ameliorate apoptosis. Moreover, miR-30d is secreted primarily in extracellular vesicles by cardiomyocytes and inhibits fibroblast proliferation and activation by directly targeting integrin α5 in the acute phase via paracrine signaling to cardiac fibroblasts. In the chronic phase of ischemic remodeling, lower expression of miR-30d in the heart and plasma extracellular vesicles is associated with adverse remodeling in rodent models and human subjects and is linked to whole-blood expression of genes implicated in fibrosis and inflammation, consistent with observations in model systems. CONCLUSIONS These findings provide the mechanistic underpinning for the cardioprotective association of miR-30d in human HF. More broadly, our findings support an emerging paradigm involving intercellular communication of extracellular vesicle-contained miRNAs (microRNAs) to transregulate distinct signaling pathways across cell types. Functionally validated RNA biomarkers and their signaling networks may warrant further investigation as novel therapeutic targets in HF.
Collapse
Affiliation(s)
- Jin Li
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Ane M. Salvador
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Nedyalka Valkov
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Olivia Ziegler
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Ashish Yeri
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Chun Yang Xiao
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | - Eric Alsop
- Neurogenomics Division, TGen, Phoenix, AZ 85004, USA
| | - Rodosthenis S. Rodosthenous
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Piyusha Kundu
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Tianxiao Huan
- The Framingham Heart Study and The Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Daniel Levy
- The Framingham Heart Study and The Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - John Tigges
- Division of Allergy and Inflammation, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | | | - Ionita Ghiran
- Division of Allergy and Inflammation, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Michael G. Silverman
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Xiangmin Meng
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Robert Kitchen
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jiahong Xu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | | | - Ravi Shah
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
29
|
Yekula A, Muralidharan K, Rosh Z, Youngkin AE, Kang KM, Balaj L, Carter BS. Liquid Biopsy Strategies to Distinguish Progression from Pseudoprogression and Radiation Necrosis in Glioblastomas. ADVANCED BIOSYSTEMS 2020; 4:e2000029. [PMID: 32484293 PMCID: PMC7708392 DOI: 10.1002/adbi.202000029] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/20/2020] [Indexed: 12/13/2022]
Abstract
Liquid biopsy for the detection and monitoring of central nervous system tumors is of significant clinical interest. At initial diagnosis, the majority of patients with central nervous system tumors undergo magnetic resonance imaging (MRI), followed by invasive brain biopsy to determine the molecular diagnosis of the WHO 2016 classification paradigm. Despite the importance of MRI for long-term treatment monitoring, in the majority of patients who receive chemoradiation therapy for glioblastoma, it can be challenging to distinguish between radiation treatment effects including pseudoprogression, radiation necrosis, and recurrent/progressive disease based on imaging alone. Tissue biopsy-based monitoring is high risk and not always feasible. However, distinguishing these entities is of critical importance for the management of patients and can significantly affect survival. Liquid biopsy strategies including circulating tumor cells, circulating free DNA, and extracellular vesicles have the potential to afford significant useful molecular information at both the stage of diagnosis and monitoring for these tumors. Here, current liquid biopsy-based approaches in the context of tumor monitoring to differentiate progressive disease from pseudoprogression and radiation necrosis are reviewed.
Collapse
Affiliation(s)
- Anudeep Yekula
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | | | - Zachary Rosh
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Anna E. Youngkin
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
- Trinity College of Arts and Sciences, Duke University, Durham, NC, USA
| | - Keiko M. Kang
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
- School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Bob S. Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
30
|
Gharbi T, Zhang Z, Yang GY. The Function of Astrocyte Mediated Extracellular Vesicles in Central Nervous System Diseases. Front Cell Dev Biol 2020; 8:568889. [PMID: 33178687 PMCID: PMC7593543 DOI: 10.3389/fcell.2020.568889] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022] Open
Abstract
Astrocyte activation plays an important role during disease-induced inflammatory response in the brain. Exosomes in the brain could be released from bone marrow (BM)-derived stem cells, neuro stem cells (NSC), mesenchymal stem cells (MSC), etc. We summarized that exosomes release and transport signaling to the target cells, and then produce function. Furthermore, we discussed the pathological interactions between astrocytes and other brain cells, which are related to brain diseases such as stroke, Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS) disease, multiple sclerosis (MS), psychiatric, traumatic brain injury (TBI), etc. We provide up-to-date, comprehensive and valuable information on the involvement of exosomes in brain diseases, which is beneficial for basic researchers and clinical physicians.
Collapse
Affiliation(s)
- Tahereh Gharbi
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zhijun Zhang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
31
|
Kesidou D, da Costa Martins PA, de Windt LJ, Brittan M, Beqqali A, Baker AH. Extracellular Vesicle miRNAs in the Promotion of Cardiac Neovascularisation. Front Physiol 2020; 11:579892. [PMID: 33101061 PMCID: PMC7546892 DOI: 10.3389/fphys.2020.579892] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality worldwide claiming almost 17. 9 million deaths annually. A primary cause is atherosclerosis within the coronary arteries, which restricts blood flow to the heart muscle resulting in myocardial infarction (MI) and cardiac cell death. Despite substantial progress in the management of coronary heart disease (CHD), there is still a significant number of patients developing chronic heart failure post-MI. Recent research has been focused on promoting neovascularisation post-MI with the ultimate goal being to reduce the extent of injury and improve function in the failing myocardium. Cardiac cell transplantation studies in pre-clinical models have shown improvement in cardiac function; nonetheless, poor retention of the cells has indicated a paracrine mechanism for the observed improvement. Cell communication in a paracrine manner is controlled by various mechanisms, including extracellular vesicles (EVs). EVs have emerged as novel regulators of intercellular communication, by transferring molecules able to influence molecular pathways in the recipient cell. Several studies have demonstrated the ability of EVs to stimulate angiogenesis by transferring microRNA (miRNA, miR) molecules to endothelial cells (ECs). In this review, we describe the process of neovascularisation and current developments in modulating neovascularisation in the heart using miRNAs and EV-bound miRNAs. Furthermore, we critically evaluate methods used in cell culture, EV isolation and administration.
Collapse
Affiliation(s)
- Despoina Kesidou
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Paula A. da Costa Martins
- Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, Maastricht, Netherlands
- Faculty of Health, Medicine and Life Sciences, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Leon J. de Windt
- Department of Molecular Genetics, Faculty of Science and Engineering, Maastricht University, Maastricht, Netherlands
| | - Mairi Brittan
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Abdelaziz Beqqali
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew Howard Baker
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
32
|
Mondragón-García I, Flores-Guzmán P, Mayani H. Human cord blood hematopoietic cells acquire neural features when cultured in the presence of neurogenic cytokines. Blood Cells Mol Dis 2020; 85:102485. [PMID: 32836190 DOI: 10.1016/j.bcmd.2020.102485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 10/23/2022]
Abstract
In vitro growth of hematopoietic cells depends on the presence of hematopoietic cytokines. To date, it is unclear if these cells would be able to respond to non-hematopoietic cytokines. In the present study, we have explored this by culturing human hematopoietic cells in presence of neurogenic cytokines. Lineage-negative (Lin-) umbilical cord blood (UCB)-derived cells -enriched for hematopoietic stem and progenitor cells- were cultured in presence of different combinations of hematopoietic cytokines, neurotrophins, epidermal growth factor, fibroblast growth factor, and neurogenic culture media, in a 3-phase culture system. A proportion (1-22%) of Lin- UCB hematopoietic cells normally express neural markers and are capable of responding to neural cytokines. Neural cytokines did not have effects on hematopoietic cell proliferation; however, we observed generation of neural-like cells, assessed by morphology, and a significant increase in the proportion of cells expressing neural markers. Such neural-like cells, however, retained expression of hematopoietic markers. It seems that under our culture conditions, no actual transdifferentiation of hematopoietic cells into neural cells occurred; instead, the cells generated in culture seem to be hematopoietic cells that acquired neural features upon contact with neurogenic factors. The identity of UCB cells that acquired a neural phenotype is still unclear.
Collapse
Affiliation(s)
- Ileana Mondragón-García
- Hematopoietic Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Mexico City, Mexico
| | - Patricia Flores-Guzmán
- Hematopoietic Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Mexico City, Mexico
| | - Hector Mayani
- Hematopoietic Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Mexico City, Mexico.
| |
Collapse
|