1
|
Duan J, Lv A, Guo Z, Liu Q, Tian C, Yang Y, Bi J, Yu X, Peng G, Luo B, Cai Z, Xu B, Fu Y, Zhang J. CX3CR1 +/UCHL1 + microglial extracellular vesicles in blood: a potential biomarker for multiple sclerosis. J Neuroinflammation 2024; 21:254. [PMID: 39385200 PMCID: PMC11465848 DOI: 10.1186/s12974-024-03243-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
In neuroinflammation, distinguishing microglia from macrophages and identifying microglial-specific biomarkers in peripheral blood pose significant challenges. This study comprehensively profiled the extracellular vesicles (EVs) of microglia and macrophages, respectively, revealing co-expressed EVs with UCHL1 and CX3CR1 as EVs derived specifically from microglia in human blood. After extensive validation, using optimized nano flow cytometry, we evaluated plasma CX3CR1+/UCHL1+ EVs across clinical cohorts [multiple sclerosis (MS), HTLV-1 associated myelopathy (HAM), Alzheimer's disease (AD), and Parkinson's disease (PD)], along with established neurodegenerative markers (NMDAR2A and NFL). The findings discovered a notable rise in CX3CR1+/UCHL1+ EVs in MS, particularly heightened in HAM, in contrast to controls. Conversely, AD and PD exhibited unaltered or diminished levels of microglial EVs. An integrated model of CX3CR1+/UCHL1+, NMDAR2A+, and NFL+ EVs demonstrated promising diagnostic potential for distinguishing MS from controls and HAM. As to the disease duration, CX3CR1+/UCHL1+ EVs increased in the initial five years of MS, stabilizing thereafter, whereas NMDAR2A+ and NFL+ EVs remained stable initially but increased significantly in the subsequent five years, suggesting their correlation with disease duration. This study uncovers unique blood microglial EVs with potential as biomarkers for MS diagnosis, differentiation from HAM, and correlation with disease duration.
Collapse
Affiliation(s)
- Jing Duan
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Aowei Lv
- Department of Neurology, Institute of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhen Guo
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qi Liu
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chen Tian
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Ying Yang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jin Bi
- Department of Neurology, Institute of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xintong Yu
- Department of Neurology, Institute of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Guoping Peng
- Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Benyan Luo
- Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhijian Cai
- Institute of Immunology, Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Bin Xu
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Ying Fu
- Department of Neurology, Institute of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China.
| | - Jing Zhang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- National Human Brain Bank for Health and Disease, Zhejiang University, Hangzhou, 310012, China.
| |
Collapse
|
2
|
Cooper CG, Kafetzis KN, Patabendige A, Tagalakis AD. Blood-brain barrier disruption in dementia: Nano-solutions as new treatment options. Eur J Neurosci 2024; 59:1359-1385. [PMID: 38154805 DOI: 10.1111/ejn.16229] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/28/2023] [Accepted: 12/02/2023] [Indexed: 12/30/2023]
Abstract
Candidate drugs targeting the central nervous system (CNS) demonstrate extremely low clinical success rates, with more than 98% of potential treatments being discontinued due to poor blood-brain barrier (BBB) permeability. Neurological conditions were shown to be the second leading cause of death globally in 2016, with the number of people currently affected by neurological disorders increasing rapidly. This increasing trend, along with an inability to develop BBB permeating drugs, is presenting a major hurdle in the treatment of CNS-related disorders, like dementia. To overcome this, it is necessary to understand the structure and function of the BBB, including the transport of molecules across its interface in both healthy and pathological conditions. The use of CNS drug carriers is rapidly gaining popularity in CNS research due to their ability to target BBB transport systems. Further research and development of drug delivery vehicles could provide essential information that can be used to develop novel treatments for neurological conditions. This review discusses the BBB and its transport systems and evaluates the potential of using nanoparticle-based delivery systems as drug carriers for CNS disease with a focus on dementia.
Collapse
Affiliation(s)
| | | | - Adjanie Patabendige
- Department of Biology, Edge Hill University, Ormskirk, UK
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK
| | - Aristides D Tagalakis
- Department of Biology, Edge Hill University, Ormskirk, UK
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
3
|
Afonso GJM, Cavaleiro C, Valero J, Mota SI, Ferreiro E. Recent Advances in Extracellular Vesicles in Amyotrophic Lateral Sclerosis and Emergent Perspectives. Cells 2023; 12:1763. [PMID: 37443797 PMCID: PMC10340215 DOI: 10.3390/cells12131763] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a severe and incurable neurodegenerative disease characterized by the progressive death of motor neurons, leading to paralysis and death. It is a rare disease characterized by high patient-to-patient heterogeneity, which makes its study arduous and complex. Extracellular vesicles (EVs) have emerged as important players in the development of ALS. Thus, ALS phenotype-expressing cells can spread their abnormal bioactive cargo through the secretion of EVs, even in distant tissues. Importantly, owing to their nature and composition, EVs' formation and cargo can be exploited for better comprehension of this elusive disease and identification of novel biomarkers, as well as for potential therapeutic applications, such as those based on stem cell-derived exosomes. This review highlights recent advances in the identification of the role of EVs in ALS etiopathology and how EVs can be promising new therapeutic strategies.
Collapse
Affiliation(s)
- Gonçalo J. M. Afonso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (G.J.M.A.); (C.C.)
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- III-Institute of Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Carla Cavaleiro
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (G.J.M.A.); (C.C.)
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- III-Institute of Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Jorge Valero
- Instituto de Neurociencias de Castilla y León, University of Salamanca, 37007 Salamanca, Spain;
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Department of Cell Biology and Pathology, University of Salamanca, 37007 Salamanca, Spain
| | - Sandra I. Mota
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (G.J.M.A.); (C.C.)
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- III-Institute of Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Elisabete Ferreiro
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (G.J.M.A.); (C.C.)
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- III-Institute of Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| |
Collapse
|
4
|
Balbi M, Bonanno G, Bonifacino T, Milanese M. The Physio-Pathological Role of Group I Metabotropic Glutamate Receptors Expressed by Microglia in Health and Disease with a Focus on Amyotrophic Lateral Sclerosis. Int J Mol Sci 2023; 24:5240. [PMID: 36982315 PMCID: PMC10048889 DOI: 10.3390/ijms24065240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
Microglia cells are the resident immune cells of the central nervous system. They act as the first-line immune guardians of nervous tissue and central drivers of neuroinflammation. Any homeostatic alteration that can compromise neuron and tissue integrity could activate microglia. Once activated, microglia exhibit highly diverse phenotypes and functions related to either beneficial or harmful consequences. Microglia activation is associated with the release of protective or deleterious cytokines, chemokines, and growth factors that can in turn determine defensive or pathological outcomes. This scenario is complicated by the pathology-related specific phenotypes that microglia can assume, thus leading to the so-called disease-associated microglia phenotypes. Microglia express several receptors that regulate the balance between pro- and anti-inflammatory features, sometimes exerting opposite actions on microglial functions according to specific conditions. In this context, group I metabotropic glutamate receptors (mGluRs) are molecular structures that may contribute to the modulation of the reactive phenotype of microglia cells, and this is worthy of exploration. Here, we summarize the role of group I mGluRs in shaping microglia cells' phenotype in specific physio-pathological conditions, including some neurodegenerative disorders. A significant section of the review is specifically focused on amyotrophic lateral sclerosis (ALS) since it represents an entirely unexplored topic of research in the field.
Collapse
Affiliation(s)
- Matilde Balbi
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy (M.M.)
| | - Giambattista Bonanno
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy (M.M.)
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy (M.M.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| | - Marco Milanese
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy (M.M.)
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| |
Collapse
|
5
|
Marinelli S, Marrone MC, Di Domenico M, Marinelli S. Endocannabinoid signaling in microglia. Glia 2022; 71:71-90. [PMID: 36222019 DOI: 10.1002/glia.24281] [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: 04/06/2022] [Revised: 09/02/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022]
Abstract
Microglia, the innate immune cells of the central nervous system (CNS), execute their sentinel, housekeeping and defense functions through a panoply of genes, receptors and released cytokines, chemokines and neurotrophic factors. Moreover, microglia functions are closely linked to the constant communication with other cell types, among them neurons. Depending on the signaling pathway and type of stimuli involved, the outcome of microglia operation can be neuroprotective or neurodegenerative. Accordingly, microglia are increasingly becoming considered cellular targets for therapeutic intervention. Among signals controlling microglia activity, the endocannabinoid (EC) system has been shown to exert a neuroprotective role in many neurological diseases. Like neurons, microglia express functional EC receptors and can produce and degrade ECs. Interestingly, boosting EC signaling leads to an anti-inflammatory and neuroprotective microglia phenotype. Nonetheless, little evidence is available on the microglia-mediated therapeutic effects of EC compounds. This review focuses on the EC signals acting on the CNS microglia in physiological and pathological conditions, namely on the CB1R, CB2R and TRPV1-mediated regulation of microglia properties. It also provides new evidence, which strengthens the understanding of mechanisms underlying the control of microglia functions by ECs. Given the broad expression of the EC system in glial and neuronal cells, the resulting picture is the need for in vivo studies in transgenic mouse models to dissect the contribution of EC microglia signaling in the neuroprotective effects of EC-derived compounds.
Collapse
Affiliation(s)
- Sara Marinelli
- CNR-National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | - Maria Cristina Marrone
- EBRI-Fondazione Rita Levi Montalcini, Rome, Italy.,Ministry of University and Research, Mission Unity for Recovery and Resilience Plan, Rome, Italy
| | - Marina Di Domenico
- EBRI-Fondazione Rita Levi Montalcini, Rome, Italy.,Bio@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
| | | |
Collapse
|
6
|
Emerging Role of Neuron-Glia in Neurological Disorders: At a Glance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3201644. [PMID: 36046684 PMCID: PMC9423989 DOI: 10.1155/2022/3201644] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/05/2022] [Indexed: 11/18/2022]
Abstract
Based on the diverse physiological influence, the impact of glial cells has become much more evident on neurological illnesses, resulting in the origins of many diseases appearing to be more convoluted than previously happened. Since neurological disorders are often random and unknown, hence the construction of animal models is difficult to build, representing a small fraction of people with a gene mutation. As a result, an immediate necessity is grown to work within in vitro techniques for examining these illnesses. As the scientific community recognizes cell-autonomous contributions to a variety of central nervous system illnesses, therapeutic techniques involving stem cells for treating neurological diseases are gaining traction. The use of stem cells derived from a variety of sources is increasingly being used to replace both neuronal and glial tissue. The brain's energy demands necessitate the reliance of neurons on glial cells in order for it to function properly. Furthermore, glial cells have diverse functions in terms of regulating their own metabolic activities, as well as collaborating with neurons via secreted signaling or guidance molecules, forming a complex network of neuron-glial connections in health and sickness. Emerging data reveals that metabolic changes in glial cells can cause morphological and functional changes in conjunction with neuronal dysfunction under disease situations, highlighting the importance of neuron-glia interactions in the pathophysiology of neurological illnesses. In this context, it is required to improve our understanding of disease mechanisms and create potential novel therapeutics. According to research, synaptic malfunction is one of the features of various mental diseases, and glial cells are acting as key ingredients not only in synapse formation, growth, and plasticity but also in neuroinflammation and synaptic homeostasis which creates critical physiological capacity in the focused sensory system. The goal of this review article is to elaborate state-of-the-art information on a few glial cell types situated in the central nervous system (CNS) and highlight their role in the onset and progression of neurological disorders.
Collapse
|
7
|
Cazzaro S, Fang C, Khan H, Witas R, Kee TR, Woo JAA, Kang DE. Slingshot homolog-1 mediates the secretion of small extracellular vesicles containing misfolded proteins by regulating autophagy cargo receptors and actin dynamics. Front Aging Neurosci 2022; 14:933979. [PMID: 36092812 PMCID: PMC9452914 DOI: 10.3389/fnagi.2022.933979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Increasing evidence indicates that the accumulation misfolded proteins in Alzheimer's disease (AD) arises from clearance defects in the autophagy-lysosome pathway. Misfolded proteins such as Aβ and tau are secreted in small extracellular vesicles (i.e., exosomes) and are propagated from cell to cell in part through secreted small extracellular vesicles (sEVs). Recent studies suggest that autophagic activity and exosome secretion are coregulated events, and multiple autophagy-related proteins are found in sEVs, including the cargo receptors Sqstm1/p62 and optineurin. However, whether and how autophagy cargo receptors per se regulate the secretion of sEVs is unknown. Moreover, despite the prominent role of actin dynamics in secretory vesicle release, its role in EV secretion is unknown. In this study, we leveraged the dual axes of Slingshot Homolog-1 (SSH1), which inhibits Sqstm1/p62-mediated autophagy and activates cofilin-mediated actin dynamics, to study the regulation of sEV secretion. Here we show that cargo receptors Sqstm1/p62 and optineurin inhibit sEV secretion, an activity that requires their ability to bind ubiquitinated cargo. Conversely, SSH1 increases sEV secretion by dephosphorylating Sqstm1/p62 at pSer403, the phospho-residue that allows Sqstm1/p62 to bind ubiquitinated cargo. In addition, increasing actin dynamics through the SSH1-cofilin activation pathway also increases sEV secretion, which is mimicked by latrunculin B treatment. Finally, Aβ42 oligomers and mutant tau increase sEV secretion and are physically associated with secreted sEVs. These findings suggest that increasing cargo receptor engagement with autophagic cargo and reducing actin dynamics (i.e., SSH1 inhibition) represents an attractive strategy to promote misfolded protein degradation while reducing sEV-mediated cell to cell spread of pathology.
Collapse
Affiliation(s)
- Sara Cazzaro
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, United States
| | - Cenxiao Fang
- Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, United States
| | - Hirah Khan
- Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, United States
| | - Richard Witas
- Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, United States
| | - Teresa R. Kee
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, United States
| | - Jung-A. A. Woo
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - David E. Kang
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH, United States
| |
Collapse
|
8
|
Hua SQ, Hu JL, Zou FL, Liu JP, Luo HL, Hu DX, Wu LD, Zhang WJ. P2X7 receptor in inflammation and pain. Brain Res Bull 2022; 187:199-209. [PMID: 35850190 DOI: 10.1016/j.brainresbull.2022.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/15/2022] [Accepted: 07/13/2022] [Indexed: 11/02/2022]
Abstract
Different studies have confirmed P2X7 receptor-mediated inflammatory mediators play a key role in the development of pain. P2X7 receptor activation can induce the development of pain by mediating the release of inflammatory mediators. In view of the fact that P2X7 receptor is expressed in the nervous system and immune system, it is closely related to the stability and maintenance of the nervous system function. ATP activates P2X7 receptor, opens non-selective cation channels, activates multiple intracellular signaling, releases multiple inflammatory cytokines, and induces pain. At present, the role of P2X7 receptor in inflammatory response and pain has been widely recognized and affirmed. Therefore, in this paper, we discussed the pathological mechanism of P2X7 receptor-mediated inflammation and pain, focused on the internal relationship between P2X7 receptor and pain. Moreover, we also described the effects of some antagonists on pain relief by inhibiting the activities of P2X7 receptor. Thus, targeting to inhibit activation of P2X7 receptor is expected to become another potential target for the relief of pain.
Collapse
Affiliation(s)
- Shi-Qi Hua
- Nanchang University, Nanchang City 343000, Jiangxi Province, China
| | - Jia-Ling Hu
- Emergency Department, The Second Affiliated Hospital, Nanchang University, Nanchang City 343000, Jiangxi Province, China
| | - Fei-Long Zou
- Gastrointestinal Surgery, The Second Affiliated Hospital, Nanchang University, Nanchang City 343000, Jiangxi Province, China
| | - Ji-Peng Liu
- Gastrointestinal Surgery, The Second Affiliated Hospital, Nanchang University, Nanchang City 343000, Jiangxi Province, China
| | - Hong-Liang Luo
- Gastrointestinal Surgery, The Second Affiliated Hospital, Nanchang University, Nanchang City 343000, Jiangxi Province, China
| | - Dong-Xia Hu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City 343000, Jiangxi Province, China.
| | - Li-Dong Wu
- Emergency Department, The Second Affiliated Hospital, Nanchang University, Nanchang City 343000, Jiangxi Province, China.
| | - Wen-Jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City 343000, Jiangxi Province, China.
| |
Collapse
|
9
|
Raffaele S, Fumagalli M. Dynamics of Microglia Activation in the Ischemic Brain: Implications for Myelin Repair and Functional Recovery. Front Cell Neurosci 2022; 16:950819. [PMID: 35899017 PMCID: PMC9309466 DOI: 10.3389/fncel.2022.950819] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Ischemic stroke is a neurological disorder representing a leading cause of death and permanent disability world-wide, for which effective regenerative treatments are missing. Oligodendrocyte degeneration and consequent myelin disruption are considered major contributing factors to stroke-associated neurological deficits. Therefore, fostering myelin reconstruction by oligodendrocyte precursor cells (OPCs) has emerged as a promising therapeutic approach to enhance functional recovery in stroke patients. A pivotal role in regulating remyelination is played by microglia, the resident immune cells of the brain. Early after stroke, microglial cells exert beneficial functions, promoting OPC recruitment toward the ischemic lesion and preserving myelin integrity. However, the protective features of microglia are lost during disease progression, contributing to remyelination failure. Unveiling the mechanisms driving the pro-remyelination properties of microglia may provide important opportunities for both reducing myelin damage and promoting its regeneration. Here, we summarize recent evidence describing microglia activation kinetics in experimental models of ischemic injury, focusing on the contribution of these innate immune cells to myelin damage and repair. Some molecular signals regulating the pro-regenerative functions of microglia after stroke have been highlighted to provide new possible therapeutic targets involved in the protective functions of these cells. Finally, we analyzed the impact of microglia-to-OPCs communication via extracellular vesicles on post-stroke remyelination and functional recovery. The results collected in this review underline the importance of supporting the pro-remyelination functions of microglial cells after stroke.
Collapse
|
10
|
Cui Y, Yu H, Bu Z, Wen L, Yan L, Feng J. Focus on the Role of the NLRP3 Inflammasome in Multiple Sclerosis: Pathogenesis, Diagnosis, and Therapeutics. Front Mol Neurosci 2022; 15:894298. [PMID: 35694441 PMCID: PMC9175009 DOI: 10.3389/fnmol.2022.894298] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/05/2022] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is initiated with an aberrant innate immune response in the central nervous system (CNS) and is involved in many neurological diseases. Inflammasomes are intracellular multiprotein complexes that can be used as platforms to induce the maturation and secretion of proinflammatory cytokines and pyroptosis, thus playing a pivotal role in neuroinflammation. Among the inflammasomes, the nucleotide-binding oligomerization domain-, leucine-rich repeat- and pyrin domain-containing 3 (NLRP3) inflammasome is well-characterized and contributes to many neurological diseases, such as multiple sclerosis (MS), Alzheimer's disease (AD), and ischemic stroke. MS is a chronic autoimmune disease of the CNS, and its hallmarks include chronic inflammation, demyelination, and neurodegeneration. Studies have demonstrated a relationship between MS and the NLRP3 inflammasome. To date, the pathogenesis of MS is not fully understood, and clinical studies on novel therapies are still underway. Here, we review the activation mechanism of the NLRP3 inflammasome, its role in MS, and therapies targeting related molecules, which may be beneficial in MS.
Collapse
|
11
|
Shi Y, Bao Q, Chen W, Wang L, Peng D, Liu J, Liu Q, Zhang Y, Ji Z, Shen A. Potential Roles of Extracellular Vesicles as Diagnosis Biomarkers and Therapeutic Approaches for Cognitive Impairment in Alzheimer’s Disease. J Alzheimers Dis 2022; 87:1-15. [DOI: 10.3233/jad-215666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cognitive dysfunction, the major clinical manifestation of Alzheimer’s disease (AD), is caused by irreversible progressive neurological dysfunction. With the aging of the population, the incidence of AD is increasing year by year. However, there is neither a simple and accurate early diagnosis method, nor an effective method to alleviate or prevent the occurrence and progression of AD. Extracellular vesicles (EVs) are a number of heterogeneous membrane structures that arise from the endosome system or shed from the plasma membrane. In the brain, almost every kind of cell may have EVs, which are related to cell-cell communication and regulate cellular function. At present, an increasing body of evidence suggests that EVs play a crucial role in the pathogenesis of AD, and it is of great significance to use them as specific biomarkers and novel therapeutic targets for cognitive impairment in AD. This article reviews the potential role of EVs as diagnostic biomarkers and treatments for cognitive dysfunction in AD.
Collapse
Affiliation(s)
- Yun Shi
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Qianqian Bao
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Weidong Chen
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, China
- Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui Hefei, China
| | - Lei Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, China
- Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui Hefei, China
| | - Daiyin Peng
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, China
| | - Jie Liu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Qing Liu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yanchun Zhang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Zhaojie Ji
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Aizong Shen
- Department of Pharmacy, Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
12
|
Chand S, Gowen A, Savine M, Moore D, Clark A, Huynh W, Wu N, Odegaard K, Weyrich L, Bevins RA, Fox HS, Pendyala G, Yelamanchili SV. A comprehensive study to delineate the role of an extracellular vesicle-associated microRNA-29a in chronic methamphetamine use disorder. J Extracell Vesicles 2021; 10:e12177. [PMID: 34913274 PMCID: PMC8674191 DOI: 10.1002/jev2.12177] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 10/27/2021] [Accepted: 11/25/2021] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs), which express a repertoire of cargo molecules (cf. proteins, microRNA, lipids, etc.), have been garnering a prominent role in the modulation of several cellular processes. Here, using both non-human primate and rodent model systems, we provide evidence that brain-derived EV (BDE) miRNA, miR-29a-3p (mir-29a), is significantly increased during chronic methamphetamine (MA) exposure. Further, miR-29a levels show significant increase both with drug-seeking and reinstatement in a rat MA self-administration model. We also show that EV-associated miR-29a is enriched in EV pool comprising of small EVs and exomeres and further plays a critical role in MA-induced inflammation and synaptodendritic damage. Furthermore, treatment with the anti-inflammatory drug ibudilast (AV411), which is known to reduce MA relapse, decreased the expression of miR-29a and subsequently attenuated inflammation and rescued synaptodendritic injury. Finally, using plasma from MUD subjects, we provide translational evidence that EV-miR29a could potentially serve as a biomarker to detect neuronal damage in humans diagnosed with MA use disorder (MUD). In summary, our work suggests that EV-associated miR-29a-3p plays a crucial role in MUD and might be used as a potential blood-based biomarker for detecting chronic inflammation and synaptic damage.
Collapse
Affiliation(s)
- Subhash Chand
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Austin Gowen
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Mason Savine
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Dalia Moore
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Alexander Clark
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Wendy Huynh
- Department of PsychologyUniversity of Nebraska–Lincoln (UNL)LincolnNebraskaUSA
| | - Niming Wu
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Katherine Odegaard
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | | | - Rick A. Bevins
- Department of PsychologyUniversity of Nebraska–Lincoln (UNL)LincolnNebraskaUSA
| | - Howard S. Fox
- Department of Neurological SciencesUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Gurudutt Pendyala
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| | - Sowmya V. Yelamanchili
- Department of AnesthesiologyUniversity of Nebraska Medical Center (UNMC)OmahaNebraskaUSA
| |
Collapse
|
13
|
Ren WJ, Illes P. Involvement of P2X7 receptors in chronic pain disorders. Purinergic Signal 2021; 18:83-92. [PMID: 34799827 PMCID: PMC8850523 DOI: 10.1007/s11302-021-09796-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic pain is caused by cellular damage with an obligatory inflammatory component. In response to noxious stimuli, high levels of ATP leave according to their concentration gradient, the intracellular space through discontinuities generated in the plasma membrane or diffusion through pannexin-1 hemichannels, and activate P2X7Rs localized at peripheral and central immune cells. Because of the involvement of P2X7Rs in immune functions and especially the initiation of macrophage/microglial and astrocytic secretion of cytokines, chemokines, prostaglandins, proteases, reactive oxygen, and nitrogen species as well as the excitotoxic glutamate/ATP, this receptor type has a key role in chronic pain processes. Microglia are equipped with a battery of pattern recognition receptors that detect pathogen-associated molecular patterns (PAMPs) such as lipopolysaccharide (LPS) from bacterial infections or danger associated molecular patterns (DAMPs) such as ATP. The co-stimulation of these receptors leads to the activation of the NLRP3 inflammasome and interleukin-1β (IL-1β) release. In the present review, we invite you to a journey through inflammatory and neuropathic pain, primary headache, and regulation of morphine analgesic tolerance, in the pathophysiology of which P2X7Rs are centrally involved. P2X7R bearing microglia and astrocyte-like cells playing eminent roles in chronic pain will be also discussed.
Collapse
Affiliation(s)
- Wen-Jing Ren
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Peter Illes
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, 04109, Leipzig, Germany.
| |
Collapse
|
14
|
Liu Y, Li J, Bu H, Wang H, Zhang Y, Shen Q, Li M, Lu Z, Rong X, Zheng D, Peng Y. Circular RNA expression alteration identifies a novel circulating biomarker in serum exosomal for detection of alcohol dependence. Addict Biol 2021; 26:e13031. [PMID: 33821559 DOI: 10.1111/adb.13031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/22/2020] [Accepted: 03/05/2021] [Indexed: 12/15/2022]
Abstract
Alcohol dependence (AD) is one of the most common and detrimental neuropsychological disorders. Recently, more and more studies have focused on circular RNA as markers for central nervous system (CNS) diseases. The present study was conducted to evaluate the circular RNA expression alteration in serum exosomal and to identify a novel circulating biomarker for the detection of AD. We first isolated exosomes from serum and then investigated the circRNA expression alterations by high throughput whole transcriptome sequencing. The data were then analyzed using bioinformatics methods. Moreover, we verified the circRNA-seq by qRT-PCR. Furthermore, we analyzed the correlations between the levels of hsa_circ_0004771 and both Severity of Alcohol Dependence Questionnaire (SADQ) and Alcohol Dependence Scale (ADS). The diagnostic value of hsa_circ_0004771 in AD patients was evaluated by receiver operating characteristic (ROC). In this study, 254 differentially expressed circRNAs were identified, with 149 upregulated and 105 downregulated. GO analysis showed that these differentially expressed circRNAs from exosomes might be associated with the regulation of neuron projection and axon regeneration. KEGG analysis revealed that T cell receptor signaling and antigen processing and presentation pathway had a regulating effect on upstream levels. We found that hsa_circ_0004771 was related to the severity of AD. The AUC for the diagnostic value of hsa_circ_0004771 in AD patients was 0.874. These findings indicated that circRNA in serum exosomes provide novel targets for further research on molecular mechanisms of AD. Among these, hsa_circ_0004771 may be a sensitive biomarker that was related to the severity of AD.
Collapse
Affiliation(s)
- Yunyun Liu
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| | - Jiande Li
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| | - Huanhuan Bu
- Department of Neurology The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
| | - Hongxuan Wang
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| | - Yuanpei Zhang
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| | - Qingyu Shen
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| | - Mei Li
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| | - Zijing Lu
- Department of Plastic and Cosmetic Surgery Nanfang Hospital, Southern Medical University Guangzhou China
| | - Xiaoming Rong
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| | - Dong Zheng
- Department of Neurology The Affiliated Brain Hospital of Guangzhou Medical University Guangzhou China
| | - Ying Peng
- Department of Neurology Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation Sun Yat‐sen Memorial Hospital, Sun Yat‐sen University Guangzhou China
| |
Collapse
|
15
|
Zhang CN, Li FJ, Zhao ZL, Zhang JN. The role of extracellular vesicles in traumatic brain injury-induced acute lung injury. Am J Physiol Lung Cell Mol Physiol 2021; 321:L885-L891. [PMID: 34549593 DOI: 10.1152/ajplung.00023.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Acute lung injury (ALI), a common complication after traumatic brain injury (TBI), can evolve into acute respiratory distress syndrome (ARDS) and has a mortality rate of 30%-40%. Secondary ALI after TBI exhibits the following typical pathological features: infiltration of neutrophils into the alveolar and interstitial space, alveolar septal thickening, alveolar edema, and hemorrhage. Extracellular vesicles (EVs) were recently identified as key mediators in TBI-induced ALI. Due to their small size and lipid bilayer, they can pass through the disrupted blood-brain barrier (BBB) into the peripheral circulation and deliver their contents, such as genetic material and proteins, to target cells through processes such as fusion, receptor-mediated interactions, and uptake. Acting as messengers, EVs contribute to mediating brain-lung cross talk after TBI. In this review, we aim to summarize the mechanism of EVs in TBI-induced ALI, which may provide new ideas for clinical treatment.
Collapse
Affiliation(s)
- Chao-Nan Zhang
- Department of Neurosurgery, Tianjin Institute of Neurology, grid.412645.0Tianjin Medical University General Hospital, Tianjin, China
| | - Fan-Jian Li
- Department of Neurosurgery, Tianjin Institute of Neurology, grid.412645.0Tianjin Medical University General Hospital, Tianjin, China
| | - Zi-Long Zhao
- Department of Neurosurgery, Tianjin Institute of Neurology, grid.412645.0Tianjin Medical University General Hospital, Tianjin, China
| | - Jian-Ning Zhang
- Department of Neurosurgery, Tianjin Institute of Neurology, grid.412645.0Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
16
|
Zhang T, Ma S, Lv J, Wang X, Afewerky HK, Li H, Lu Y. The emerging role of exosomes in Alzheimer's disease. Ageing Res Rev 2021; 68:101321. [PMID: 33727157 DOI: 10.1016/j.arr.2021.101321] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/20/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD), manifested by memory loss and a decline in cognitive functions, is the most prevalent neurodegenerative disease accounting for 60-80 % of dementia cases. But, to-date, there is no effective treatment available to slow or stop the progression of AD. Exosomes are small extracellular vesicles that carry constituents, such as functional messenger RNAs, non-coding RNAs, proteins, lipids, DNA, and other bioactive substances of their source cells. In the brain, exosomes are likely to be sourced by almost all cell types and involve in cell communication to regulate cellular functions. The yet, accumulated evidence on the roles of exosomes and their constituents in the AD pathological process suggests their significance as additional biomarkers and therapeutic targets for AD. This review summarizes the current reported research findings on exosomes roles in the pathogenesis, diagnosis, and treatment of AD.
Collapse
|
17
|
Pelissier Vatter FA, Cioffi M, Hanna SJ, Castarede I, Caielli S, Pascual V, Matei I, Lyden D. Extracellular vesicle- and particle-mediated communication shapes innate and adaptive immune responses. J Exp Med 2021; 218:212439. [PMID: 34180950 PMCID: PMC8241538 DOI: 10.1084/jem.20202579] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/25/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
Intercellular communication among immune cells is vital for the coordination of proper immune responses. Extracellular vesicles and particles (EVPs) act as messengers in intercellular communication, with important consequences for target cell and organ physiology in both health and disease. Under normal physiological conditions, immune cell-derived EVPs participate in immune responses by regulating innate and adaptive immune responses. EVPs play a major role in antigen presentation and immune activation. On the other hand, immune cell-derived EVPs exert immunosuppressive and regulatory effects. Consequently, EVPs may contribute to pathological conditions, such as autoimmune and inflammatory diseases, graft rejection, and cancer progression and metastasis. Here, we provide an overview of the role of EVPs in immune homeostasis and pathophysiology, with a particular focus on their contribution to innate and adaptive immunity and their potential use for immunotherapies.
Collapse
Affiliation(s)
- Fanny A Pelissier Vatter
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY
| | - Michele Cioffi
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY
| | - Samer J Hanna
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY
| | - Ines Castarede
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY.,Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Simone Caielli
- Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Virginia Pascual
- Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Irina Matei
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY
| | - David Lyden
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY
| |
Collapse
|
18
|
Guo M, Hao Y, Feng Y, Li H, Mao Y, Dong Q, Cui M. Microglial Exosomes in Neurodegenerative Disease. Front Mol Neurosci 2021; 14:630808. [PMID: 34045943 PMCID: PMC8148341 DOI: 10.3389/fnmol.2021.630808] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 04/09/2021] [Indexed: 12/20/2022] Open
Abstract
Microglia play an important role in neurodegenerative disease [i.e., Parkinson's disease (PD), Alzheimer's disease (AD), and amyotrophic lateral sclerosis (ALS)]. These diseases share some similar pathological changes and several microglia-associated processes, including immune response, neuroinflammation, phagocytosis, elimination of synapses et al. Microglia in the central nervous system (CNS) has been described as having both destructive and protective effects in neurological disorders. Besides, considerable evidence also indicates that microglia play a significant role in neurogenesis, neuronal cell death, and synaptic interactions. The communication between microglia and neurons is of vital role in regulating complex functions which are key to appropriate the activity of the brain. Accumulating studies have also demonstrated that exosomes with sizes ranging from 40-100 nm, released by microglia, could serve as key mediators in intercellular signaling. These exosomes, identified in terms of cellular origin in many kinds of biological fluids, exert their effects by delivering specific cargos such as proteins, microRNAs (miRNAs), and mRNAs. It was shown that microglial exosomes could transport to and be uptake by neurons, which may either be beneficial or instead, detrimental to CNS diseases. The focus of this review is to summarize the involvement of microglial exosomes in critical pathologies associated with neurodegenerative disease and how they contribute to these disorders, including PD, AD, and ALS. We also review the application of microglia exosomes as potential biomarkers in monitoring disease progression, as well as focusing on their roles as drug delivery vehicles in treating neurodegenerative disorders.
Collapse
Affiliation(s)
- Min Guo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yining Hao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yiwei Feng
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Haiqing Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yiting Mao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Edler MK, Mhatre-Winters I, Richardson JR. Microglia in Aging and Alzheimer's Disease: A Comparative Species Review. Cells 2021; 10:1138. [PMID: 34066847 PMCID: PMC8150617 DOI: 10.3390/cells10051138] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia are the primary immune cells of the central nervous system that help nourish and support neurons, clear debris, and respond to foreign stimuli. Greatly impacted by their environment, microglia go through rapid changes in cell shape, gene expression, and functional behavior during states of infection, trauma, and neurodegeneration. Aging also has a profound effect on microglia, leading to chronic inflammation and an increase in the brain's susceptibility to neurodegenerative processes that occur in Alzheimer's disease. Despite the scientific community's growing knowledge in the field of neuroinflammation, the overall success rate of drug treatment for age-related and neurodegenerative diseases remains incredibly low. Potential reasons for the lack of translation from animal models to the clinic include the use of a single species model, an assumption of similarity in humans, and ignoring contradictory data or information from other species. To aid in the selection of validated and predictive animal models and to bridge the translational gap, this review evaluates similarities and differences among species in microglial activation and density, morphology and phenotype, cytokine expression, phagocytosis, and production of oxidative species in aging and Alzheimer's disease.
Collapse
Affiliation(s)
- Melissa K. Edler
- Department of Anthropology, School of Biomedical Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44240, USA;
| | - Isha Mhatre-Winters
- School of Biomedical Sciences, College of Arts and Sciences, Kent State University, Kent, OH 44240, USA;
- Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Jason R. Richardson
- Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
20
|
Tiberi M, Chiurchiù V. Specialized Pro-resolving Lipid Mediators and Glial Cells: Emerging Candidates for Brain Homeostasis and Repair. Front Cell Neurosci 2021; 15:673549. [PMID: 33981203 PMCID: PMC8107215 DOI: 10.3389/fncel.2021.673549] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
Astrocytes and oligodendrocytes are known to play critical roles in the central nervous system development, homeostasis and response to injury. In addition to their well-defined functions in synaptic signaling, blood-brain barrier control and myelination, it is now becoming clear that both glial cells also actively produce a wide range of immune-regulatory factors and engage in an intricate communication with neurons, microglia or with infiltrated immune cells, thus taking a center stage in both inflammation and resolution processes occurring within the brain. Resolution of inflammation is operated by the superfamily of specialized pro-resolving lipid mediators (SPMs), that include lipoxins, resolvins, protectins and maresins, and that altogether activate a series of cellular and molecular events that lead to spontaneous regression of inflammatory processes and restoration of tissue homeostasis. Here, we review the manifold effects of SPMs on modulation of astrocytes and oligodendrocytes, along with the mechanisms through which they either inhibit inflammatory pathways or induce the activation of protective ones. Furthermore, the possible role of SPMs in modulating the cross-talk between microglia, astrocytes and oligodendrocytes is also summarized. This SPM-mediated mechanism uncovers novel pathways of immune regulation in the brain that could be further exploited to control neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Marta Tiberi
- Laboratory of Resolution of Neuroinflammation, European Center for Brain Research, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Valerio Chiurchiù
- Laboratory of Resolution of Neuroinflammation, European Center for Brain Research, IRCCS Santa Lucia Foundation, Rome, Italy.,Institute of Translational Pharmacology, National Research Council, Rome, Italy
| |
Collapse
|
21
|
Lysosomal Functions in Glia Associated with Neurodegeneration. Biomolecules 2021; 11:biom11030400. [PMID: 33803137 PMCID: PMC7999372 DOI: 10.3390/biom11030400] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Lysosomes are cellular organelles that contain various acidic digestive enzymes. Despite their small size, they have multiple functions. Lysosomes remove or recycle unnecessary cell parts. They repair damaged cellular membranes by exocytosis. Lysosomes also sense cellular energy status and transmit signals to the nucleus. Glial cells are non-neuronal cells in the nervous system and have an active role in homeostatic support for neurons. In response to dynamic cues, glia use lysosomal pathways for the secretion and uptake of regulatory molecules, which affect the physiology of neighboring neurons. Therefore, functional aberration of glial lysosomes can trigger neuronal degeneration. Here, we review lysosomal functions in oligodendrocytes, astrocytes, and microglia, with emphasis on neurodegeneration.
Collapse
|
22
|
Izco M, Carlos E, Alvarez-Erviti L. The Two Faces of Exosomes in Parkinson's Disease: From Pathology to Therapy. Neuroscientist 2021; 28:180-193. [PMID: 33530851 DOI: 10.1177/1073858421990001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Accumulating evidence suggests that exosomes play a key role in Parkinson's disease (PD). Exosomes may contribute to the PD progression facilitating the spread of pathological alpha-synuclein or activating immune cells. Glial cells also release exosomes, and transmission of exosomes derived from activated glial cells containing inflammatory mediators may contribute to the propagation of the neuroinflammatory response. Glia-to-neuron transmission of exosomes containing alpha-synuclein may contribute to alpha-synuclein propagation and neurodegeneration. Additionally, miRNAs can be transmitted among cells via exosomes inducing changes in the genetic program of the target cell contributing to PD progression. Exosomes also represent a promising drug delivery system. The brain is a difficult target for drugs of all classes because the blood-brain barrier excludes most macromolecular drugs. One of the major challenges is the development of vehicles for robust delivery to the brain. Targeted exosomes may have the potential for delivering therapeutic agents, including proteins and gene therapy molecules, into the brain. This review summarizes recent advances in the role of exosomes in PD pathology progression and their potential use as drug delivery system for PD treatment, the two faces of the exosomes in PD.
Collapse
Affiliation(s)
- Maria Izco
- Laboratory of Molecular Neurobiology, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Estefania Carlos
- Laboratory of Molecular Neurobiology, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Lydia Alvarez-Erviti
- Laboratory of Molecular Neurobiology, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| |
Collapse
|
23
|
Gualerzi A, Lombardi M, Verderio C. Microglia-oligodendrocyte intercellular communication: role of extracellular vesicle lipids in functional signalling. Neural Regen Res 2021; 16:1194-1195. [PMID: 33269772 PMCID: PMC8224135 DOI: 10.4103/1673-5374.300430] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|
24
|
Alcohol Increases Exosome Release from Microglia to Promote Complement C1q-Induced Cellular Death of Proopiomelanocortin Neurons in the Hypothalamus in a Rat Model of Fetal Alcohol Spectrum Disorders. J Neurosci 2020; 40:7965-7979. [PMID: 32887744 DOI: 10.1523/jneurosci.0284-20.2020] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/23/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023] Open
Abstract
Microglia, a type of CNS immune cell, have been shown to contribute to ethanol-activated neuronal death of the stress regulatory proopiomelanocortin (POMC) neuron-producing β-endorphin peptides in the hypothalamus in a postnatal rat model of fetal alcohol spectrum disorders. We determined whether the microglial extracellular vesicle exosome is involved in the ethanol-induced neuronal death of the β-endorphin neuron. Extracellular vesicles were prepared from hypothalamic tissues collected from postnatal rats (both males and females) fed daily with 2.5 mg/kg ethanol or control milk formula for 5 d or from hypothalamic microglia cells obtained from postnatal rats, grown in cultures for several days, and then challenged with ethanol or vehicle for 24 h. Nanoparticle tracking analysis and transmission electron microscopy indicated that these vesicles had the size range and shape of exosomes. Ethanol treatments increased the number and the β-endorphin neuronal killing activity of microglial exosomes both in vivo and in vitro Proteomics analyses of exosomes of cultured microglial cells identified a large number of proteins, including various complements, which were elevated following ethanol treatment. Proteomics data involving complements were reconfirmed using quantitative protein assays. Ethanol treatments also increased deposition of the complement protein C1q in β-endorphin neuronal cells in both in vitro and in vivo systems. Recombinant C1q protein increased while C1q blockers reduced ethanol-induced C3a/b, C4, and membrane attack complex/C5b9 formations; ROS production; and ultimately cellular death of β-endorphin neurons. These data suggest that the complement system involving C1q-C3-C4-membrane attack complex and ROS regulates exosome-mediated, ethanol-induced β-endorphin neuronal death.SIGNIFICANCE STATEMENT Neurotoxic action of alcohol during the developmental period is recognized for its involvement in fetal alcohol spectrum disorders, but the lack of clear understanding of the mechanism of alcohol action has delayed the progress in therapeutic intervention of this disease. Proopiomelanocortin neurons known to regulate stress, energy homeostasis, and immune functions are reported to be killed by developmental alcohol exposure because of activation of microglial immune cells in the brain. While microglia are known to use extracellular vesicles to communicate with neurons for maintaining homeostasis, we show here that ethanol exposure during the developmental period hijacks this system to spread apoptotic factors, including complement protein C1q, to induce the membrane attack complex and reactive super-oxygen species for proopiomelanocortin neuronal killing.
Collapse
|
25
|
P2X7 Receptors Amplify CNS Damage in Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21175996. [PMID: 32825423 PMCID: PMC7504621 DOI: 10.3390/ijms21175996] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
ATP is a (co)transmitter and signaling molecule in the CNS. It acts at a multitude of ligand-gated cationic channels termed P2X to induce rapid depolarization of the cell membrane. Within this receptor-channel family, the P2X7 receptor (R) allows the transmembrane fluxes of Na+, Ca2+, and K+, but also allows the slow permeation of larger organic molecules. This is supposed to cause necrosis by excessive Ca2+ influx, as well as depletion of intracellular ions and metabolites. Cell death may also occur by apoptosis due to the activation of the caspase enzymatic cascade. Because P2X7Rs are localized in the CNS preferentially on microglia, but also at a lower density on neuroglia (astrocytes, oligodendrocytes) the stimulation of this receptor leads to the release of neurodegeneration-inducing bioactive molecules such as pro-inflammatory cytokines, chemokines, proteases, reactive oxygen and nitrogen molecules, and the excitotoxic glutamate/ATP. Various neurodegenerative reactions of the brain/spinal cord following acute harmful events (mechanical CNS damage, ischemia, status epilepticus) or chronic neurodegenerative diseases (neuropathic pain, Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, amyotrophic lateral sclerosis) lead to a massive release of ATP via the leaky plasma membrane of neural tissue. This causes cellular damage superimposed on the original consequences of neurodegeneration. Hence, blood-brain-barrier permeable pharmacological antagonists of P2X7Rs with excellent bioavailability are possible therapeutic agents for these diseases. The aim of this review article is to summarize our present state of knowledge on the involvement of P2X7R-mediated events in neurodegenerative illnesses endangering especially the life quality and duration of the aged human population.
Collapse
|
26
|
Aires ID, Ribeiro-Rodrigues T, Boia R, Catarino S, Girão H, Ambrósio AF, Santiago AR. Exosomes derived from microglia exposed to elevated pressure amplify the neuroinflammatory response in retinal cells. Glia 2020; 68:2705-2724. [PMID: 32645245 DOI: 10.1002/glia.23880] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022]
Abstract
Glaucoma is a degenerative disease that causes irreversible loss of vision and is characterized by retinal ganglion cell (RGC) loss. Others and we have demonstrated that chronic neuroinflammation mediated by reactive microglial cells plays a role in glaucomatous pathology. Exosomes are extracellular vesicles released by most cells, including microglia, that mediate intercellular communication. The role of microglial exosomes in glaucomatous degeneration remains unknown. Taking the prominent role of microglial exosomes in brain neurodegenerative diseases, we studied the contribution of microglial-derived exosomes to the inflammatory response in experimental glaucoma. Microglial cells were exposed to elevated hydrostatic pressure (EHP), to mimic elevated intraocular pressure, the main risk factor for glaucoma. Naïve microglia (BV-2 cells or retinal microglia) were exposed to exosomes derived from BV-2 cells under EHP conditions (BV-Exo-EHP) or cultured in control pressure (BV-Exo-Control). We found that BV-Exo-EHP increased the production of pro-inflammatory cytokines, promoted retinal microglia motility, phagocytic efficiency, and proliferation. Furthermore, the incubation of primary retinal neural cell cultures with BV-Exo-EHP increased cell death and the production of reactive oxygen species. Exosomes derived from retinal microglia (MG-Exo-Control or MG-Exo-EHP) were injected in the vitreous of C57BL/6J mice. MG-Exo-EHP sustained activation of retinal microglia, mediated cell death, and impacted RGC number. Herein, we show that exosomes derived from retinal microglia have an autocrine function and propagate the inflammatory signal in conditions of elevated pressure, contributing to retinal degeneration in glaucomatous conditions.
Collapse
Affiliation(s)
- Inês Dinis Aires
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Teresa Ribeiro-Rodrigues
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Raquel Boia
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Steve Catarino
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Henrique Girão
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - António Francisco Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.,Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal
| | - Ana Raquel Santiago
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.,Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal
| |
Collapse
|
27
|
Regulation of Microglial Functions by Purinergic Mechanisms in the Healthy and Diseased CNS. Cells 2020; 9:cells9051108. [PMID: 32365642 PMCID: PMC7290360 DOI: 10.3390/cells9051108] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Microglial cells, the resident macrophages of the central nervous system (CNS), exist in a process-bearing, ramified/surveying phenotype under resting conditions. Upon activation by cell-damaging factors, they get transformed into an amoeboid phenotype releasing various cell products including pro-inflammatory cytokines, chemokines, proteases, reactive oxygen/nitrogen species, and the excytotoxic ATP and glutamate. In addition, they engulf pathogenic bacteria or cell debris and phagocytose them. However, already resting/surveying microglia have a number of important physiological functions in the CNS; for example, they shield small disruptions of the blood–brain barrier by their processes, dynamically interact with synaptic structures, and clear surplus synapses during development. In neurodegenerative illnesses, they aggravate the original disease by a microglia-based compulsory neuroinflammatory reaction. Therefore, the blockade of this reaction improves the outcome of Alzheimer’s Disease, Parkinson’s Disease, multiple sclerosis, amyotrophic lateral sclerosis, etc. The function of microglia is regulated by a whole array of purinergic receptors classified as P2Y12, P2Y6, P2Y4, P2X4, P2X7, A2A, and A3, as targets of endogenous ATP, ADP, or adenosine. ATP is sequentially degraded by the ecto-nucleotidases and 5′-nucleotidase enzymes to the almost inactive inosine as an end product. The appropriate selective agonists/antagonists for purinergic receptors as well as the respective enzyme inhibitors may profoundly interfere with microglial functions and reconstitute the homeostasis of the CNS disturbed by neuroinflammation.
Collapse
|
28
|
The Messenger Apps of the cell: Extracellular Vesicles as Regulatory Messengers of Microglial Function in the CNS. J Neuroimmune Pharmacol 2020; 15:473-486. [PMID: 32337651 DOI: 10.1007/s11481-020-09916-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 03/20/2020] [Indexed: 02/08/2023]
Abstract
The intense effort of investigators, in particular during the past decade, has highlighted the importance of extracellular vesicles (EVs) such as exosomes in regulating both innate and adaptive immunity in the course of a variety of infections, with clear implications for development of novel vaccines, therapeutics, and diagnostics. Current and future efforts now need to focus strongly on teasing apart the intricate and complex molecular mechanisms that operate during EV regulation of immunity. In this review, we discuss recent advances that bear on our current understanding of how EVs, including exosomes, can contribute to the innate immune functions of microglia within the central nervous system (CNS), and we also highlight future important mechanistic questions that need to be addressed. In particular, recent findings that highlight the crosstalk between autophagy and exosome pathways and their implications for innate immune functions of microglia will be presented. Microglial activation has been shown to play a key role in neuroAIDS, a neuro-infectious disease for which the importance of exosome functions, including exosome-autophagy interplay, has been reported. The importance of exosomes and exosome-autophagy crosstalk involving microglia has also been shown for the Parkinson's disease (PD), a neurodegenerative disease that is thought to be linked with immune dysfunction and involve infectious agents as trigger. Considering the accumulation of recent findings and the vibrancy of the EV field, we anticipate that future studies will continue to have a deep impact on our understanding of the CNS pathologies that are influenced by the functions of microglia and of the infectious disease mechanisms in general. Graphical Abstract.
Collapse
|
29
|
Detrimental and protective action of microglial extracellular vesicles on myelin lesions: astrocyte involvement in remyelination failure. Acta Neuropathol 2019; 138:987-1012. [PMID: 31363836 PMCID: PMC6851224 DOI: 10.1007/s00401-019-02049-1] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 12/17/2022]
Abstract
Microglia are highly plastic immune cells which exist in a continuum of activation states. By shaping the function of oligodendrocyte precursor cells (OPCs), the brain cells which differentiate to myelin-forming cells, microglia participate in both myelin injury and remyelination during multiple sclerosis. However, the mode(s) of action of microglia in supporting or inhibiting myelin repair is still largely unclear. Here, we analysed the effects of extracellular vesicles (EVs) produced in vitro by either pro-inflammatory or pro-regenerative microglia on OPCs at demyelinated lesions caused by lysolecithin injection in the mouse corpus callosum. Immunolabelling for myelin proteins and electron microscopy showed that EVs released by pro-inflammatory microglia blocked remyelination, whereas EVs produced by microglia co-cultured with immunosuppressive mesenchymal stem cells promoted OPC recruitment and myelin repair. The molecular mechanisms responsible for the harmful and beneficial EV actions were dissected in primary OPC cultures. By exposing OPCs, cultured either alone or with astrocytes, to inflammatory EVs, we observed a blockade of OPC maturation only in the presence of astrocytes, implicating these cells in remyelination failure. Biochemical fractionation revealed that astrocytes may be converted into harmful cells by the inflammatory EV cargo, as indicated by immunohistochemical and qPCR analyses, whereas surface lipid components of EVs promote OPC migration and/or differentiation, linking EV lipids to myelin repair. Although the mechanisms through which the lipid species enhance OPC maturation still remain to be fully defined, we provide the first demonstration that vesicular sphingosine 1 phosphate stimulates OPC migration, the first fundamental step in myelin repair. From this study, microglial EVs emerge as multimodal and multitarget signalling mediators able to influence both OPCs and astrocytes around myelin lesions, which may be exploited to develop novel approaches for myelin repair not only in multiple sclerosis, but also in neurological and neuropsychiatric diseases characterized by demyelination.
Collapse
|
30
|
Arab T, Raffo-Romero A, Van Camp C, Lemaire Q, Le Marrec-Croq F, Drago F, Aboulouard S, Slomianny C, Lacoste AS, Guigon I, Touzet H, Salzet M, Fournier I, Lefebvre C, Vizioli J, Sautière PE. Proteomic characterisation of leech microglia extracellular vesicles (EVs): comparison between differential ultracentrifugation and Optiprep™ density gradient isolation. J Extracell Vesicles 2019; 8:1603048. [PMID: 31069026 PMCID: PMC6493217 DOI: 10.1080/20013078.2019.1603048] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 01/04/2023] Open
Abstract
In Mammals, microglial cells are considered as the resident immune cells in central nervous system (CNS). Many studies demonstrated that, after injury, these cells are activated and recruited at the lesion site. Leech microglia present a similar pattern of microglial activation and migration upon experimental lesion of CNS. This activation is associated with the release of a large amount of extracellular vesicles (EVs). We collected EVs released by microglia primary culture and compared two different protocols of isolation: one with differential ultracentrifugation (UC) and one using an additional Optiprep™ Density Gradient (ODG) ultracentrifugation. Nanoparticles tracking analysis (NTA) and transmission electron microscopy (TEM) were used to assess vesicles size and morphology. The protein content of isolated EVs was assessed by mass spectrometry approaches. Results showed the presence of EV-specific proteins in both procedures. The extensive proteomic analysis of each single ODG fractions confirmed the efficiency of this protocol in limiting the presence of co-isolated proteins aggregates and other membranous particles during vesicles isolation. The present study permitted for the first time the characterisation of microglial EV protein content in an annelid model. Interestingly, an important amount of proteins found in leech vesicles was previously described in EV-specific databases. Finally, purified EVs were assessed for neurotrophic activity and promote neurites outgrowth on primary cultured neurons.
Collapse
Affiliation(s)
- T Arab
- U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse – PRISM, Univ. Lille, Inserm, Lille, France
| | - A Raffo-Romero
- U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse – PRISM, Univ. Lille, Inserm, Lille, France
| | - C Van Camp
- U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse – PRISM, Univ. Lille, Inserm, Lille, France
| | - Q Lemaire
- U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse – PRISM, Univ. Lille, Inserm, Lille, France
| | - F Le Marrec-Croq
- U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse – PRISM, Univ. Lille, Inserm, Lille, France
| | - F Drago
- U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse – PRISM, Univ. Lille, Inserm, Lille, France
| | - S Aboulouard
- U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse – PRISM, Univ. Lille, Inserm, Lille, France
| | - C Slomianny
- Inserm U1003, PHYCEL Laboratory, Physiologie Cellulaire, Univ Lille, Lille, France
- Bio Imaging Center Lille, Univ. Lille, Lille, France
| | - A-S Lacoste
- Bio Imaging Center Lille, Univ. Lille, Lille, France
| | - I Guigon
- CNRS, Centrale Lille, UMR 9189 - CRIStAL - Centre de Recherche en Informatique Signal et Automatique de Lille, Bilille and Univ. Lille, Lille, France
| | - H Touzet
- CNRS, Centrale Lille, Inria, UMR 9189 - CRIStAL - Centre de Recherche en Informatique Signal et Automatique de Lille, Univ. Lille, Lille, France
| | - M Salzet
- U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse – PRISM, Univ. Lille, Inserm, Lille, France
| | - I Fournier
- U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse – PRISM, Univ. Lille, Inserm, Lille, France
| | - C Lefebvre
- U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse – PRISM, Univ. Lille, Inserm, Lille, France
| | - J Vizioli
- U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse – PRISM, Univ. Lille, Inserm, Lille, France
| | - P-E Sautière
- U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse – PRISM, Univ. Lille, Inserm, Lille, France
| |
Collapse
|
31
|
Delpech JC, Herron S, Botros MB, Ikezu T. Neuroimmune Crosstalk through Extracellular Vesicles in Health and Disease. Trends Neurosci 2019; 42:361-372. [PMID: 30926143 DOI: 10.1016/j.tins.2019.02.007] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 02/21/2019] [Accepted: 02/28/2019] [Indexed: 12/11/2022]
Abstract
The dynamics of CNS function rely upon omnidirectional communication among CNS cell types. Extracellular vesicles (EVs) have emerged as key mediators of this communication and are actively involved in response to CNS injury, mediating inflammatory response and inflammation-related neuroprotection as they display dual beneficial and detrimental roles. Neuroimmune interactions include communication between neurons and microglia, the resident macrophages within the CNS, and these interactions are a critical mediator of healthy brain functions, mounting an inflammatory response, and disease pathogenesis. This review aims to organize recent research highlighting the role of EVs in health and neurodegenerative disorders, with a specific focus on neuroimmune interactions between neurons and glia in Alzheimer's disease.
Collapse
Affiliation(s)
- Jean-Christophe Delpech
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Shawn Herron
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Mina B Botros
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Tsuneya Ikezu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA; Neurology, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
32
|
Antibody-mediated clearance of tau in primary mouse microglial cultures requires Fcγ-receptor binding and functional lysosomes. Sci Rep 2019; 9:4658. [PMID: 30874605 PMCID: PMC6420568 DOI: 10.1038/s41598-019-41105-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 03/01/2019] [Indexed: 12/16/2022] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease are characterized by the progressive spreading and accumulation of hyper-phosphorylated tau protein in the brain. Anti-tau antibodies have been shown to reduce tau pathology in in vivo models and antibody-mediated clearance of tau exerted by microglia has been proposed as a contributing factor. By subjecting primary microglia cultured in vitro to anti-phospho-tau antibodies in complex with pathological tau, we show that microglia internalise and degrade tau in a manner that is dependent on FcγR interaction and functional lysosomes. It has recently been discussed if anti-tau antibody effector-functions are required for induction of tau clearance. Using antibodies with compromised FcγR binding and non-compromised control antibodies we show that antibody effector functions are required for induction of microglial clearance of tau. Understanding the inflammatory consequences of targeting microglia using therapeutic antibodies is important when developing these molecules for clinical use. Using RNA sequencing, we show that treatment with anti-tau antibodies increases transcription of mRNA encoding pro-inflammatory markers, but that the mRNA expression profile of antibody-treated cells differ from the profile of LPS activated microglia. We further demonstrate that microglia activation alone is not sufficient to induce significant tau clearance.
Collapse
|
33
|
Malloci M, Perdomo L, Veerasamy M, Andriantsitohaina R, Simard G, Martínez MC. Extracellular Vesicles: Mechanisms in Human Health and Disease. Antioxid Redox Signal 2019; 30:813-856. [PMID: 29634347 DOI: 10.1089/ars.2017.7265] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Secreted extracellular vesicles (EVs) are now considered veritable entities for diagnosis, prognosis, and therapeutics. These structures are able to interact with target cells and modify their phenotype and function. Recent Advances: Since composition of EVs depends on the cell type of origin and the stimulation that leads to their release, the analysis of EV content remains an important input to understand the potential effects of EVs on target cells. CRITICAL ISSUES Here, we review recent data related to the mechanisms involved in the formation of EVs and the methods allowing specific EV isolation and identification. Also, we analyze the potential use of EVs as biomarkers in different pathologies such as diabetes, obesity, atherosclerosis, neurodegenerative diseases, and cancer. Besides, their role in these diseases is discussed. Finally, we consider EVs enriched in microRNA or drugs as potential therapeutic cargo able to deliver desirable information to target cells/tissues. FUTURE DIRECTIONS We underline the importance of the homogenization of the parameters of isolation of EVs and their characterization, which allow considering EVs as excellent biomarkers for diagnosis and prognosis.
Collapse
Affiliation(s)
- Marine Malloci
- 1 INSERM UMR 1063, Stress Oxydant et Pathologies Métaboliques, UNIV Angers, Université Bretagne Loire, Angers, France
| | - Liliana Perdomo
- 1 INSERM UMR 1063, Stress Oxydant et Pathologies Métaboliques, UNIV Angers, Université Bretagne Loire, Angers, France
| | - Maëva Veerasamy
- 1 INSERM UMR 1063, Stress Oxydant et Pathologies Métaboliques, UNIV Angers, Université Bretagne Loire, Angers, France
| | - Ramaroson Andriantsitohaina
- 1 INSERM UMR 1063, Stress Oxydant et Pathologies Métaboliques, UNIV Angers, Université Bretagne Loire, Angers, France.,2 Centre Hospitalo-Universitaire d'Angers, Angers, France
| | - Gilles Simard
- 1 INSERM UMR 1063, Stress Oxydant et Pathologies Métaboliques, UNIV Angers, Université Bretagne Loire, Angers, France.,2 Centre Hospitalo-Universitaire d'Angers, Angers, France
| | - M Carmen Martínez
- 1 INSERM UMR 1063, Stress Oxydant et Pathologies Métaboliques, UNIV Angers, Université Bretagne Loire, Angers, France.,2 Centre Hospitalo-Universitaire d'Angers, Angers, France
| |
Collapse
|
34
|
Stampanoni Bassi M, Gentile A, Iezzi E, Zagaglia S, Musella A, Simonelli I, Gilio L, Furlan R, Finardi A, Marfia GA, Guadalupi L, Bullitta S, Mandolesi G, Centonze D, Buttari F. Transient Receptor Potential Vanilloid 1 Modulates Central Inflammation in Multiple Sclerosis. Front Neurol 2019; 10:30. [PMID: 30761069 PMCID: PMC6361812 DOI: 10.3389/fneur.2019.00030] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 01/10/2019] [Indexed: 12/04/2022] Open
Abstract
Introduction: Disease course of multiple sclerosis (MS) is negatively influenced by proinflammatory molecules released by activated T and B lymphocytes and local immune cells. The endovanilloid system plays different physiological functions, and preclinical data suggest that transient receptor potential vanilloid type 1 (TRPV1) could modulate neuroinflammation in this disorder. Methods: The effect of TRPV1 activation on the release of two main proinflammatory cytokines, tumor necrosis factor (TNF) and interleukin (IL)-6, was explored in activated microglial cells. Furthermore, in a group of 132 MS patients, the association between the cerebrospinal fluid (CSF) levels of TNF and IL-6 and a single nucleotide polymorphisms (SNP) influencing TRPV1 protein expression and function (rs222747) was assessed. Results: In in vitro experiments, TRPV1 stimulation by capsaicin significantly reduced TNF and IL-6 release by activated microglial cells. Moreover, the anti-inflammatory effect of TRPV1 activation was confirmed by another TRPV1 agonist, the resiniferatoxin (RTX), whose effects were significantly inhibited by the TRPV1 antagonist, 5-iodoresiniferatoxin (5-IRTX). Vice versa, BV2 pre-treatment with 5-IRTX increased the inflammatory response induced by LPS. Moreover, in MS patients, a significant association emerged between TRPV1 SNP rs222747 and CSF TNF levels. In particular, the presence of a G allele, known to result in increased TRPV1 protein expression and function, was associated to lower CSF levels of TNF. Conclusions: Our results indicate that TRPV1 influences central inflammation in MS by regulating cytokine release by activated microglial cells. The modulation of the endovanilloid system may represent a useful approach to contrast neuroinflammation in MS.
Collapse
Affiliation(s)
| | - Antonietta Gentile
- Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Ennio Iezzi
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| | - Sara Zagaglia
- Clinica Neurologica, Università Politecnica delle Marche, Ancona, Italy
| | - Alessandra Musella
- Laboratory of Neuroimmunology and Synaptic Plasticity, University & IRCCS San Raffaele, Rome, Italy
| | - Ilaria Simonelli
- Servizio di Statistica Medica & Information Technology, Fondazione Fatebenefratelli per la Ricerca e la Formazione Sanitaria e Sociale, Rome, Italy
| | - Luana Gilio
- Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Roberto Furlan
- Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | - Annamaria Finardi
- Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | - Girolama A Marfia
- Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Livia Guadalupi
- Laboratory of Neuroimmunology and Synaptic Plasticity, University & IRCCS San Raffaele, Rome, Italy
| | - Silvia Bullitta
- Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Georgia Mandolesi
- Laboratory of Neuroimmunology and Synaptic Plasticity, University & IRCCS San Raffaele, Rome, Italy
| | - Diego Centonze
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy.,Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Fabio Buttari
- Unit of Neurology and Neurorehabilitation, IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
35
|
Szepesi Z, Manouchehrian O, Bachiller S, Deierborg T. Bidirectional Microglia-Neuron Communication in Health and Disease. Front Cell Neurosci 2018; 12:323. [PMID: 30319362 PMCID: PMC6170615 DOI: 10.3389/fncel.2018.00323] [Citation(s) in RCA: 296] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022] Open
Abstract
Microglia are ramified cells that exhibit highly motile processes, which continuously survey the brain parenchyma and react to any insult to the CNS homeostasis. Although microglia have long been recognized as a crucial player in generating and maintaining inflammatory responses in the CNS, now it has become clear, that their function are much more diverse, particularly in the healthy brain. The innate immune response and phagocytosis represent only a little segment of microglia functional repertoire that also includes maintenance of biochemical homeostasis, neuronal circuit maturation during development and experience-dependent remodeling of neuronal circuits in the adult brain. Being equipped by numerous receptors and cell surface molecules microglia can perform bidirectional interactions with other cell types in the CNS. There is accumulating evidence showing that neurons inform microglia about their status and thus are capable of controlling microglial activation and motility while microglia also modulate neuronal activities. This review addresses the topic: how microglia communicate with other cell types in the brain, including fractalkine signaling, secreted soluble factors and extracellular vesicles. We summarize the current state of knowledge of physiological role and function of microglia during brain development and in the mature brain and further highlight microglial contribution to brain pathologies such as Alzheimer’s and Parkinson’s disease, brain ischemia, traumatic brain injury, brain tumor as well as neuropsychiatric diseases (depression, bipolar disorder, and schizophrenia).
Collapse
Affiliation(s)
- Zsuzsanna Szepesi
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Oscar Manouchehrian
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Sara Bachiller
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
36
|
Cypryk W, Nyman TA, Matikainen S. From Inflammasome to Exosome-Does Extracellular Vesicle Secretion Constitute an Inflammasome-Dependent Immune Response? Front Immunol 2018; 9:2188. [PMID: 30319640 PMCID: PMC6167409 DOI: 10.3389/fimmu.2018.02188] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/04/2018] [Indexed: 12/11/2022] Open
Abstract
Inflammasomes are intracellular protein complexes of pattern recognition receptors and caspase-1, with essential functions in regulating inflammatory responses of macrophages and dendritic cells. The primary role of inflammasomes is to catalyze processing and secretion of pro-inflammatory cytokines IL-1β and IL-18. Recently, intracellular non-canonical inflammasome activation by caspases-4/5, which are also regulators of pyroptosis via processing gasdermin D, has been elucidated. Caspase-1, the effector protease of inflammasome complex, is also known to modulate secretion of large number of other proteins. Thereby, besides its known role in processing pro-inflammatory cytokines, the inflammasome turns into a universal regulator of protein secretion, which allows the danger-exposed cells to release various proteins in order to alert and guide neighboring cells. Majority of these proteins are not secreted through the conventional ER-Golgi secretory pathway. Instead, they are segregated in membrane-enclosed compartment and secreted in nanosized extracellular vesicles, which protect their cargo and guide it for delivery. Growing evidence indicates that inflammasome activity correlates with enhanced secretion of extracellular vesicles and modulation of their protein cargo. This inflammasome-driven unconventional, vesicle-mediated secretion of multitude of immunoregulatory proteins may constitute a novel paradigm in inflammatory responses. In this mini review we discuss the current knowledge and highlight unsolved questions about metabolic processes, signals, and mechanisms linking inflammasome activity with regulated extracellular vesicle secretion of proteins. Further investigations on this relationship may in the future help understanding the significance of extracellular vesicle secretion in inflammatory diseases such as atherosclerosis, gouty arthritis, asthma, Alzheimer's and many others.
Collapse
Affiliation(s)
- Wojciech Cypryk
- Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Lodz, Poland
| | - Tuula A Nyman
- Department of Immunology, Institute of Clinical Medicine, University of Oslo and Rikshospitalet Oslo, Oslo, Norway
| | - Sampsa Matikainen
- Division of Rheumatology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
37
|
Arroba AI, Campos-Caro A, Aguilar-Diosdado M, Valverde ÁM. IGF-1, Inflammation and Retinal Degeneration: A Close Network. Front Aging Neurosci 2018; 10:203. [PMID: 30026694 PMCID: PMC6041402 DOI: 10.3389/fnagi.2018.00203] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/14/2018] [Indexed: 01/10/2023] Open
Abstract
Retinal degenerative diseases are a group of heterogeneous diseases that include age-related macular degeneration (AMD), retinitis pigmentosa (RP), and diabetic retinopathy (DR). The progressive degeneration of the retinal neurons results in a severe deterioration of the visual function. Neuroinflammation is an early hallmark of many neurodegenerative disorders of the retina including AMD, RP and DR. Microglial cells, key components of the retinal immune defense system, are activated in retinal degenerative diseases. In the microglia the interplay between the proinflammatory/classically activated or antiinflammatory/alternatively activated phenotypes is a complex dynamic process that occurs during the course of disease due to the different environmental signals related to pathophysiological conditions. In this regard, an adequate transition from the proinflammatory to the anti-inflammatory response is necessary to counteract retinal neurodegeneration and its subsequent damage that leads to the loss of visual function. Insulin like-growth factor-1 (IGF-1) has been considered as a pleiotropic factor in the retina under health or disease conditions and several effects of IGF-1 in retinal immune modulation have been described. In this review, we provide recent insights of inflammation as a common feature of retinal diseases (AMD, RP and RD) highlighting the role of microglia, exosomes and IGF-1 in this process.
Collapse
Affiliation(s)
- Ana I Arroba
- Alberto Sols Biomedical Research Institute (IIBm) (CSIC/UAM), Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERdem), ISCIII, Madrid, Spain.,Research Unit, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), University Hospital "Puerta del Mar", Cádiz, Spain
| | - Antonio Campos-Caro
- Research Unit, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), University Hospital "Puerta del Mar", Cádiz, Spain
| | - Manuel Aguilar-Diosdado
- Research Unit, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), University Hospital "Puerta del Mar", Cádiz, Spain.,Department of Endocrinology and Metabolism, Instituto de Investigación e Innovación en Ciencias Biomédicas de la Provincia de Cádiz (INiBICA), University Hospital "Puerta del Mar", Cádiz, Spain
| | - Ángela M Valverde
- Alberto Sols Biomedical Research Institute (IIBm) (CSIC/UAM), Madrid, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERdem), ISCIII, Madrid, Spain
| |
Collapse
|
38
|
Paolicelli RC, Bergamini G, Rajendran L. Cell-to-cell Communication by Extracellular Vesicles: Focus on Microglia. Neuroscience 2018; 405:148-157. [PMID: 29660443 DOI: 10.1016/j.neuroscience.2018.04.003] [Citation(s) in RCA: 239] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/30/2018] [Accepted: 04/04/2018] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles, including exosomes and microvesicles, are small, nano-to-micrometer vesicles that are released from cells. While initially observed in immune cells and reticulocytes as vesicles meant to remove archaic proteins, now they have been observed in almost all cell types of multicellular organisms. Growing evidence indicates that extracellular vesicles, containing lipids, proteins and RNAs, represent an efficient way to transfer functional cargoes from one cell to another. In the central nervous system, the extensive cross-talk ongoing between neurons and glia, including microglia, the immune cells of the brain, takes advantage of secreted vesicles, which mediate intercellular communication over long range distance. Recent literature supports a critical role for extracellular vesicles in mediating complex and coordinated communication among neurons, astrocytes and microglia, both in the healthy and in the diseased brain. In this review, we focus on the biogenesis and function of microglia-related extracellular vesicles and focus on their putative role in Alzheimer's disease pathology.
Collapse
Affiliation(s)
- Rosa C Paolicelli
- Systems and Cell Biology of Neurodegeneration, IREM - Institute for Regenerative Medicine, University of Zurich, Switzerland.
| | - Giorgio Bergamini
- Preclinical Laboratory for Translational Research into Affective Disorders (PLaTRAD), Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zurich, Switzerland
| | - Lawrence Rajendran
- Systems and Cell Biology of Neurodegeneration, IREM - Institute for Regenerative Medicine, University of Zurich, Switzerland
| |
Collapse
|
39
|
Plastira I, Bernhart E, Goeritzer M, DeVaney T, Reicher H, Hammer A, Lohberger B, Wintersperger A, Zucol B, Graier WF, Kratky D, Malle E, Sattler W. Lysophosphatidic acid via LPA-receptor 5/protein kinase D-dependent pathways induces a motile and pro-inflammatory microglial phenotype. J Neuroinflammation 2017; 14:253. [PMID: 29258556 PMCID: PMC5735906 DOI: 10.1186/s12974-017-1024-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/06/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Extracellular lysophosphatidic acid (LPA) species transmit signals via six different G protein-coupled receptors (LPAR1-6) and are indispensible for brain development and function of the nervous system. However, under neuroinflammatory conditions or brain damage, LPA levels increase, thereby inducing signaling cascades that counteract brain function. We describe a critical role for 1-oleyl-2-hydroxy-sn-glycero-3-phosphate (termed "LPA" throughout our study) in mediating a motile and pro-inflammatory microglial phenotype via LPAR5 that couples to protein kinase D (PKD)-mediated pathways. METHODS Using the xCELLigence system and time-lapse microscopy, we investigated the migrational response of microglial cells. Different M1 and M2 markers were analyzed by confocal microscopy, flow cytometry, and immunoblotting. Using qPCR and ELISA, we studied the expression of migratory genes and quantitated the secretion of pro-inflammatory cytokines and chemokines, respectively. Different transcription factors that promote the regulation of pro-inflammatory genes were analyzed by western blot. Reactive oxygen species (ROS) and nitric oxide (NO) production, phagocytosis, and microglial cytotoxicity were determined using commercially available assay kits. RESULTS LPA induces MAPK family and AKT activation and pro-inflammatory transcription factors' phosphorylation (NF-κB, c-Jun, STAT1, and STAT3) that were inhibited by both LPAR5 and PKD family antagonists. LPA increases migratory capacity, induces secretion of pro-inflammatory cytokines and chemokines and expression of M1 markers, enhances production of ROS and NO by microglia, and augments cytotoxicity of microglial cell-conditioned medium towards neurons. The PKD family inhibitor blunted all of these effects. We propose that interference with this signaling axis could aid in the development of new therapeutic approaches to control neuroinflammation under conditions of overshooting LPA production. CONCLUSIONS In the present study, we show that inflammatory LPA levels increased the migratory response of microglia and promoted a pro-inflammatory phenotype via the LPAR5/PKD axis. Interference with this signaling axis reduced microglial migration, blunted microglial cytotoxicity, and abrogated the expression and secretion of pro-inflammatory mediators.
Collapse
Affiliation(s)
- I. Plastira
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - E. Bernhart
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - M. Goeritzer
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria ,grid.452216.6BioTechMed-Graz, Graz, Austria
| | - T. DeVaney
- 0000 0000 8988 2476grid.11598.34Institute of Biophysics, Medical University of Graz, Graz, Austria
| | - H. Reicher
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - A. Hammer
- 0000 0000 8988 2476grid.11598.34Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - B. Lohberger
- 0000 0000 8988 2476grid.11598.34Department of Orthopedic Surgery, Medical University of Graz, Graz, Austria
| | - A. Wintersperger
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - B. Zucol
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - W. F. Graier
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria ,grid.452216.6BioTechMed-Graz, Graz, Austria
| | - D. Kratky
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria ,grid.452216.6BioTechMed-Graz, Graz, Austria
| | - E. Malle
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria
| | - W. Sattler
- 0000 0000 8988 2476grid.11598.34Institute of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, 8010 Graz, Austria ,grid.452216.6BioTechMed-Graz, Graz, Austria
| |
Collapse
|
40
|
Drago F, Lombardi M, Prada I, Gabrielli M, Joshi P, Cojoc D, Franck J, Fournier I, Vizioli J, Verderio C. ATP Modifies the Proteome of Extracellular Vesicles Released by Microglia and Influences Their Action on Astrocytes. Front Pharmacol 2017; 8:910. [PMID: 29321741 PMCID: PMC5733563 DOI: 10.3389/fphar.2017.00910] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 11/29/2017] [Indexed: 11/23/2022] Open
Abstract
Extracellular ATP is among molecules promoting microglia activation and inducing the release of extracellular vesicles (EVs), which are potent mediators of intercellular communication between microglia and the microenvironment. We previously showed that EVs produced under ATP stimulation (ATP-EVs) propagate a robust inflammatory reaction among astrocytes and microglia in vitro and in mice with subclinical neuroinflammation (Verderio et al., 2012). However, the proteome of EVs released upon ATP stimulation has not yet been elucidated. In this study we applied a label free proteomic approach to characterize the proteome of EVs released constitutively and during microglia activation with ATP. We show that ATP drives sorting in EVs of a set of proteins implicated in cell adhesion/extracellular matrix organization, autophagy-lysosomal pathway and cellular metabolism, that may influence the response of recipient astrocytes to EVs. These data provide new clues to molecular mechanisms involved in microglia response to ATP and in microglia signaling to the environment via EVs.
Collapse
Affiliation(s)
- Francesco Drago
- Univ. Lille, INSERM, U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse - PRISM, Lille, France.,Fondazione Istituto Oncologico del Mediterraneo, Viagrande, Italy
| | | | | | | | - Pooja Joshi
- Institute of Neuroscience (CNR), Milan, Italy
| | - Dan Cojoc
- Institute of Materials (CNR), Trieste, Italy
| | - Julien Franck
- Univ. Lille, INSERM, U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse - PRISM, Lille, France
| | - Isabelle Fournier
- Univ. Lille, INSERM, U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse - PRISM, Lille, France
| | - Jacopo Vizioli
- Univ. Lille, INSERM, U1192 - Protéomique Réponse Inflammatoire Spectrométrie de Masse - PRISM, Lille, France
| | - Claudia Verderio
- IRCCS Humanitas, Rozzano, Italy.,Institute of Neuroscience (CNR), Milan, Italy
| |
Collapse
|
41
|
Layé S, Nadjar A, Joffre C, Bazinet RP. Anti-Inflammatory Effects of Omega-3 Fatty Acids in the Brain: Physiological Mechanisms and Relevance to Pharmacology. Pharmacol Rev 2017; 70:12-38. [PMID: 29217656 DOI: 10.1124/pr.117.014092] [Citation(s) in RCA: 240] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 09/05/2017] [Indexed: 12/17/2022] Open
Abstract
Classically, polyunsaturated fatty acids (PUFA) were largely thought to be relatively inert structural components of brain, largely important for the formation of cellular membranes. Over the past 10 years, a host of bioactive lipid mediators that are enzymatically derived from arachidonic acid, the main n-6 PUFA, and docosahexaenoic acid, the main n-3 PUFA in the brain, known to regulate peripheral immune function, have been detected in the brain and shown to regulate microglia activation. Recent advances have focused on how PUFA regulate the molecular signaling of microglia, especially in the context of neuroinflammation and behavior. Several active drugs regulate brain lipid signaling and provide proof of concept for targeting the brain. Because brain lipid metabolism relies on a complex integration of diet, peripheral metabolism, including the liver and blood, which supply the brain with PUFAs that can be altered by genetics, sex, and aging, there are many pathways that can be disrupted, leading to altered brain lipid homeostasis. Brain lipid signaling pathways are altered in neurologic disorders and may be viable targets for the development of novel therapeutics. In this study, we discuss in particular how n-3 PUFAs and their metabolites regulate microglia phenotype and function to exert their anti-inflammatory and proresolving activities in the brain.
Collapse
Affiliation(s)
- Sophie Layé
- Institut National pour la Recherche Agronomique and Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France (S.L., A.N., C.J.); and Department of Nutritional Sciences, University of Toronto, Ontario, Canada (R.P.B.)
| | - Agnès Nadjar
- Institut National pour la Recherche Agronomique and Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France (S.L., A.N., C.J.); and Department of Nutritional Sciences, University of Toronto, Ontario, Canada (R.P.B.)
| | - Corinne Joffre
- Institut National pour la Recherche Agronomique and Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France (S.L., A.N., C.J.); and Department of Nutritional Sciences, University of Toronto, Ontario, Canada (R.P.B.)
| | - Richard P Bazinet
- Institut National pour la Recherche Agronomique and Bordeaux University, Nutrition et Neurobiologie Intégrée, UMR 1286, Bordeaux, France (S.L., A.N., C.J.); and Department of Nutritional Sciences, University of Toronto, Ontario, Canada (R.P.B.)
| |
Collapse
|
42
|
Beneventano M, Spampinato SF, Merlo S, Chisari M, Platania P, Ragusa M, Purrello M, Nicoletti F, Sortino MA. Shedding of Microvesicles from Microglia Contributes to the Effects Induced by Metabotropic Glutamate Receptor 5 Activation on Neuronal Death. Front Pharmacol 2017; 8:812. [PMID: 29170640 PMCID: PMC5684115 DOI: 10.3389/fphar.2017.00812] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/26/2017] [Indexed: 01/08/2023] Open
Abstract
Metabotropic glutamate (mGlu) receptor 5 is involved in neuroinflammation and has been shown to mediate reduced inflammation and neurotoxicity and to modify microglia polarization. On the other hand, blockade of mGlu5 receptor results in inhibition of microglia activation. To dissect this controversy, we investigated whether microvesicles (MVs) released from microglia BV2 cells could contribute to the communication between microglia and neurons and whether this interaction was modulated by mGlu5 receptor. Activation of purinergic ionotropic P2X7 receptor with the stable ATP analog benzoyl-ATP (100 μM) caused rapid MVs shedding from BV2 cells. Ionic currents through P2X7 receptor increased in BV2 cells pretreated for 24 h with the mGlu5 receptor agonist CHPG (200 μM) as by patch-clamp recording. This increase was blunted when microglia cells were activated by exposure to lipopolysaccharide (LPS; 0.1 μg/ml for 6 h). Accordingly, a greater amount of MVs formed after CHPG treatment, an effect prevented by the mGlu5 receptor antagonist MTEP (100 μM), as measured by expression of flotillin, a membrane protein enriched in MVs. Transferred MVs were internalized by SH-SY5Y neurons where they did not modify neuronal death induced by a low concentration of rotenone (0.1 μM for 24 h), but significantly increased rotenone neurotoxicity when shed from CHPG-treated BV2 cells. miR146a was increased in CHPG-treated MVs, an effect concealed in MVs from LPS-activated BV2 cells that showed per se an increase in miRNA146a levels. The present data support a role for microglia-shed MVs in mGlu5-mediated modulation of neuronal death and identify miRNAs as potential critical mediators of this interaction.
Collapse
Affiliation(s)
- Martina Beneventano
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Simona F Spampinato
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sara Merlo
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Mariangela Chisari
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Paola Platania
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Marco Ragusa
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Michele Purrello
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy.,I.R.C.C.S. Neuromed, Pozzilli, Italy
| | - Maria Angela Sortino
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
43
|
Molecular Characterization of Peripheral Extracellular Vesicles in Clinically Isolated Syndrome: Preliminary Suggestions from a Pilot Study. Med Sci (Basel) 2017; 5:medsci5030019. [PMID: 29099035 PMCID: PMC5635803 DOI: 10.3390/medsci5030019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 09/07/2017] [Accepted: 09/14/2017] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs), nanoparticles originated from different cell types, seem to be implicated in several cellular activities. In the Central Nervous System (CNS), glia and neurons secrete EVs and recent studies have demonstrated that the intercellular communication mediated by EVs has versatile functional impact in the cerebral homeostasis. This essential role may be due to their proteins and RNAs cargo that possibly modify the phenotypes of the targeted cells. Despite the increasing importance of EVs, little is known about their fluctuations in physiological as well as in pathological conditions. Furthermore, only few studies have investigated the contents of contemporary EVs subgroups (microvesicles, MVs and exosomes, EXOs) with the purpose of discriminating between their features and functional roles. In order to possibly shed light on these issues, we performed a pilot study in which MVs and EXOs extracted from serum samples of a little cohort of subjects (patients with the first clinical evidence of CNS demyelination, also known as Clinically Isolated Syndrome and Healthy Controls) were submitted to deep small-RNA sequencing. Data were analysed by an in-home bioinformatics platform. In line with previous reports, distinct classes of non-coding RNAs have been detected in both the EVs subsets, offering interesting suggestions on their origins and functions. We also verified the feasibility of this extensive molecular approach, thus supporting its valuable use for the analysis of circulating biomarkers (e.g., microRNAs) in order to investigate and monitor specific diseases.
Collapse
|
44
|
Du C, Wang Y, Zhang F, Yan S, Guan Y, Gong X, Zhang T, Cui X, Wang X, Zhang CX. Synaptotagmin-11 inhibits cytokine secretion and phagocytosis in microglia. Glia 2017; 65:1656-1667. [PMID: 28686317 DOI: 10.1002/glia.23186] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 05/18/2017] [Accepted: 06/20/2017] [Indexed: 12/18/2022]
Abstract
Cytokine secretion and phagocytosis are key functions of activated microglia. However, the molecular mechanisms underlying their regulation in microglia remain largely unknown. Here, we report that synaptotagmin-11 (Syt11), a non-Ca2+ -binding Syt implicated in Parkinson disease and schizophrenia, inhibits cytokine secretion and phagocytosis in microglia. We found Syt11 expression in microglia in brain slices and primary microglia. Interestingly, Syt11-knockdown (KD) increased cytokine secretion and NO release in primary microglia both in the absence and presence of lipopolysaccharide. NF-κB was activated in untreated KD microglia together with enhanced synthesis of IL-6, TNF-α, IL-1β, and iNOS. When the release capacity was assessed by the ratio of extracellular to intracellular levels, only the IL-6 and TNF-α secretion capacity was increased in Syt11-KD cells, suggesting that Syt11 specifically regulates conventional secretion. Consistently, Syt11 localized to the trans-Golgi network and recycling endosomes. In addition, Syt11 was recruited to phagosomes and its deficiency enhanced microglial phagocytosis. All the KD phenotypes were rescued by expression of an shRNA-resistant Syt11, while overexpression of Syt11 suppressed cytokine secretion and phagocytosis. Importantly, Syt11 also inhibited microglial phagocytosis of α-synuclein fibrils, supporting its association with Parkinson disease. Taken together, we propose that Syt11 suppresses microglial activation under both physiological and pathological conditions through the inhibition of cytokine secretion and phagocytosis.
Collapse
Affiliation(s)
- Cuilian Du
- Key Laboratory for the Neurodegenerative Disorders of the Chinese Ministry of Education, Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,Department of Neurobiology, School of Basic Medical Science, Key Laboratory for the Neurodegenerative Disorders of the Chinese Ministry of Education, Capital Medical University, Beijing, China
| | - Yalong Wang
- Key Laboratory for the Neurodegenerative Disorders of the Chinese Ministry of Education, Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Feifan Zhang
- Key Laboratory for the Neurodegenerative Disorders of the Chinese Ministry of Education, Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Shuxin Yan
- Key Laboratory for the Neurodegenerative Disorders of the Chinese Ministry of Education, Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yuan Guan
- Key Laboratory for the Neurodegenerative Disorders of the Chinese Ministry of Education, Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Xiaoli Gong
- Department of Physiology, School of Basic Medical Science, Capital Medical University, Beijing, China
| | - Ting Zhang
- Department of Neurobiology, School of Basic Medical Science, Key Laboratory for the Neurodegenerative Disorders of the Chinese Ministry of Education, Capital Medical University, Beijing, China
| | - Xiuyu Cui
- Key Laboratory for the Neurodegenerative Disorders of the Chinese Ministry of Education, Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Xiaomin Wang
- Key Laboratory for the Neurodegenerative Disorders of the Chinese Ministry of Education, Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,Department of Neurobiology, School of Basic Medical Science, Key Laboratory for the Neurodegenerative Disorders of the Chinese Ministry of Education, Capital Medical University, Beijing, China
| | - Claire Xi Zhang
- Key Laboratory for the Neurodegenerative Disorders of the Chinese Ministry of Education, Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
45
|
TRPV1 channels are critical brain inflammation detectors and neuropathic pain biomarkers in mice. Nat Commun 2017; 8:15292. [PMID: 28489079 PMCID: PMC5436240 DOI: 10.1038/ncomms15292] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 03/14/2017] [Indexed: 12/12/2022] Open
Abstract
The capsaicin receptor TRPV1 has been widely characterized in the sensory system as a key component of pain and inflammation. A large amount of evidence shows that TRPV1 is also functional in the brain although its role is still debated. Here we report that TRPV1 is highly expressed in microglial cells rather than neurons of the anterior cingulate cortex and other brain areas. We found that stimulation of microglial TRPV1 controls cortical microglia activation per se and indirectly enhances glutamatergic transmission in neurons by promoting extracellular microglial microvesicles shedding. Conversely, in the cortex of mice suffering from neuropathic pain, TRPV1 is also present in neurons affecting their intrinsic electrical properties and synaptic strength. Altogether, these findings identify brain TRPV1 as potential detector of harmful stimuli and a key player of microglia to neuron communication.
Collapse
|
46
|
Solé-Domènech S, Cruz DL, Capetillo-Zarate E, Maxfield FR. The endocytic pathway in microglia during health, aging and Alzheimer's disease. Ageing Res Rev 2016; 32:89-103. [PMID: 27421577 DOI: 10.1016/j.arr.2016.07.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 07/01/2016] [Accepted: 07/05/2016] [Indexed: 12/14/2022]
Abstract
Microglia, the main phagocytes of the central nervous system (CNS), are involved in the surveillance and maintenance of nervous tissue. During normal tissue homeostasis, microglia migrates within the CNS, phagocytose dead cells and tissue debris, and modulate synapse pruning and spine formation via controlled phagocytosis. In the event of an invasion by a foreign body, microglia are able to phagocytose the invading pathogen and process it proteolytically for antigen presentation. Internalized substrates are incorporated and sorted within the endocytic pathway and thereafter transported via complex vesicular routes. When targeted for degradation, substrates are delivered to acidic late endosomes and lysosomes. In these, the enzymatic degradation relies on pH and enzyme content. Endocytosis, sorting, transport, compartment acidification and degradation are regulated by complex signaling mechanisms, and these may be altered during aging and pathology. In this review, we discuss the endocytic pathway in microglia, with insight into the mechanisms controlling lysosomal biogenesis and pH regulation. We also discuss microglial lysosome function associated with Alzheimer's disease (AD) and the mechanisms of amyloid-beta (Aβ) internalization and degradation. Finally, we explore some therapies currently being investigated to treat AD and their effects on microglial response to Aβ, with insight in those involving enhancement of lysosomal function.
Collapse
|
47
|
Schiera G, Di Liegro CM, Puleo V, Colletta O, Fricano A, Cancemi P, Di Cara G, Di Liegro I. Extracellular vesicles shed by melanoma cells contain a modified form of H1.0 linker histone and H1.0 mRNA-binding proteins. Int J Oncol 2016; 49:1807-1814. [PMID: 27633859 PMCID: PMC5063456 DOI: 10.3892/ijo.2016.3692] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/13/2016] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs) are now recognized as a fundamental way for cell-to-cell horizontal transfer of properties, in both physiological and pathological conditions. Most of EV-mediated cross-talk among cells depend on the exchange of proteins, and nucleic acids, among which mRNAs, and non-coding RNAs such as different species of miRNAs. Cancer cells, in particular, use EVs to discard molecules which could be dangerous to them (for example differentiation-inducing proteins such as histone H1.0, or antitumor drugs), to transfer molecules which, after entering the surrounding cells, are able to transform their phenotype, and even to secrete factors, which allow escaping from immune surveillance. Herein we report that melanoma cells not only secrete EVs which contain a modified form of H1.0 histone, but also transport the corresponding mRNA. Given the already known role in tumorigenesis of some RNA binding proteins (RBPs), we also searched for proteins of this class in EVs. This study revealed the presence in A375 melanoma cells of at least three RBPs, with apparent MW of about 65, 45 and 38 kDa, which are able to bind H1.0 mRNA. Moreover, we purified one of these proteins, which by MALDI-TOF mass spectrometry was identified as the already known transcription factor MYEF2.
Collapse
Affiliation(s)
- Gabriella Schiera
- Department of Biological Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), I-90128 Palermo, Italy
| | - Carlo Maria Di Liegro
- Department of Biological Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), I-90128 Palermo, Italy
| | - Veronica Puleo
- Department of Biological Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), I-90128 Palermo, Italy
| | - Oriana Colletta
- Department of Biological Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), I-90128 Palermo, Italy
| | - Anna Fricano
- Department of Biological Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), I-90128 Palermo, Italy
| | - Patrizia Cancemi
- Department of Biological Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), I-90128 Palermo, Italy
| | - Gianluca Di Cara
- Center of Experimental Oncobiology (C.OB.S.), La Maddalena Hospital III Level Oncological Dept., Palermo, Italy
| | - Italia Di Liegro
- Department of Experimental Biomedicine and Clinical Neurosciences (BIONEC), University of Palermo, I-90127 Palermo, Italy
| |
Collapse
|
48
|
Abstract
Functional neural competence and integrity require interactive exchanges among sensory and motor neurons, interneurons and glial cells. Recent studies have attributed some of the tasks needed for these exchanges to extracellular vesicles (such as exosomes and microvesicles), which are most prominently involved in shuttling reciprocal signals between myelinating glia and neurons, thus promoting neuronal survival, the immune response mediated by microglia, and synapse assembly and plasticity. Such vesicles have also been identified as important factors in the spread of neurodegenerative disorders and brain cancer. These extracellular vesicle functions add a previously unrecognized level of complexity to transcellular interactions within the nervous system.
Collapse
|
49
|
P2 receptors in cancer progression and metastatic spreading. Curr Opin Pharmacol 2016; 29:17-25. [PMID: 27262778 DOI: 10.1016/j.coph.2016.05.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/27/2016] [Accepted: 05/08/2016] [Indexed: 10/21/2022]
Abstract
Tumor microenvironment is nucleoside and nucleotide rich. Adenosine is a key determinant of the highly immunosuppressive tumor interstitium. Extracellular ATP also affects anti-tumor immunity, albeit its effects on host-tumor interaction are incompletely understood. We give here an overview of recent literature covering the role of nucleotide-selective (P2) plasma membrane receptors in tumor growth and progression. P2 receptors are expressed on both host and cancer cells, where depending on the receptor subtype, the inflammatory infiltrate and the tumor cell type they may drive an anti-tumor response or promote tumor progression. It is anticipated that knowledge of the pharmacology, biochemistry and functional activity of the P2 receptors will allow a better understanding of host-tumor interaction and the development of innovative anti-cancer therapy.
Collapse
|
50
|
Koniusz S, Andrzejewska A, Muraca M, Srivastava AK, Janowski M, Lukomska B. Extracellular Vesicles in Physiology, Pathology, and Therapy of the Immune and Central Nervous System, with Focus on Extracellular Vesicles Derived from Mesenchymal Stem Cells as Therapeutic Tools. Front Cell Neurosci 2016; 10:109. [PMID: 27199663 PMCID: PMC4852177 DOI: 10.3389/fncel.2016.00109] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 04/14/2016] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane-surrounded structures released by most cell types. They are characterized by a specific set of proteins, lipids and nucleic acids. EVs have been recognized as potent vehicles of intercellular communication to transmit biological signals between cells. In addition, pathophysiological roles of EVs in conditions like cancer, infectious diseases and neurodegenerative disorders are well established. In recent years focus has been shifted on therapeutic use of stem cell derived-EVs. Use of stem cell derived-EVs present distinct advantage over the whole stem cells as EVs do not replicate and after intravenous administration, they are less likely to trap inside the lungs. From the therapeutic perspective, the most promising cellular sources of EVs are mesenchymal stem cells (MSCs), which are easy to obtain and maintain. Therapeutic activity of MSCs has been shown in numerous animal models and the beneficial paracrine effect of MSCs may be mediated by EVs. The various components of MSC derived-EVs such as proteins, lipids, and RNA might play a specific therapeutic role. In this review, we characterize the role of EVs in immune and central nervous system (CNS); present evidences for defective signaling of these vesicles in neurodegeneration and therapeutic role of EVs in CNS.
Collapse
Affiliation(s)
- Sylwia Koniusz
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences Warsaw, Poland
| | - Anna Andrzejewska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences Warsaw, Poland
| | - Maurizio Muraca
- Department of Women's and Children's Health, University of Padua Padua, Italy
| | - Amit K Srivastava
- Russel H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore MD, USA
| | - Miroslaw Janowski
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of SciencesWarsaw, Poland; Russel H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, BaltimoreMD, USA
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences Warsaw, Poland
| |
Collapse
|