Beyond Macromolecules: Extracellular Vesicles as Regulators of Inflammatory Diseases
Abstract
:1. Introduction
Isolation Method | Purity | Yield | Time Consumption | Sample Volume Needed |
---|---|---|---|---|
Centrifugation | ||||
Differential ultracentrifugation | Low | Low-moderate | 8 h | 100 mL |
Density-gradient centrifugation | >Differential ultracentrifugation | Low-moderate | 20 h | 1 mL |
Rate-zonal Centrifugation | High | >Density-gradient centrifugation | 2 h | 0.5 mL |
Precipitation | Low | 16 h | 1 mL | |
Size exclusion | ||||
Ultrafiltration | >Differential ultracentrifugation | Very high | 18 h | 0.5 mL |
Sequential filtration | High | <Differential Ultracentrifugation | - | 150 mL |
Isolation kits | High | High | - | 10–100 µL |
Field-flow fractionation | High | High | <1 h | 100 µL |
Size-exclusion Chromatography | High | High | ~1.5 h | 50 mL |
Hydrostatic Filtration dialysis | - | >Differential ultracentrifugation | 9 h | 15–200 mL |
Affinity purification | Very high | Poor | ~45 min | ~100 µL |
Micro/nano-fluidics or chips | ||||
Immune microfluidic | - | Almost 100% | ~100 min | 30 µL |
Viscoelastic flow | Very high | Very high | 5–25 min | <100 µL |
Acoustic separation | Very high | Very high | 25 min | 100 µL |
Membrane Component | Type | Specific Name | Function | Reference |
---|---|---|---|---|
Membrane proteins | Tetraspanins | CD63, CD9, CD82, CD81 | EVs’ tetraspanins, in association with adhesion molecules, such as ICAM, bind to cellular integrins and other adhesion molecules, hence promoting EVs uptake by target recipient cells | [78] |
Integrins | α6β4, α6β1, αvβ5 | Targeting α6β4- and α6β1-integrins on the EVs decreases lung metastasis, whereas αvβ5-integrin targeting of EVs reduces liver metastasis, via interfering with the uptake of the EVs | [80] | |
Membrane lipids | Glycero-Phospholipids | PS | EVs-PS is indirectly recognized by Gas6, leading to MERTK activation in the recipient macrophages, thereby facilitating EVs’ uptake and associated anti-inflammatory response | [81] |
Membrane glycans | Proteoglycans | - | Proteoglycans are abundant on the EVs’ surface, and targeting proteoglycans would reduce EVs’ uptake by inhibiting the glycan–lectin interaction | [82] |
Mannose-containing glycoproteins | - | Mannose-containing glycoproteins are enriched on the EVs’ surface, the blocking of which significantly attenuates EVs’ uptake by ovarian cancer cells | [36] |
Disease | EVs Found in | Function | Reference/s |
---|---|---|---|
Neuroinflammatory disease | |||
MS | CSF and Plasma | Endothelial- or platelet-EVs from MS patients’ plasma promote BBB leakage, resulting in myeloid- and T-cells’ transmigration into CNS contributing to MS neuropathology | [115,116,117] |
AD and PD | CSF and Plasma | Microglia and neuronal-EVs from AD or PD patients transport Aβ, α-synuclein, and tau to the local/distant neurons, leading to neuronal loss | [118,119,120,121] |
CJD | Plasma | PrPSc is selectively packaged into neuronal EVs and EV-mediated transfer of PrPSc contributes to the pathogenetic spread of CJD | [123] |
PML | Serum | JCPyV transfer via the serum EVs of PML patients between the glial cells is infectious and contributes to PML pathogenesis | [124] |
CM | Plasma | Plasmodium-infected red blood cell-derived EVs are implicated in the pathogenesis of CM, and blocking EV biogenesis shows protection against CM | [126] |
Stroke | - | MSC-EVs inhibit pro-inflammatory M1 microglial differentiation, preventing neuroinflammation and brain injury following MCAO | [127] |
SCI | - | EVs released from infiltrating macrophages are loaded with NOX2 which targets PTEN in the recipient neurons and promotes PI3K-AKT-driven outgrowth | [128] |
TBI | Serum | Microglial EVs transfer miR-124-3p to the neurons and target PDE4B to down-regulate the mTOR pathway leading to inhibition of neuronal inflammation and thus promoting neurite growth | [129] |
Cardiovascular inflammatory diseases | |||
Atherosclerosis | Plaque and plasma | EVs from atherogenic plaque, monocytes, and neutrophils trigger the endothelial ICAM-1 expression leading to leukocyte recruitment, adhesion, and trans-endothelial migration via pro-inflammatory signaling mechanisms | [85,130,131] |
Plasma | During plaque maturation stages, platelet-EVs trigger the phagocytosis of ox-LDL by macrophages, and adipose cell-derived EVs stimulate cholesterol efflux by macrophages via pro-inflammatory signaling, both of which lead to the formation of foam cells | [132,133] | |
Plasma | During plaque progression, EVs from inflammatory macrophages promote microcalcification | [134,135] | |
MI and ischemic heart disease | Myocardium | Cardiomyocytes and endothelial-EVs induce the release of pro-inflammatory cytokines and chemokines from infiltrating monocytes, thereby contributing to MI and ischemic heart disease progression | [136] |
Myocardium | Activated macrophage-derived miR-155-enriched EVs are incorporated into cardiac fibroblasts and promote inflammation while suppressing fibroblast proliferation, leading to cardiac rupture | [137] | |
HF | Plasma | Cardiac fibroblast-derived EVs are enriched with miR27a* and miR-21*, promoting cardiac hypertrophy | [138,139] |
Plasma | Cardiomyocyte-derived EVs promote fibroblast proliferation depending on miR-217 transfer | [140] | |
Aneurysms | Intraluminal thrombus of aortic aneurysm | Neutrophil-EVs carry proteases ADAM10 and ADAM17 which degrade aortic walls | [141] |
Plasma | Ficolin-3 + platelet-EVs often contribute to the progression of aortic aneurysms | [142] | |
Respiratory inflammatory diseases | |||
ALI or ARDS | BALF | EVs from alveolar macrophages or alveolar type-I epithelial cells upon infection or sterile stimulation, respectively, trigger pro-inflammatory cytokines and mediators’ release from naïve alveolar macrophages, contributing to the lung inflammation | [143] |
COPD | EVs from bronchial epithelial cells are enriched with miR-210, which regulates autophagy functions and myofibroblast differentiation, the dysregulation of which leads to COPD pathogenesis | [144] | |
PH | miR-143-loaded EVs from PASMCs promote migration and differentiation of PAECs, leading to PH pathogenesis | [145] | |
ILF | BALF | BALF-EVs, loaded with WNT5A, trigger the proliferation of lung fibroblasts, leading to ILF pathogenesis | [146] |
Asthma | Plasma | EVs derived from the plasma of asthma patients are related to epithelial and smooth muscle cell functions | [147] |
Inflammatory diseases of the digestive system | |||
NEC | Breast milk | BM-EVs protect IEC against H/R-induced apoptosis and loss of proliferation | [149] |
Amniotic fluid | AFSC-EVs promote intestinal epithelial proliferation and anti-inflammation, leading to epithelial regeneration to help intestinal recovery from NEC | [150] | |
IBD | Intestinal luminal fluid | TGF-β1+ EVs from IECs under physiological conditions induce Treg and immunosuppressive dendritic cells, leading to the downregulation of IBD severity | [152] |
Intestinal mucosa | miR-223+ EVs from MCs target CLDN8 in the IECs, resulting in the loss of intestinal epithelial tight junctions and increased epithelial permeability | [153] | |
Serum | ANXA1+ EVs from injury induced IECs help in the activation of the wound repair process | [154] | |
Integumentary inflammatory diseases | |||
SLE | Plasma | SLE plasma-EVs promote endothelial release of pro-inflammatory cytokines, endothelial apoptosis, and increased vascular permeability, contributing to secondary tissue leukocyte infiltration | [155] |
Psoriasis | Plasma | IFN-α-triggered mast cell-derived cytoplasmic PLA2+ EVs promote neo-lipid antigen presentation by CD1a+ cells and their concomitant recognition by CD1a-reactive T-cells, leading to IL22 and IL17A release and skin inflammation | [156] |
AD | Plasma | SEVs trigger DMECs to induce the expression of E-selectin, VCAM-1, and ICAM-1 as well as IL-6 release to promote endothelial adhesion and subsequent transmigration of leukocytes, leading to AD progression | [157] |
Musculoskeletal inflammatory diseases | |||
OP | BMIF | aBMSCs, under oxidative stress, release miR-183-5p-laden EVs which target Hmox1 in yBMSCs, thereby leading to the inhibition of proliferation and osteogenic differentiation as well as senescence induction of yBMSCs | [158] |
OA | - | IL-1β-stimulated SFB-derived EVs promote MMP-13 and ADAMTS-5 expression while inhibiting COL2A1 and ACAN expression in articular chondrocytes, leading to OA pathology | [159] |
Urinary inflammatory diseases | |||
AKI | Plasma and Urine | Hypoxia or I/R-induced injured TECs release miR-150-loaded EVs which trigger profibrotic manifestations in renal fibroblasts | [171] |
IgAN | Urine | CCL2 mRNA expression in urinary EVs of IgAN is significantly higher as compared to controls, which is correlated with tubular interstitial inflammation and C3 deposition, reflecting renal injury and impaired renal functions | [161] |
Reproductive system inflammatory diseases | |||
Pregnancy disorders | Plasma | Maternal macrophage derived EVs induce the release of pro-inflammatory cytokines from placental trophoblasts, contributing to maternal inflammatory responses to protect the fetus | [162] |
Amniotic fluid | Placental trophoblast derived EVs, via the transfer of C19MC, prevent virus replication in non-placental cells, leading to embryonic protection against viral infections | [163] | |
Inflammatory diseases of the endocrine system | |||
Obesity | Adipose tissue and Plasma | Obese adipose tissue or plasma EVs show a significant down-regulation of miR-141-3p expression which contributes to glucose intolerance and insulin resistance | [164,165] |
Serum | Brown adipocyte-derived miR-99b-laden EVs target FGF21 in the liver, leading to metabolic dysfunctions such as glucose intolerance | [166] | |
Serum | ATM-EVs are highly expressed with miR-155, which targets PPARγ in adipocytes, myocytes, and primary hepatocytes, leading to glucose intolerance and insulin resistance | [167] | |
EVs of the lymphatic system in inflammatory diseases | |||
Atherosclerosis | Lymph | Lymph-derived EVs in atherosclerotic conditions influence lymphatic dysfunction and associated inflammatory disease progression | [168] |
RA | Lymph | In RA, prolonged inflammation-induced vascular leakage promotes the egress of platelet-derived EVs in the lymphatic system, contributing to the pathogenesis of RA | [169] |
2. Conclusions and Future Directions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
Aβ | β-amyloid |
aBMSC | aged BM stromal cell |
ACAN | aggrecan |
AD | Alzheimer’s disease |
ADAM | a disintegrin and metalloproteinase |
ADAMTS-5 | ADAM metallopeptidase with thrombospondin type 1 motif 5 |
ADSC | adipose-derived stem cell |
AFSM | amniotic fluid stem cell |
AKI | acute kidney injury |
ALI | acute lung injury |
Alix | ALG-2-interacting protein X |
ANXA1 | annexin A1 |
APC | activated protein C |
AQP1 | aquaporin 1 |
ARDS | acute respiratory distress syndrome |
Atg7 | autophagy related 7 |
ATM | adipose tissue macrophage |
BALF | bronchoalveolar lavage fluid |
BBB | blood brain barrier |
BM | bone marrow |
BMIF | BM interstitial fluid |
C3 | complement component 3 |
CAF | cancer-associated fibroblasts |
CCL2 | chemokine (C-C motif) ligand 2 |
CD | cluster of differentiation |
CDC | cardiosphere-derived cell |
CJD | Creutzfeldt-Jacob disease |
CLDN8 | Claudin 8 |
CM | cerebral malaria |
C19MC | chromosome 19 miRNA cluster |
CNS | central nervous system |
COL2A1 | collagen type II alpha 1 chain |
COPD | chronic obstructive pulmonary disease |
CSF | cerebrospinal fluid |
CIA | collagen-induced arthritis |
COVID-19 | coronavirus 19 |
DLD | deterministic later displacement |
DMEC | dermal microvascular endothelial cell |
DNA | deoxyribonucleic acid |
EEV | endothelial EV |
EMT | epithelial to mesenchymal transition |
EPCR | endothelial cell protein C receptor |
ESCRT | endosomal sorting complexes required for transport |
EV | extracellular vesicle |
FGF21 | fibroblast growth factor 21 |
FPLC | fast protein liquid chromatography |
FVIIa | activated factor VII |
Gab2 | Grb2-associated binder 2 |
Gas6 | growth arrest-specific protein 6 |
GBM | glioblastoma multiforme |
GRP78 | glucose-regulated protein 78 |
HF | heart failure |
HFF | human follicular fluid |
HK | human keratinocyte |
Hmox1 | heme oxygenase 1 |
HPLC | high performance liquid chromatography |
H/R | hypoxia/reoxygenation |
HSC70 | heat shock cognate 70 kDa protein |
HSP | heat shock protein |
ICAM-1 | intercellular adhesion molecule 1 |
IBD | inflammatory bowel disease |
IEC | intestinal epithelial cell |
IFN | interferon |
IgAN | immunoglobulin A nephropathy |
IL | interleukin |
ILF | idiopathic lung fibrosis |
I/R | ischemia-reperfusion |
ISEV | international society for extracellular vesicles |
JCPyV | JC polyomavirus |
LC3 | microtubule-associated proteins 1A/1B light chain 3B |
LPS | lipopolysaccharide |
MC | mast cell |
MCAO | middle cerebral artery occlusion |
MERTR | Mer receptor tyrosine kinase |
MI | myocardial infarction |
miRNA | microRNA |
MMP | matrix metalloproteinase |
MISEV | minimal information for studies of extracellular vesicles |
MP | microparticle |
MS | multiple sclerosis |
MSC | mesenchymal stem cell |
mTOR | mammalian target of rapamycin |
MV | microvesicle |
MVB | multivesicular body |
MYD88 | myeloid differentiation primary response 88 |
NEC | necrotizing enterocolitis |
NF-ĸB | nuclear factor kappa-light-chain-enhancer of activated B-cells |
NOX2 | reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 |
NPC | nasopharyngeal carcinoma |
OA | osteoarthritis |
OP | osteoporosis |
ox-LDL | oxidized low density lipoprotein |
PAEC | pulmonary arterial endothelial cell |
PAH | pulmonary arterial hypertension |
PAR | protease-activated receptor |
PASMC | pulmonary arterial smooth muscle cell |
PBMC | peripheral blood mononuclear cell |
PCOS | polycystic ovary syndrome |
PD | Parkinson’s disease |
PDE4B | phosphodiesterase 4B |
PEG | polyethylene glycol |
PH | pulmonary hypertension |
PI3K | phosphoinositide 3-kinase |
PLA2 | phospholipase A2 |
PML | progressive multifocal leukoencephalopathy |
PPARγ | peroxisome proliferator-activated receptor gamma |
PrP | prion protein |
PrPSc | misfolded and transmissible form of PrP |
PS | phosphatidylserine |
PTEN | phosphatase and tensin homolog |
RA | rheumatoid arthritis |
ROS | reactive oxygen species |
RNA | ribonucleic acid |
SCI | spinal cord injury |
SEC | size exclusion chromatography |
SEV | Staphylococcus aureus-derived EV |
SFB | synovial fibroblast |
SLE | systemic lupus erythematosus |
SNARE | soluble N-ethylmaleimide-sensitive factor activating protein receptor |
srIĸB | super repressor NF-kappa-B inhibitor alpha (I-kappa-B) |
STAT3 | signal transducer and activator of transcription 3 |
TAK1 | TGF-β-activated kinase 1 |
TBI | traumatic brain injury |
TEC | tubular endothelial cell |
TF | tissue factor |
TGF-β1 | transforming growth factor β1 |
TH | helper T-cell |
TIC | trauma-induced coagulopathy |
TLR4 | toll-like receptor 4 |
TME | tumor microenvironment |
TNBC | triple-negative breast cancer |
TNF | tumor necrosis factor |
Treg | regulatory T-cell |
TSG101 | tumor susceptibility gene 101 |
UME | uterine microenvironment |
UTI | urinary tract infection |
VCAM-1 | vascular cell adhesion molecule 1 |
vWF | von Willebrand factor |
WNT5A | Wnt family member 5A |
yBMSC | young BMSC |
References
- Combarnous, Y.; Nguyen, T.M.D. Cell Communications among Microorganisms, Plants, and Animals: Origin, Evolution, and Interplays. Int. J. Mol. Sci. 2020, 21, 8052. [Google Scholar] [CrossRef] [PubMed]
- Das, K.; Prasad, R.; Singh, A.; Bhattacharya, A.; Roy, A.; Mallik, S.; Mukherjee, A.; Sen, P. Protease-activated receptor 2 promotes actomyosin dependent transforming microvesicles generation from human breast cancer. Mol. Carcinog. 2018, 57, 1707–1722. [Google Scholar] [CrossRef] [PubMed]
- Das, K.; Prasad, R.; Roy, S.; Mukherjee, A.; Sen, P. The Protease Activated Receptor2 Promotes Rab5a Mediated Generation of Pro-metastatic Microvesicles. Sci. Rep. 2018, 8, 7357. [Google Scholar] [CrossRef]
- Das, K.; Pendurthi, U.R.; Manco-Johnson, M.; Martin, E.J.; Brophy, D.F.; Rao, L.V.M. Factor VIIa treatment increases circulating extracellular vesicles in hemophilia patients: Implications for the therapeutic hemostatic effect of FVIIa. J. Thromb. Haemost. 2022, 20, 1928–1933. [Google Scholar] [CrossRef] [PubMed]
- Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yanez-Mo, M.; Siljander, P.R.-M.; Andreu, Z.; Zavec, A.B.; Borras, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [Green Version]
- Van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
- Das, K.; Keshava, S.; Pendurthi, U.R.; Rao, L.V.M. Factor VIIa suppresses inflammation and barrier disruption through the release of EEVs and transfer of microRNA 10a. Blood 2022, 139, 118–133. [Google Scholar] [CrossRef]
- Das, K.; Paul, S.; Singh, A.; Ghosh, A.; Roy, A.; Ansari, S.A.; Prasad, R.; Mukherjee, A.; Sen, P. Triple-negative breast cancer-derived microvesicles transfer microRNA221 to the recipient cells and thereby promote epithelial-to-mesenchymal transition. J. Biol. Chem. 2019, 294, 13681–13696. [Google Scholar] [CrossRef]
- Das, K.; Rao, L.V.M. The Role of microRNAs in Inflammation. Int. J. Mol. Sci. 2022, 23, 15479. [Google Scholar] [CrossRef]
- Parashar, D.; Singh, A.; Gupta, S.; Sharma, A.; Sharma, M.K.; Roy, K.K.; Chauhan, S.C.; Kashyap, V.K. Emerging Roles and Potential Applications of Non-Coding RNAs in Cervical Cancer. Genes 2022, 13, 1254. [Google Scholar] [CrossRef]
- Parashar, D.; Geethadevi, A.; Aure, M.R.; Mishra, J.; George, J.; Chen, C.; Mishra, M.K.; Tahiri, A.; Zhao, W.; Nair, B.; et al. miRNA551b-3p Activates an Oncostatin Signaling Module for the Progression of Triple-Negative Breast Cancer. Cell Rep. 2019, 29, 4389–4406.e4310. [Google Scholar] [CrossRef] [Green Version]
- Mulcahy, L.A.; Pink, R.C.; Carter, D.R. Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles 2014, 3, 24641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [PubMed]
- Neven, K.Y.; Nawrot, T.S.; Bollati, V. Extracellular Vesicles: How the External and Internal Environment Can Shape Cell-To-Cell Communication. Curr. Environ. Health Rep. 2017, 4, 30–37. [Google Scholar] [CrossRef] [PubMed]
- Pulliero, A.; Pergoli, L.; Maestra, S.L.A.; Micale, R.T.; Camoirano, C.; Bollati, V.; Izzotti, A.; Flora, S.D.E. Extracellular vesicles in biological fluids. A biomarker of exposure to cigarette smoke and treatment with chemopreventive drugs. J. Prev. Med. Hyg. 2019, 60, E327–E336. [Google Scholar] [CrossRef]
- Admyre, C.; Johansson, S.M.; Qazi, K.R.; Filen, J.-J.; Lahesmaa, R.; Norman, M.; Neve, E.P.A.; Scheynius, A.; Gabrielsson, S. Exosomes with immune modulatory features are present in human breast milk. J. Immunol. 2007, 179, 1969–1978. [Google Scholar] [CrossRef] [Green Version]
- Carnino, J.M.; Lee, H.; Jin, Y. Isolation and characterization of extracellular vesicles from Broncho-alveolar lavage fluid: A review and comparison of different methods. Respir. Res. 2019, 20, 240. [Google Scholar] [CrossRef] [Green Version]
- Das, K.; Keshava, S.; Ansari, S.A.; Kondreddy, V.; Esmon, C.T.; Griffin, J.H.; Pendurthi, U.R.; Rao, L.V.M. Factor VIIa induces extracellular vesicles from the endothelium: A potential mechanism for its hemostatic effect. Blood 2021, 137, 3428–3442. [Google Scholar] [CrossRef]
- Ratajczak, M.Z.; Ratajczak, J. Extracellular microvesicles/exosomes: Discovery, disbelief, acceptance, and the future? Leukemia 2020, 34, 3126–3135. [Google Scholar] [CrossRef]
- Holliday, L.S.; Faria, L.P.; Rody, W.J., Jr. Actin and Actin-Associated Proteins in Extracellular Vesicles Shed by Osteoclasts. Int. J. Mol. Sci. 2019, 21, 158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muralidharan-Chari, V.; Clancy, J.; Plou, C.; Romao, M.; Chavrier, P.; Raposo, G.; D’Souza-Schorey, C. ARF6-regulated shedding of tumor cell-derived plasma membrane microvesicles. Curr. Biol. 2009, 19, 1875–1885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cai, H.; Reinisch, K.; Ferro-Novick, S. Coats, tethers, Rabs, and SNAREs work together to mediate the intracellular destination of a transport vesicle. Dev. Cell 2007, 12, 671–682. [Google Scholar] [CrossRef] [Green Version]
- Escola, J.M.; Kleijmeer, M.J.; Stoorvogel, W.; Griffith, J.M.; Yoshie, O.; Geuze, H.J. Selective enrichment of tetraspan proteins on the internal vesicles of multivesicular endosomes and on exosomes secreted by human B-lymphocytes. J. Biol. Chem. 1998, 273, 20121–20127. [Google Scholar] [CrossRef] [Green Version]
- Das, K.; Mukherjee, T.; Shankar, P. The Role of Extracellular Vesicles in the Pathogenesis of Hematological Malignancies: Interaction with Tumor Microenvironment; a Potential Biomarker and Targeted Therapy. Biomolecules 2023, 13, 897. [Google Scholar]
- Kowal, J.; Tkach, M.; Thery, C. Biogenesis and secretion of exosomes. Curr. Opin. Cell Biol. 2014, 29, 116–125. [Google Scholar] [CrossRef] [Green Version]
- Geminard, C.; De Gassart, A.; Blanc, L.; Vidal, M. Degradation of AP2 during reticulocyte maturation enhances binding of hsc70 and Alix to a common site on TFR for sorting into exosomes. Traffic 2004, 5, 181–193. [Google Scholar] [CrossRef] [PubMed]
- van Niel, G.; Porto-Carreiro, I.; Simoes, S.; Raposo, G. Exosomes: A common pathway for a specialized function. J. Biochem. 2006, 140, 13–21. [Google Scholar] [CrossRef]
- Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brügger, B.; Simons, M. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef]
- Glick, D.; Barth, S.; Macleod, K.F. Autophagy: Cellular and molecular mechanisms. J. Pathol. 2010, 221, 3–12. [Google Scholar] [CrossRef] [Green Version]
- Fader, C.M.; Colombo, M.I. Autophagy and multivesicular bodies: Two closely related partners. Cell Death Differ. 2009, 16, 70–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, J.; Camfield, R.; Gorski, S.M. The interplay between exosomes and autophagy-partners in crime. J. Cell Sci. 2018, 131, jcs215210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Borges, F.T.; Reis, L.A.; Schor, N. Extracellular vesicles: Structure, function, and potential clinical uses in renal diseases. Braz. J. Med. Biol. Res. 2013, 46, 824–830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wickman, G.; Julian, L.; Olson, M.F. How apoptotic cells aid in the removal of their own cold dead bodies. Cell Death Differ. 2012, 19, 735–742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thery, C.; Boussac, M.; Véron, P.; Ricciardi-Castagnoli, P.; Raposo, G.; Garin, J.; Amigorena, S. Proteomic analysis of dendritic cell-derived exosomes: A secreted subcellular compartment distinct from apoptotic vesicles. J. Immunol. 2001, 166, 7309–7318. [Google Scholar] [CrossRef]
- Escrevente, C.; Keller, S.; Altevogt, P.; Costa, J. Interaction and uptake of exosomes by ovarian cancer cells. BMC Cancer 2011, 11, 108. [Google Scholar] [CrossRef] [Green Version]
- Kerr, J.F.; Wyllie, A.H.; Currie, A.R. Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer 1972, 26, 239–257. [Google Scholar] [CrossRef] [Green Version]
- Akbar, A.; Malekian, F.; Baghban, N.; Kodam, S.P.; Ullah, M. Methodologies to Isolate and Purify Clinical Grade Extracellular Vesicles for Medical Applications. Cells 2022, 11, 186. [Google Scholar] [CrossRef]
- Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [Green Version]
- Pillalamarri, N.; Abdullah; Ren, G.; Khan, L.; Ullah, A.; Jonnakuti, S.; Ullah, M. Exploring the utility of extracellular vesicles in ameliorating viral infection-associated inflammation, cytokine storm and tissue damage. Transl. Oncol. 2021, 14, 101095. [Google Scholar] [CrossRef]
- Muller, L.; Hong, C.S.; Stolz, D.B.; Watkins, S.C.; Whiteside, T.L. Isolation of biologically-active exosomes from human plasma. J. Immunol. Methods 2014, 411, 55–65. [Google Scholar] [CrossRef]
- Baranyai, T.; Herczeg, K.; Onódi, Z.; Voszka, I.; Módos, K.; Marton, N.; Nagy, G.; Mäger, I.; Wood, M.J.; El Andaloussi, S.; et al. Isolation of Exosomes from Blood Plasma: Qualitative and Quantitative Comparison of Ultracentrifugation and Size Exclusion Chromatography Methods. PLoS ONE 2015, 10, e0145686. [Google Scholar] [CrossRef] [Green Version]
- Ullah, M.; Kodam, S.P.; Mu, Q.; Akbar, A. Microbubbles versus Extracellular Vesicles as Therapeutic Cargo for Targeting Drug Delivery. ACS Nano 2021, 15, 3612–3620. [Google Scholar] [CrossRef] [PubMed]
- Soares Martins, T.; Catita, J.; Martins Rosa, I.; AB da Cruz e Silva, O.; Henriques, A.G. Exosome isolation from distinct biofluids using precipitation and column-based approaches. PLoS ONE 2018, 13, e0198820. [Google Scholar] [CrossRef] [Green Version]
- Serrano-Pertierra, E.; Oliveira-Rodríguez, M.; Rivas, M.; Oliva, P.; Villafani, J.; Navarro, A. Blanco-López, M.C.; Cernuda-Morollón, E. Characterization of Plasma-Derived Extracellular Vesicles Isolated by Different Methods: A Comparison Study. Bioengineering 2019, 6, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, Z.; Wijerathne, H.; Godwin, A.K.; Soper, S.A. Isolation and analysis methods of extracellular vesicles (EVs). Extracell. Vesicles Circ. Nucl. Acids 2021, 2, 80–103. [Google Scholar] [CrossRef]
- Livshits, M.A.; Khomyakova, E.; Evtushenko, E.G.; Lazarev, V.N.; Kulemin, N.A.; Semina, S.E.; Generozov, E.V.; Govorun, V.M. Isolation of exosomes by differential centrifugation: Theoretical analysis of a commonly used protocol. Sci. Rep. 2015, 5, 17319. [Google Scholar] [CrossRef] [Green Version]
- Zarovni, N.; Corrado, A.; Guazzi, P.; Zocco, D.; Lari, E.; Radano, G.; Muhhina, J.; Fondelli, C.; Gavrilova, J.; Chiesi, A. Integrated isolation and quantitative analysis of exosome shuttled proteins and nucleic acids using immunocapture approaches. Methods 2015, 87, 46–58. [Google Scholar] [CrossRef]
- Taylor, D.D.; Shah, S. Methods of isolating extracellular vesicles impact down-stream analyses of their cargoes. Methods 2015, 87, 3–10. [Google Scholar] [CrossRef]
- Salih, M.; Zietse, R.; Hoorn, E.J. Urinary extracellular vesicles and the kidney: Biomarkers and beyond. Am. J. Physiol. Renal Physiol. 2014, 306, F1251–F1259. [Google Scholar] [CrossRef] [Green Version]
- Van Deun, J.; Mestdagh, P.; Sormunen, R.; Cocquyt, V.; Vermaelen, K.; Vandesompele, J.; Bracke, M.; De Wever, O.; Hendrix, A. The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling. J. Extracell. Vesicles 2014, 3, 24858. [Google Scholar] [CrossRef] [Green Version]
- Rikkert, L.G.; Engelaer, M.; Hau, C.M.; Terstappen, L.W.M.M.; Nieuwland, R.; Coumans, F.A.W. Rate zonal centrifugation can partially separate platelets from platelet-derived vesicles. Res. Pract. Thromb. Haemost. 2020, 4, 1053–1059. [Google Scholar] [CrossRef] [PubMed]
- Ullah, A.; Mabood, N.; Maqbool, M.; Khan, L.; Khan, M.; Ullah, M. SAR-CoV-2 infection, emerging new variants and the role of activation induced cytidine deaminase (AID) in lasting immunity. Saudi Pharm. J. 2021, 29, 1181–1184. [Google Scholar] [CrossRef] [PubMed]
- Li, P.; Kaslan, M.; Lee, S.H.; Yao, J.; Gao, Z. Progress in Exosome Isolation Techniques. Theranostics 2017, 7, 789–804. [Google Scholar] [CrossRef]
- Kim, D.K.; Nishida, H.; An, S.Y.; Shetty, A.K.; Bartosh, T.J.; Prockop, D.J. Chromatographically isolated CD63+CD81+ extracellular vesicles from mesenchymal stromal cells rescue cognitive impairments after TBI. Proc. Natl. Acad. Sci. USA 2016, 113, 170–175. [Google Scholar] [CrossRef]
- Zeringer, E.; Barta, T.; Li, M.; Vlassov, A.V. Strategies for isolation of exosomes. Cold Spring Harb. Protoc. 2015, 2015, 319–323. [Google Scholar] [CrossRef] [Green Version]
- Batrakova, E.V.; Kim, M.S. Using exosomes, naturally-equipped nanocarriers, for drug delivery. J. Control. Release 2015, 219, 396–405. [Google Scholar] [CrossRef] [Green Version]
- Yang, D.; Zhang, W.; Zhang, H.; Zhang, F.; Chen, L.; Ma, L.; Larcher, L.M.; Chen, S.; Liu, N.; Zhao, Q.; et al. Progress, opportunity, and perspective on exosome isolation-efforts for efficient exosome-based theranostics. Theranostics 2020, 10, 3684–3707. [Google Scholar] [CrossRef] [PubMed]
- Lobb, R.J.; Becker, M.; Wen, S.W.; Wong, C.S.; Wiegmans, A.P.; Leimgruber, A.; Möller, A. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J. Extracell. Vesicles 2015, 4, 27031. [Google Scholar] [CrossRef]
- Soda, N.; Rehm, B.H.A.; Sonar, P.; Nguyen, N.T.; Shiddiky, M.J.A. Advanced liquid biopsy technologies for circulating biomarker detection. J. Mater. Chem. B 2019, 7, 6670–6704. [Google Scholar] [CrossRef]
- Peterson, M.F.; Otoc, N.; Sethi, J.K.; Gupta, A.; Antes, T.J. Integrated systems for exosome investigation. Methods 2015, 87, 31–45. [Google Scholar] [CrossRef] [PubMed]
- Liu, F.; Vermesh, O.; Mani, V.; Ge, T.J.; Madsen, S.J.; Sabour, A.; Hsu, E.C.; Gowrishankar, G.; Kanada, M.; Jokerst, J.V.; et al. The Exosome Total Isolation Chip. ACS Nano 2017, 11, 10712–10723. [Google Scholar] [CrossRef] [PubMed]
- Kang, D.; Oh, S.; Ahn, S.M.; Lee, B.H.; Moon, M.H. Proteomic analysis of exosomes from human neural stem cells by flow field-flow fractionation and nanoflow liquid chromatography-tandem mass spectrometry. J. Proteome Res. 2008, 7, 3475–3480. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Lyden, D. Asymmetric-flow field-flow fractionation technology for exomere and small extracellular vesicle separation and characterization. Nat. Protoc. 2019, 14, 1027–1053. [Google Scholar] [CrossRef]
- Gamez-Valero, A.; Monguió-Tortajada, M.; Carreras-Planella, L.; Franquesa, M.l.; Beyer, K.; Borràs, F.E. Size-Exclusion Chromatography-based isolation minimally alters Extracellular Vesicles’ characteristics compared to precipitating agents. Sci. Rep. 2016, 6, 33641. [Google Scholar] [CrossRef] [Green Version]
- Gheinani, A.H.; Vögeli, M.; Baumgartner, U.; Vassella, E.; Draeger, A.; Burkhard, F.C.; Monastyrskaya, K. Improved isolation strategies to increase the yield and purity of human urinary exosomes for biomarker discovery. Sci. Rep. 2018, 8, 3945. [Google Scholar] [CrossRef]
- Musante, L.; Tataruch, D.; Gu, D.; Benito-Martin, A.; Calzaferri, G.; Aherne, S.; Holthofer, H. A simplified method to recover urinary vesicles for clinical applications, and sample banking. Sci. Rep. 2014, 4, 7532. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Nguyen, L.T.H.; Hickey, R.; Walters, N.; Wang, X.; Kwak, K.J.; Lee, L.J.; Palmer, A.F.; Reátegui, E. Immunomagnetic sequential ultrafiltration (iSUF) platform for enrichment and purification of extracellular vesicles from biofluids. Sci. Rep. 2021, 11, 8034. [Google Scholar] [CrossRef]
- Guo, S.C.; Tao, S.C.; Dawn, H. Microfluidics-based on-a-chip systems for isolating and analysing extracellular vesicles. J. Extracell. Vesicles 2018, 7, 1508271. [Google Scholar] [CrossRef] [Green Version]
- Liu, C.; Guo, J.; Tian, F.; Yang, N.; Yan, F.; Ding, Y.; Wei, J.; Hu, G.; Nie, G.; Sun, J. Field-Free Isolation of Exosomes from Extracellular Vesicles by Microfluidic Viscoelastic Flows. ACS Nano 2017, 11, 6968–6976. [Google Scholar] [CrossRef] [Green Version]
- Lee, K.; Shao, H.; Weissleder, R.; Lee, H. Acoustic purification of extracellular microvesicles. ACS Nano 2015, 9, 2321–2327. [Google Scholar] [CrossRef] [Green Version]
- Wu, M.; Chen, C.; Wang, Z.; Bachman, H.; Ouyang, Y.; Huang, P.H.; Sadovsky, Y.; Huang, T.J. Separating extracellular vesicles and lipoproteins via acoustofluidics. Lab Chip 2019, 19, 1174–1182. [Google Scholar] [CrossRef] [PubMed]
- Thery, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [Green Version]
- Gardiner, C.; Di Vizio, D.; Sahoo, S.; Théry, C.; Witwer, K.W.; Wauben, M.; Hill, A.F. Techniques used for the isolation and characterization of extracellular vesicles: Results of a worldwide survey. J. Extracell. Vesicles 2016, 5, 32945. [Google Scholar] [CrossRef]
- Horibe, S.; Tanahashi, T.; Kawauchi, S.; Murakami, Y.; Rikitake, Y. Mechanism of recipient cell-dependent differences in exosome uptake. BMC Cancer 2018, 18, 47. [Google Scholar] [CrossRef] [Green Version]
- Takahashi, Y.; Nishikawa, M.; Shinotsuka, H.; Matsui, Y.; Ohara, S.; Imai, T.; Takakura, Y. Visualization and in vivo tracking of the exosomes of murine melanoma B16-BL6 cells in mice after intravenous injection. J. Biotechnol. 2013, 165, 77–84. [Google Scholar] [CrossRef]
- Sharif, S.; Ghahremani, M.H.; Soleimani, M. Delivery of Exogenous miR-124 to Glioblastoma Multiform Cells by Wharton’s Jelly Mesenchymal Stem Cells Decreases Cell Proliferation and Migration, and Confers Chemosensitivity. Stem Cell Rev. Rep. 2018, 14, 236–246. [Google Scholar] [CrossRef] [PubMed]
- Andreu, Z.; Yanez-Mo, M. Tetraspanins in extracellular vesicle formation and function. Front. Immunol. 2014, 5, 442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liangsupree, T.; Multia, E.; Forssen, P.; Fornstedt, T.; Riekkola, M.L. Kinetics and interaction studies of anti-tetraspanin antibodies and ICAM-1 with extracellular vesicle subpopulations using continuous flow quartz crystal microbalance biosensor. Biosens. Bioelectron. 2022, 206, 114151. [Google Scholar] [CrossRef]
- Hoshino, A.; Costa-Silva, B.; Shen, T.L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [Green Version]
- Graham, D.K.; DeRyckere, D.; Davies, K.D.; Earp, H.S. The TAM family: Phosphatidylserine sensing receptor tyrosine kinases gone awry in cancer. Nat. Rev. Cancer 2014, 14, 769–785. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.J.; Wang, C. A review of the regulatory mechanisms of extracellular vesicles-mediated intercellular communication. Cell Commun. Signal 2023, 21, 77. [Google Scholar] [CrossRef]
- Woei, A.J.F.J.; van der Starre, W.E.; Tesselaar, M.E.; Garcia Rodriguez, P.; van Nieuwkoop, C.; Bertina, R.M.; van Dissel, J.T.; Osanto, S. Procoagulant tissue factor activity on microparticles is associated with disease severity and bacteremia in febrile urinary tract infections. Thromb. Res. 2014, 133, 799–803. [Google Scholar] [CrossRef] [PubMed]
- Tans, G.; Rosing, J.; Thomassen, M.C.; Heeb, M.J.; Zwaal, R.F.; Griffin, J.H. Comparison of anticoagulant and procoagulant activities of stimulated platelets and platelet-derived microparticles. Blood 1991, 77, 2641–2648. [Google Scholar] [CrossRef] [Green Version]
- Rautou, P.E.; Leroyer, A.S.; Ramkhelawon, B.; Devue, C.; Duflaut, D.; Vion, A.C.; Nalbone, G.; Castier, Y.; Leseche, G.; Lehoux, S.; et al. Microparticles from human atherosclerotic plaques promote endothelial ICAM-1-dependent monocyte adhesion and transendothelial migration. Circ. Res. 2011, 108, 335–343. [Google Scholar] [CrossRef]
- Zhang, W.; Zhou, X.; Zhang, H.; Yao, Q.; Liu, Y.; Dong, Z. Extracellular vesicles in diagnosis and therapy of kidney diseases. Am. J. Physiol. Ren. Physiol. 2016, 311, F844–F851. [Google Scholar] [CrossRef] [Green Version]
- Hu, J.; Tang, T.; Zeng, Z.; Wu, J.; Tan, X.; Yan, J. The expression of small RNAs in exosomes of follicular fluid altered in human polycystic ovarian syndrome. PeerJ 2020, 8, e8640. [Google Scholar] [CrossRef]
- Herman, S.; Djaldetti, R.; Mollenhauer, B.; Offen, D. CSF-derived extracellular vesicles from patients with Parkinson’s disease induce symptoms and pathology. Brain 2023, 146, 209–224. [Google Scholar] [CrossRef] [PubMed]
- Murphy, C.; Withrow, J.; Hunter, M.; Liu, Y.; Tang, Y.L.; Fulzele, S.; Hamrick, M.W. Emerging role of extracellular vesicles in musculoskeletal diseases. Mol. Asp. Med. 2018, 60, 123–128. [Google Scholar] [CrossRef]
- Yang, J.; Shin, T.S.; Kim, J.S.; Jee, Y.K.; Kim, Y.K. A new horizon of precision medicine: Combination of the microbiome and extracellular vesicles. Exp. Mol. Med. 2022, 54, 466–482. [Google Scholar] [CrossRef]
- Vu, L.T.; Peng, B.; Zhang, D.X.; Ma, V.; Mathey-Andrews, C.A.; Lam, C.K.; Kiomourtzis, T.; Jin, J.; McReynolds, L.; Huang, L.; et al. Tumor-secreted extracellular vesicles promote the activation of cancer-associated fibroblasts via the transfer of microRNA-125b. J. Extracell. Vesicles 2019, 8, 1599680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tian, X.; Liu, Y.; Wang, Z.; Wu, S. miR-144 delivered by nasopharyngeal carcinoma-derived EVs stimulates angiogenesis through the FBXW7/HIF-1alpha/VEGF-A axis. Mol. Ther. Nucleic Acids 2021, 24, 1000–1011. [Google Scholar] [CrossRef] [PubMed]
- Nathan, C.; Ding, A. Nonresolving inflammation. Cell 2010, 140, 871–882. [Google Scholar] [CrossRef] [Green Version]
- Schaue, D.; Micewicz, E.D.; Ratikan, J.A.; Xie, M.W.; Cheng, G.; McBride, W.H. Radiation and inflammation. Semin. Radiat. Oncol. 2015, 25, 4–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parke, D.V.; Parke, A.L. Chemical-induced inflammation and inflammatory diseases. Int. J. Occup. Med. Environ. Health 1996, 9, 211–217. [Google Scholar]
- Medzhitov, R. Inflammation 2010: New adventures of an old flame. Cell 2010, 140, 771–776. [Google Scholar] [CrossRef] [Green Version]
- Chen, X.; Bi, M.; Yang, J.; Cai, J.; Zhang, H.; Zhu, Y.; Zheng, Y.; Liu, Q.; Shi, G.; Zhang, Z. Cadmium exposure triggers oxidative stress, necroptosis, Th1/Th2 imbalance and promotes inflammation through the TNF-alpha/NF-kappaB pathway in swine small intestine. J. Hazard. Mater. 2022, 421, 126704. [Google Scholar] [CrossRef]
- Li, Y.; Yuan, Y.; Huang, Z.X.; Chen, H.; Lan, R.; Wang, Z.; Lai, K.; Chen, H.; Chen, Z.; Zou, Z.; et al. GSDME-mediated pyroptosis promotes inflammation and fibrosis in obstructive nephropathy. Cell Death Differ. 2021, 28, 2333–2350. [Google Scholar] [CrossRef]
- Zeng, Z.; Huang, H.; Zhang, J.; Liu, Y.; Zhong, W.; Chen, W.; Lu, Y.; Qiao, Y.; Zhao, H.; Meng, X.; et al. HDM induce airway epithelial cell ferroptosis and promote inflammation by activating ferritinophagy in asthma. FASEB J. 2022, 36, e22359. [Google Scholar] [CrossRef]
- Cronkite, D.A.; Strutt, T.M. The Regulation of Inflammation by Innate and Adaptive Lymphocytes. J. Immunol. Res. 2018, 2018, 1467538. [Google Scholar] [CrossRef]
- Miyauchi, M.; Ito, Y.; Nakahara, F.; Hino, T.; Nakamura, F.; Iwasaki, Y.; Kawagoshi, T.; Koya, J.; Yoshimi, A.; Arai, S.; et al. Efficient production of human neutrophils from iPSCs that prevent murine lethal infection with immune cell recruitment. Blood 2021, 138, 2555–2569. [Google Scholar] [CrossRef] [PubMed]
- Puschel, F.; Favaro, F.; Redondo-Pedraza, J.; Lucendo, E.; Iurlaro, R.; Marchetti, S.; Majem, B.; Eldering, E.; Nadal, E.; Ricci, J.E.; et al. Starvation and antimetabolic therapy promote cytokine release and recruitment of immune cells. Proc. Natl. Acad. Sci. USA 2020, 117, 9932–9941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chua, R.L.; Lukassen, S.; Trump, S.; Hennig, B.P.; Wendisch, D.; Pott, F.; Debnath, O.; Thürmann, L.; Kurth, F.; Völker, M.T.; et al. COVID-19 severity correlates with airway epithelium-immune cell interactions identified by single-cell analysis. Nat. Biotechnol. 2020, 38, 970–979. [Google Scholar] [CrossRef] [PubMed]
- Tu, C.; Lu, H.; Zhou, T.; Zhang, W.; Deng, L.; Cao, W.; Yang, Z.; Wang, Z.; Wu, X.; Ding, J.; et al. Promoting the healing of infected diabetic wound by an anti-bacterial and nano-enzyme-containing hydrogel with inflammation-suppressing, ROS-scavenging, oxygen and nitric oxide-generating properties. Biomaterials 2022, 286, 121597. [Google Scholar] [CrossRef] [PubMed]
- Siddiqui, Y.D.; Omori, K.; Ito, T.; Yamashiro, K.; Nakamura, S.; Okamoto, K.; Ono, M.; Yamamoto, T.; Van Dyke, T.E.; Takashiba, S. Resolvin D2 Induces Resolution of Periapical Inflammation and Promotes Healing of Periapical Lesions in Rat Periapical Periodontitis. Front. Immunol. 2019, 10, 307. [Google Scholar] [CrossRef]
- Xu, X.; Zeng, Y.; Chen, Z.; Yu, Y.; Wang, H.; Lu, X.; Zhao, J.; Wang, S. Chitosan-based multifunctional hydrogel for sequential wound inflammation elimination, infection inhibition, and wound healing. Int. J. Biol. Macromol. 2023, 235, 123847. [Google Scholar] [CrossRef]
- Pahlavani, N.; Malekahmadi, M.; Firouzi, S.; Rostami, D.; Sedaghat, A.; Moghaddam, A.B.; Ferns, G.A.; Navashenaq, J.G.; Reazvani, R.; Safarian, M.; et al. Molecular and cellular mechanisms of the effects of Propolis in inflammation, oxidative stress and glycemic control in chronic diseases. Nutr. Metab. (Lond) 2020, 17, 65. [Google Scholar] [CrossRef]
- Gomez-Pastora, J.; Weigand, M.; Kim, J.; Wu, X.; Strayer, J.; Palmer, A.F.; Zborowski, M.; Yazer, M.; Chalmers, J.J. Hyperferritinemia in critically ill COVID-19 patients-Is ferritin the product of inflammation or a pathogenic mediator? Clin. Chim. Acta 2020, 509, 249–251. [Google Scholar] [CrossRef]
- Ranneh, Y.; Akim, A.M.; Hamid, H.A.; Khazaai, H.; Fadel, A.; Mahmoud, A.M. Stingless bee honey protects against lipopolysaccharide induced-chronic subclinical systemic inflammation and oxidative stress by modulating Nrf2, NF-kappaB and p38 MAPK. Nutr. Metab. (Lond) 2019, 16, 15. [Google Scholar] [CrossRef] [Green Version]
- Braun, T.P.; Zhu, X.; Szumowski, M.; Scott, G.D.; Grossberg, A.J.; Levasseur, P.R.; Graham, K.; Khan, S.; Damaraju, S.; Colmers, W.F. Central nervous system inflammation induces muscle atrophy via activation of the hypothalamic-pituitary-adrenal axis. J. Exp. Med. 2011, 208, 2449–2463. [Google Scholar] [CrossRef]
- Foley, J.H.; Conway, E.M. Cross Talk Pathways Between Coagulation and Inflammation. Circ. Res. 2016, 118, 1392–1408. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y. Contact pathway of coagulation and inflammation. Thromb. J. 2015, 13, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Levi, M.; Keller, T.T.; van Gorp, E.; ten Cate, H. Infection and inflammation and the coagulation system. Cardiovasc. Res. 2003, 60, 26–39. [Google Scholar] [CrossRef]
- Saenz-Cuesta, M.; Osorio-Querejeta, I.; Otaegui, D. Extracellular Vesicles in Multiple Sclerosis: What are They Telling Us? Front. Cell Neurosci. 2014, 8, 100. [Google Scholar] [CrossRef] [PubMed]
- Saenz-Cuesta, M.; Irizar, H.; Castillo-Triviño, T.; Muñoz-Culla, M.; Osorio-Querejeta, I.; Prada, A.; Sepúlveda, L.; López-Mato, M.P.; López de Munain, A.; Comabella, M.; et al. Circulating microparticles reflect treatment effects and clinical status in multiple sclerosis. Biomark. Med. 2014, 8, 653–661. [Google Scholar] [CrossRef]
- Jimenez, J.; Jy, W.; Mauro, L.M.; Horstman, L.L.; Ahn, E.R.; Ahn, Y.S.; Minagar, A. Elevated endothelial microparticle-monocyte complexes induced by multiple sclerosis plasma and the inhibitory effects of interferon-beta 1b on release of endothelial microparticles, formation and transendothelial migration of monocyte-endothelial microparticle complexes. Mult. Scler. 2005, 11, 310–315. [Google Scholar] [CrossRef]
- Marcos-Ramiro, B.; Oliva Nacarino, P.; Serrano-Pertierra, E.; Blanco-Gelaz, M.A.; Weksler, B.B.; Romero, I.A.; Couraud, P.O.; Tuñón, A.; López-Larrea, C.; Millán, J.; et al. Microparticles in multiple sclerosis and clinically isolated syndrome: Effect on endothelial barrier function. BMC Neurosci. 2014, 15, 110. [Google Scholar] [CrossRef] [Green Version]
- Asai, H.; Ikezu, S.; Tsunoda, S.; Medalla, M.; Luebke, J.; Haydar, T.; Wolozin, B.; Butovsky, O.; Kügler, S.; Ikezu, T. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat. Neurosci. 2015, 18, 1584–1593. [Google Scholar] [CrossRef] [Green Version]
- Joshi, P.; Turola, E.; Ruiz, A.; Bergami, A.; Libera, D.D.; Benussi, L.; Giussani, P.; Magnani, G.; Comi, G.; Legname, G.; et al. Microglia convert aggregated amyloid-beta into neurotoxic forms through the shedding of microvesicles. Cell Death Differ. 2014, 21, 582–593. [Google Scholar] [CrossRef] [Green Version]
- Agosta, F.; Dalla Libera, D.; Spinelli, E.G.; Finardi, A.; Canu, E.; Bergami, A.; Bocchio Chiavetto, L.; Baronio, M.; Comi, G.; Martino, G.; et al. Myeloid microvesicles in cerebrospinal fluid are associated with myelin damage and neuronal loss in mild cognitive impairment and Alzheimer disease. Ann. Neurol. 2014, 76, 813–825. [Google Scholar] [CrossRef]
- Guo, M.; Wang, J.; Zhao, Y.; Feng, Y.; Han, S.; Dong, Q.; Cui, M.; Tieu, K. Microglial exosomes facilitate alpha-synuclein transmission in Parkinson’s disease. Brain 2020, 143, 1476–1497. [Google Scholar] [CrossRef] [PubMed]
- Robertson, C.; Booth, S.A.; Beniac, D.R.; Coulthart, M.B.; Booth, T.F.; McNicol, A. Cellular prion protein is released on exosomes from activated platelets. Blood 2006, 107, 3907–3911. [Google Scholar] [CrossRef] [PubMed]
- Guo, B.B.; Bellingham, S.A.; Hill, A.F. The neutral sphingomyelinase pathway regulates packaging of the prion protein into exosomes. J. Biol. Chem. 2015, 290, 3455–3467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morris-Love, J.; Gee, G.V.; O’Hara, B.A.; Assetta, B.; Atkinson, A.L.; Dugan, A.S.; Haley, S.A.; Atwood, W.J. JC Polyomavirus Uses Extracellular Vesicles To Infect Target Cells. mBio 2019, 10, e00379-19. [Google Scholar] [CrossRef] [Green Version]
- Opadokun, T.; Rohrbach, P. Extracellular vesicles in malaria: An agglomeration of two decades of research. Malar. J. 2021, 20, 442. [Google Scholar] [CrossRef]
- Debs, S.; Cohen, A.; Hosseini-Beheshti, E.; Chimini, G.; Hunt, N.H.; Grau, G.E.R. Interplay of extracellular vesicles and other players in cerebral malaria pathogenesis. Biochim. Biophys. Acta Gen. Subj. 2019, 1863, 325–331. [Google Scholar] [CrossRef]
- Zhao, Y.; Gan, Y.; Xu, G.; Yin, G.; Liu, D. MSCs-Derived Exosomes Attenuate Acute Brain Injury and Inhibit Microglial Inflammation by Reversing CysLT2R-ERK1/2 Mediated Microglia M1 Polarization. Neurochem. Res. 2020, 45, 1180–1190. [Google Scholar] [CrossRef]
- Hervera, A.; De Virgiliis, F.; Palmisano, I.; Zhou, L.; Tantardini, E.; Kong, G.; Hutson, T.; Danzi, M.C.; Perry, R.B.; Santos, C.X.C.; et al. Publisher Correction: Reactive oxygen species regulate axonal regeneration through the release of exosomal NADPH oxidase 2 complexes into injured axons. Nat. Cell Biol. 2018, 20, 1098. [Google Scholar] [CrossRef] [Green Version]
- Huang, S.; Ge, X.; Yu, J.; Han, Z.; Yin, Z.; Li, Y.; Chen, F.; Wang, H.; Zhang, J.; Lei, P. Increased miR-124-3p in microglial exosomes following traumatic brain injury inhibits neuronal inflammation and contributes to neurite outgrowth via their transfer into neurons. FASEB J. 2018, 32, 512–528. [Google Scholar] [CrossRef] [Green Version]
- Gomez, I.; Ward, B.; Souilhol, C.; Recarti, C.; Ariaans, M.; Johnston, J.; Burnett, A.; Mahmoud, M.; Luong, L.A.; West, L.; et al. Neutrophil microvesicles drive atherosclerosis by delivering miR-155 to atheroprone endothelium. Nat. Commun. 2020, 11, 214. [Google Scholar] [CrossRef] [Green Version]
- Tang, N.; Sun, B.; Gupta, A.; Rempel, H.; Pulliam, L. Monocyte exosomes induce adhesion molecules and cytokines via activation of NF-kappaB in endothelial cells. FASEB J. 2016, 30, 3097–3106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Feng, C.; Chen, Q.; Fan, M.; Guo, J.; Liu, Y.; Ji, T.; Zhu, J.; Zhao, X. Platelet-derived microparticles promote phagocytosis of oxidized low-density lipoprotein by macrophages, potentially enhancing foam cell formation. Ann. Transl. Med. 2019, 7, 477. [Google Scholar] [CrossRef] [PubMed]
- Xie, Z.; Wang, X.; Liu, X.; Du, H.; Sun, C.; Shao, X.; Tian, J.; Gu, X.; Wang, H.; Tian, J.; et al. Adipose-Derived Exosomes Exert Proatherogenic Effects by Regulating Macrophage Foam Cell Formation and Polarization. J. Am. Heart Assoc. 2018, 7, e007442. [Google Scholar] [CrossRef] [PubMed]
- New, S.E.; Goettsch, C.; Aikawa, M.; Marchini, J.F.; Shibasaki, M.; Yabusaki, K.; Libby, P.; Shanahan, C.M.; Croce, K.; Aikawa, E. Macrophage-derived matrix vesicles: An alternative novel mechanism for microcalcification in atherosclerotic plaques. Circ. Res. 2013, 113, 72–77. [Google Scholar] [CrossRef]
- Kawakami, R.; Katsuki, S.; Travers, R.; Romero, D.C.; Becker-Greene, D.; Passos, L.S.A.; Higashi, H.; Blaser, M.C.; Sukhova, G.K.; Buttigieg, J.; et al. S100A9-RAGE Axis Accelerates Formation of Macrophage-Mediated Extracellular Vesicle Microcalcification in Diabetes Mellitus. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 1838–1853. [Google Scholar] [CrossRef]
- Loyer, X.; Zlatanova, I.; Devue, C.; Yin, M.; Howangyin, K.Y.; Klaihmon, P.; Guerin, C.L.; Kheloufi, M.; Vilar, J.; Zannis, K.; et al. Intra-Cardiac Release of Extracellular Vesicles Shapes Inflammation Following Myocardial Infarction. Circ. Res. 2018, 123, 100–106. [Google Scholar] [CrossRef]
- Wang, C.; Zhang, C.; Liu, L.; A, X.; Chen, B.; Li, Y.; Du, J. Macrophage-Derived mir-155-Containing Exosomes Suppress Fibroblast Proliferation and Promote Fibroblast Inflammation during Cardiac Injury. Mol. Ther. 2017, 25, 192–204. [Google Scholar] [CrossRef] [Green Version]
- Tian, C.; Hu, G.; Gao, L.; Hackfort, B.T.; Zucker, I.H. Extracellular vesicular MicroRNA-27a* contributes to cardiac hypertrophy in chronic heart failure. J. Mol. Cell Cardiol. 2020, 143, 120–131. [Google Scholar] [CrossRef]
- Bang, C.; Batkai, S.; Dangwal, S.; Gupta, S.K.; Foinquinos, A.; Holzmann, A.; Just, A.; Remke, J.; Zimmer, K.; Zeug, A.; et al. Cardiac fibroblast-derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. J. Clin. Investig. 2014, 124, 2136–2146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nie, X.; Fan, J.; Li, H.; Yin, Z.; Zhao, Y.; Dai, B.; Dong, N.; Chen, C.; Wang, D.W. miR-217 Promotes Cardiac Hypertrophy and Dysfunction by Targeting PTEN. Mol. Ther. Nucleic Acids 2018, 12, 254–266. [Google Scholar] [CrossRef] [Green Version]
- Folkesson, M.; Li, C.; Frebelius, S.; Swedenborg, J.; Wågsäter, D.; Williams, K.J.; Eriksson, P.; Roy, J.; Liu, M.L. Proteolytically active ADAM10 and ADAM17 carried on membrane microvesicles in human abdominal aortic aneurysms. Thromb. Haemost. 2015, 114, 1165–1174. [Google Scholar] [CrossRef] [PubMed]
- Fernandez-Garcia, C.E.; Burillo, E.; Lindholt, J.S.; Martinez-Lopez, D.; Pilely, K.; Mazzeo, C.; Michel, J.B.; Egido, J.; Garred, P.; Blanco-Colio, L.M.; et al. Association of ficolin-3 with abdominal aortic aneurysm presence and progression. J. Thromb. Haemost. 2017, 15, 575–585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, H.; Zhang, D.; Laskin, D.L.; Jin, Y. Functional Evidence of Pulmonary Extracellular Vesicles in Infectious and Noninfectious Lung Inflammation. J. Immunol. 2018, 201, 1500–1509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fujita, Y.; Araya, J.; Ito, S.; Kobayashi, K.; Kosaka, N.; Yoshioka, Y.; Kadota, T.; Hara, H.; Kuwano, K.; Ochiya, T. Suppression of autophagy by extracellular vesicles promotes myofibroblast differentiation in COPD pathogenesis. J. Extracell. Vesicles 2015, 4, 28388. [Google Scholar] [CrossRef] [PubMed]
- Deng, L.; Blanco, F.J.; Stevens, H.; Lu, R.; Caudrillier, A.; McBride, M.; McClure, J.D.; Grant, J.; Thomas, M.; Frid, M.; et al. MicroRNA-143 Activation Regulates Smooth Muscle and Endothelial Cell Crosstalk in Pulmonary Arterial Hypertension. Circ. Res. 2015, 117, 870–883. [Google Scholar] [CrossRef]
- Martin-Medina, A.; Lehmann, M.; Burgy, O.; Hermann, S.; Baarsma, H.A.; Wagner, D.E.; De Santis, M.M.; Ciolek, F.; Hofer, T.P.; Frankenberger, M.; et al. Increased Extracellular Vesicles Mediate WNT5A Signaling in Idiopathic Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2018, 198, 1527–1538. [Google Scholar] [CrossRef]
- Nagano, T.; Katsurada, M.; Dokuni, R.; Hazama, D.; Kiriu, T.; Umezawa, K.; Kobayashi, K.; Nishimura, Y. Crucial Role of Extracellular Vesicles in Bronchial Asthma. Int. J. Mol. Sci. 2019, 20, 2589. [Google Scholar] [CrossRef] [Green Version]
- Collison, A.; Mattes, J.; Plank, M.; Foster, P.S. Inhibition of house dust mite-induced allergic airways disease by antagonism of microRNA-145 is comparable to glucocorticoid treatment. J. Allergy Clin. Immunol. 2011, 128, 160–167.e4. [Google Scholar] [CrossRef]
- Pisano, C.; Galley, J.; Elbahrawy, M.; Wang, Y.; Farrell, A.; Brigstock, D.; Besner, G.E. Human Breast Milk-Derived Extracellular Vesicles in the Protection Against Experimental Necrotizing Enterocolitis. J. Pediatr. Surg. 2020, 55, 54–58. [Google Scholar] [CrossRef] [Green Version]
- Li, B.; Lee, C.; O’Connell, J.S.; Antounians, L.; Ganji, N.; Alganabi, M.; Cadete, M.; Nascimben, F.; Koike, Y.; Hock, A.; et al. Activation of Wnt signaling by amniotic fluid stem cell-derived extracellular vesicles attenuates intestinal injury in experimental necrotizing enterocolitis. Cell Death Dis. 2020, 11, 750. [Google Scholar] [CrossRef]
- Shen, Q.; Huang, Z.; Yao, J.; Jin, Y. Extracellular vesicles-mediated interaction within intestinal microenvironment in inflammatory bowel disease. J. Adv. Res. 2022, 37, 221–233. [Google Scholar] [CrossRef] [PubMed]
- Jiang, L.; Shen, Y.; Guo, D.; Yang, D.; Liu, J.; Fei, X.; Yang, Y.; Zhang, B.; Lin, Z.; Yang, F.; et al. EpCAM-dependent extracellular vesicles from intestinal epithelial cells maintain intestinal tract immune balance. Nat. Commun. 2016, 7, 13045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, M.; Zhao, J.; Cao, M.; Liu, R.; Chen, G.; Li, S.; Xie, Y.; Xie, J.; Cheng, Y.; Huang, L.; et al. Mast cells-derived MiR-223 destroys intestinal barrier function by inhibition of CLDN8 expression in intestinal epithelial cells. Biol. Res. 2020, 53, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leoni, G.; Neumann, P.A.; Kamaly, N.; Quiros, M.; Nishio, H.; Jones, H.R.; Sumagin, R.; Hilgarth, R.S.; Alam, A.; Fredman, G.; et al. Annexin A1-containing extracellular vesicles and polymeric nanoparticles promote epithelial wound repair. J. Clin. Investig. 2015, 125, 1215–1227. [Google Scholar] [CrossRef] [Green Version]
- Atehortua, L.; Rojas, M.; Vásquez, G.; Muñoz-Vahos, C.H.; Vanegas-García, A.; Posada-Duque, R.A.; Castaño, D. Endothelial activation and injury by microparticles in patients with systemic lupus erythematosus and rheumatoid arthritis. Arthritis Res. Ther. 2019, 21, 34. [Google Scholar] [CrossRef] [Green Version]
- Cheung, K.L.; Jarrett, R.; Subramaniam, S.; Salimi, M.; Gutowska-Owsiak, D.; Chen, Y.L.; Hardman, C.; Xue, L.; Cerundolo, V.; Ogg, G. Psoriatic T cells recognize neolipid antigens generated by mast cell phospholipase delivered by exosomes and presented by CD1a. J. Exp. Med. 2016, 213, 2399–2412. [Google Scholar] [CrossRef]
- Kim, J.; Bin, B.H.; Choi, E.J.; Lee, H.G.; Lee, T.R.; Cho, E.G. Staphylococcus aureus-derived extracellular vesicles induce monocyte recruitment by activating human dermal microvascular endothelial cells in vitro. Clin. Exp. Allergy 2019, 49, 68–81. [Google Scholar] [CrossRef] [Green Version]
- Davis, C.; Dukes, A.; Drewry, M.; Helwa, I.; Johnson, M.H.; Isales, C.M.; Hill, W.D.; Liu, Y.; Shi, X.; Fulzele, S.; et al. MicroRNA-183-5p Increases with Age in Bone-Derived Extracellular Vesicles, Suppresses Bone Marrow Stromal (Stem) Cell Proliferation, and Induces Stem Cell Senescence. Tissue Eng. Part. A 2017, 23, 1231–1240. [Google Scholar] [CrossRef]
- Kato, T.; Miyaki, S.; Ishitobi, H.; Nakamura, Y.; Nakasa, T.; Lotz, M.K.; Ochi, M. Exosomes from IL-1beta stimulated synovial fibroblasts induce osteoarthritic changes in articular chondrocytes. Arthritis Res. Ther. 2014, 16, R163. [Google Scholar] [CrossRef] [Green Version]
- Souza, A.C.; Yuen, P.S.; Star, R.A. Microparticles: Markers and mediators of sepsis-induced microvascular dysfunction, immunosuppression, and AKI. Kidney Int. 2015, 87, 1100–1108. [Google Scholar] [CrossRef] [Green Version]
- Feng, Y.; Lv, L.L.; Wu, W.J.; Li, Z.L.; Chen, J.; Ni, H.F.; Zhou, L.T.; Tang, T.T.; Wang, F.M.; Wang, B.; et al. Urinary Exosomes and Exosomal CCL2 mRNA as Biomarkers of Active Histologic Injury in IgA Nephropathy. Am. J. Pathol. 2018, 188, 2542–2552. [Google Scholar] [CrossRef] [Green Version]
- Holder, B.; Jones, T.; Sancho Shimizu, V.; Rice, T.F.; Donaldson, B.; Bouqueau, M.; Forbes, K.; Kampmann, B. Macrophage Exosomes Induce Placental Inflammatory Cytokines: A Novel Mode of Maternal-Placental Messaging. Traffic 2016, 17, 168–178. [Google Scholar] [CrossRef] [Green Version]
- Delorme-Axford, E.; Donker, R.B.; Mouillet, J.F.; Chu, T.; Bayer, A.; Ouyang, Y.; Wang, T.; Stolz, D.B.; Sarkar, S.N.; Morelli, A.E.; et al. Human placental trophoblasts confer viral resistance to recipient cells. Proc. Natl. Acad. Sci. USA 2013, 110, 12048–12053. [Google Scholar] [CrossRef] [PubMed]
- Dang, S.Y.; Leng, Y.; Wang, Z.X.; Xiao, X.; Zhang, X.; Wen, T.; Gong, H.Z.; Hong, A.; Ma, Y. Exosomal transfer of obesity adipose tissue for decreased miR-141-3p mediate insulin resistance of hepatocytes. Int. J. Biol. Sci. 2019, 15, 351–368. [Google Scholar] [CrossRef] [PubMed]
- Castano, C.; Kalko, S.; Novials, A.; Parrizas, M. Obesity-associated exosomal miRNAs modulate glucose and lipid metabolism in mice. Proc. Natl. Acad. Sci. USA 2018, 115, 12158–12163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thomou, T.; Mori, M.A.; Dreyfuss, J.M.; Konishi, M.; Sakaguchi, M.; Wolfrum, C.; Rao, T.N.; Winnay, J.N.; Garcia-Martin, R.; Grinspoon, S.K.; et al. Adipose-derived circulating miRNAs regulate gene expression in other tissues. Nature 2017, 542, 450–455. [Google Scholar] [CrossRef] [Green Version]
- Ying, W.; Riopel, M.; Bandyopadhyay, G.; Dong, Y.; Birmingham, A.; Seo, J.B.; Ofrecio, J.M.; Wollam, J.; Hernandez-Carretero, A.; Fu, W.; et al. Adipose Tissue Macrophage-Derived Exosomal miRNAs Can Modulate In Vivo and In Vitro Insulin Sensitivity. Cell 2017, 171, 372–384.e312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Milasan, A.; Tessandier, N.; Tan, S.; Brisson, A.; Boilard, E.; Martel, C. Extracellular vesicles are present in mouse lymph and their level differs in atherosclerosis. J. Extracell. Vesicles 2016, 5, 31427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tessandier, N.; Melki, I.; Cloutier, N.; Allaeys, I.; Miszta, A.; Tan, S.; Milasan, A.; Michel, S.; Benmoussa, A.; Lévesque, T.; et al. Platelets Disseminate Extracellular Vesicles in Lymph in Rheumatoid Arthritis. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 929–942. [Google Scholar] [CrossRef]
- Lanna, A.; Vaz, B.; D’Ambra, C.; Valvo, S.; Vuotto, C.; Chiurchiù, V.; Devine, O.; Sanchez, M.; Borsellino, G.; Akbar, A.N.; et al. An intercellular transfer of telomeres rescues T cells from senescence and promotes long-term immunological memory. Nat. Cell Biol. 2022, 24, 1461–1474. [Google Scholar] [CrossRef]
- Guan, H.; Peng, R.; Mao, L.; Fang, F.; Xu, B.; Chen, M. Injured tubular epithelial cells activate fibroblasts to promote kidney fibrosis through miR-150-containing exosomes. Exp. Cell Res. 2020, 392, 112007. [Google Scholar] [CrossRef]
- Macfarlane, R.G. An Enzyme Cascade in the Blood Clotting Mechanism, and Its Function as a Biochemical Amplifier. Nature 1964, 202, 498–499. [Google Scholar] [CrossRef]
- Coughlin, S.R. How the protease thrombin talks to cells. Proc. Natl. Acad. Sci. USA 1999, 96, 11023–11027. [Google Scholar] [CrossRef] [PubMed]
- Jackson, S.P.; Darbousset, R.; Schoenwaelder, S.M. Thromboinflammation: Challenges of therapeutically targeting coagulation and other host defense mechanisms. Blood 2019, 133, 906–918. [Google Scholar] [CrossRef] [Green Version]
- Wang, M.; Hao, H.; Leeper, N.J.; Zhu, L.; Early Career, C. Thrombotic Regulation From the Endothelial Cell Perspectives. Arterioscler. Thromb. Vasc. Biol. 2018, 38, e90–e95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dumnicka, P.; Maduzia, D.; Ceranowicz, P.; Olszanecki, R.; Drożdż, R.; Kuśnierz-Cabala, B. The Interplay between Inflammation, Coagulation and Endothelial Injury in the Early Phase of Acute Pancreatitis: Clinical Implications. Int. J. Mol. Sci. 2017, 18, 354. [Google Scholar] [CrossRef] [Green Version]
- Garcia, J.G.; Pavalko, F.M.; Patterson, C.E. Vascular endothelial cell activation and permeability responses to thrombin. Blood Coagul. Fibrinolysis 1995, 6, 609–626. [Google Scholar] [CrossRef]
- Kondreddy, V.; Keshava, S.; Das, K.; Magisetty, J.; Rao, L.V.M.; Pendurthi, U.R. The Gab2-MALT1 axis regulates thromboinflammation and deep vein thrombosis. Blood 2022, 140, 1549–1564. [Google Scholar] [CrossRef] [PubMed]
- Giesen, P.L.; Rauch, U.; Bohrmann, B.; Kling, D.; Roqué, M.; Fallon, J.T.; Badimon, J.J.; Himber, J.; Riederer, M.A.; Nemerson, Y. Blood-borne tissue factor: Another view of thrombosis. Proc. Natl. Acad. Sci. USA 1999, 96, 2311–2315. [Google Scholar] [CrossRef]
- Reddy, E.C.; Rand, M.L. Procoagulant Phosphatidylserine-Exposing Platelets in vitro and in vivo. Front. Cardiovasc. Med. 2020, 7, 15. [Google Scholar] [CrossRef] [Green Version]
- Heemskerk, J.W.; Bevers, E.M.; Lindhout, T. Platelet activation and blood coagulation. Thromb. Haemost. 2002, 88, 186–193. [Google Scholar] [PubMed]
- Perez-Casal, M.; Downey, C.; Fukudome, K.; Marx, G.; Toh, C.H. Activated protein C induces the release of microparticle-associated endothelial protein C receptor. Blood 2005, 105, 1515–1522. [Google Scholar] [CrossRef]
- Oggero, S.; Austin-Williams, S.; Norling, L.V. The Contrasting Role of Extracellular Vesicles in Vascular Inflammation and Tissue Repair. Front. Pharmacol. 2019, 10, 1479. [Google Scholar] [CrossRef]
- Chatterjee, V.; Yang, X.; Ma, Y.; Wu, M.H.; Yuan, S.Y. Extracellular vesicles: New players in regulating vascular barrier function. Am. J. Physiol. Heart Circ. Physiol. 2020, 319, H1181–H1196. [Google Scholar] [CrossRef] [PubMed]
- Njock, M.S.; Cheng, H.S.; Dang, L.T.; Nazari-Jahantigh, M.; Lau, A.C.; Boudreau, E.; Roufaiel, M.; Cybulsky, M.I.; Schober, A.; Fish, J.E. Endothelial cells suppress monocyte activation through secretion of extracellular vesicles containing antiinflammatory microRNAs. Blood 2015, 125, 3202–3212. [Google Scholar] [CrossRef] [Green Version]
- Das, K.; Prasad, R.; Ansari, S.A.; Roy, A.; Mukherjee, A.; Sen, P. Matrix metalloproteinase-2: A key regulator in coagulation proteases mediated human breast cancer progression through autocrine signaling. Biomed. Pharmacother. 2018, 105, 395–406. [Google Scholar] [CrossRef] [PubMed]
- Dabrowska, S.; Andrzejewska, A.; Lukomska, B.; Janowski, M. Neuroinflammation as a target for treatment of stroke using mesenchymal stem cells and extracellular vesicles. J. Neuroinflamm. 2019, 16, 178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhuang, X.; Xiang, X.; Grizzle, W.; Sun, D.; Zhang, S.; Axtell, R.C.; Ju, S.; Mu, J.; Zhang, L.; Steinman, L.; et al. Treatment of brain inflammatory diseases by delivering exosome encapsulated anti-inflammatory drugs from the nasal region to the brain. Mol. Ther. 2011, 19, 1769–1779. [Google Scholar] [CrossRef] [PubMed]
- Chivero, E.T.; Liao, K.; Niu, F.; Tripathi, A.; Tian, C.; Buch, S.; Hu, G. Engineered Extracellular Vesicles Loaded With miR-124 Attenuate Cocaine-Mediated Activation of Microglia. Front. Cell Dev. Biol. 2020, 8, 573. [Google Scholar] [CrossRef]
- Cambier, L.; de Couto, G.; Ibrahim, A.; Echavez, A.K.; Valle, J.; Liu, W.; Kreke, M.; Smith, R.R.; Marbán, L.; Marbán, E. Y RNA fragment in extracellular vesicles confers cardioprotection via modulation of IL-10 expression and secretion. EMBO Mol. Med. 2017, 9, 337–352. [Google Scholar] [CrossRef]
- Liu, H.; Gao, W.; Yuan, J.; Wu, C.; Yao, K.; Zhang, L.; Ma, L.; Zhu, J.; Zou, Y.; Ge, J. Exosomes derived from dendritic cells improve cardiac function via activation of CD4(+) T lymphocytes after myocardial infarction. J. Mol. Cell Cardiol. 2016, 91, 123–133. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Jiang, M.; Deng, S.; Lu, J.; Huang, H.; Zhang, Y.; Gong, P.; Shen, X.; Ruan, H.; Jin, M.; et al. miR-93-5p-Containing Exosomes Treatment Attenuates Acute Myocardial Infarction-Induced Myocardial Damage. Mol. Ther. Nucleic Acids 2018, 11, 103–115. [Google Scholar] [CrossRef] [Green Version]
- Lanyu, Z.; Feilong, H. Emerging role of extracellular vesicles in lung injury and inflammation. Biomed. Pharmacother. 2019, 113, 108748. [Google Scholar] [CrossRef] [PubMed]
- Pinkerton, M.; Chinchilli, V.; Banta, E.; Craig, T.; August, A.; Bascom, R.; Cantorna, M.; Harvill, E.; Ishmael, F.T. Differential expression of microRNAs in exhaled breath condensates of patients with asthma, patients with chronic obstructive pulmonary disease, and healthy adults. J. Allergy Clin. Immunol. 2013, 132, 217–219. [Google Scholar] [CrossRef]
- Worthington, E.N.; Hagood, J.S. Therapeutic Use of Extracellular Vesicles for Acute and Chronic Lung Disease. Int. J. Mol. Sci. 2020, 21, 2318. [Google Scholar] [CrossRef] [Green Version]
- Liu, A.; Zhang, X.; He, H.; Zhou, L.; Naito, Y.; Sugita, S.; Lee, J.W. Therapeutic potential of mesenchymal stem/stromal cell-derived secretome and vesicles for lung injury and disease. Expert. Opin. Biol. Ther. 2020, 20, 125–140. [Google Scholar] [CrossRef]
- Li, Q.; Zhao, H.; Chen, W.; Huang, P.; Bi, J. Human keratinocyte-derived microvesicle miRNA-21 promotes skin wound healing in diabetic rats through facilitating fibroblast function and angiogenesis. Int. J. Biochem. Cell Biol. 2019, 114, 105570. [Google Scholar] [CrossRef]
- Choi, H.; Kim, Y.; Mirzaaghasi, A.; Heo, J.; Kim, Y.N.; Shin, J.H.; Kim, S.; Kim, N.H.; Cho, E.S.; In Yook, J.; et al. Exosome-based delivery of super-repressor IkappaBalpha relieves sepsis-associated organ damage and mortality. Sci. Adv. 2020, 6, eaaz6980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, S.H.; Lechman, E.R.; Bianco, N.; Menon, R.; Keravala, A.; Nash, J.; Mi, Z.; Watkins, S.C.; Gambotto, A.; Robbins, P.D. Exosomes derived from IL-10-treated dendritic cells can suppress inflammation and collagen-induced arthritis. J. Immunol. 2005, 174, 6440–6448. [Google Scholar] [CrossRef]
- Cosenza, S.; Toupet, K.; Maumus, M.; Luz-Crawford, P.; Blanc-Brude, O.; Jorgensen, C.; Noël, D. Mesenchymal stem cells-derived exosomes are more immunosuppressive than microparticles in inflammatory arthritis. Theranostics 2018, 8, 1399–1410. [Google Scholar] [CrossRef]
- Hwang, H.S.; Kim, H.; Han, G.; Lee, J.W.; Kim, K.; Kwon, I.C.; Yang, Y.; Kim, S.H. Extracellular Vesicles as Potential Therapeutics for Inflammatory Diseases. Int. J. Mol. Sci. 2021, 22, 5487. [Google Scholar] [CrossRef] [PubMed]
- Garofalo, M.; Villa, A.; Crescenti, D.; Marzagalli, M.; Kuryk, L.; Limonta, P.; Mazzaferro, V.; Ciana, P. Heterologous and cross-species tropism of cancer-derived extracellular vesicles. Theranostics 2019, 9, 5681–5693. [Google Scholar] [CrossRef] [PubMed]
- Antimisiaris, S.G.; Mourtas, S.; Marazioti, A. Exosomes and Exosome-Inspired Vesicles for Targeted Drug Delivery. Pharmaceutics 2018, 10, 218. [Google Scholar] [CrossRef] [Green Version]
- Meng, W.; He, C.; Hao, Y.; Wang, L.; Li, L.; Zhu, G. Prospects and challenges of extracellular vesicle-based drug delivery system: Considering cell source. Drug Deliv. 2020, 27, 585–598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef]
- Gudbergsson, J.M.; Jonsson, K.; Simonsen, J.B.; Johnsen, K.B. Systematic review of targeted extracellular vesicles for drug delivery—Considerations on methodological and biological heterogeneity. J. Control. Release 2019, 306, 108–120. [Google Scholar] [CrossRef]
- Stritzke, F.; Poeck, H.; Heidegger, S. In Vivo Immunogenicity Screening of Tumor-Derived Extracellular Vesicles by Flow Cytometry of Splenic T Cells. J. Vis. Exp. 2021, 175, e62811. [Google Scholar] [CrossRef]
- Kim, M.S.; Haney, M.J.; Zhao, Y.; Yuan, D.; Deygen, I.; Klyachko, N.L.; Kabanov, A.V.; Batrakova, E.V. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: In vitro and in vivo evaluations. Nanomedicine 2018, 14, 195–204. [Google Scholar] [CrossRef]
- Stickney, Z.; Losacco, J.; McDevitt, S.; Zhang, Z.; Lu, B. Development of exosome surface display technology in living human cells. Biochem. Biophys. Res. Commun. 2016, 472, 53–59. [Google Scholar] [CrossRef]
- Kooijmans, S.A.A.; Fliervoet, L.A.L.; van der Meel, R.; Fens, M.H.A.M.; Heijnen, H.F.G.; van Bergen En Henegouwen, P.M.P.; Vader, P.; Schiffelers, R.M. PEGylated and targeted extracellular vesicles display enhanced cell specificity and circulation time. J. Control. Release 2016, 224, 77–85. [Google Scholar] [CrossRef]
- Jang, S.C.; Kim, O.Y.; Yoon, C.M.; Choi, D.S.; Roh, T.Y.; Park, J.; Nilsson, J.; Lötvall, J.; Kim, Y.K.; Gho, Y.S. Bioinspired exosome-mimetic nanovesicles for targeted delivery of chemotherapeutics to malignant tumors. ACS Nano 2013, 7, 7698–7710. [Google Scholar] [CrossRef] [PubMed]
- Garcia-Manrique, P.; Gutierrez, G.; Blanco-Lopez, M.C. Fully Artificial Exosomes: Towards New Theranostic Biomaterials. Trends Biotechnol. 2018, 36, 10–14. [Google Scholar] [CrossRef] [Green Version]
- Kooijmans, S.A.; Vader, P.; van Dommelen, S.M.; van Solinge, W.W.; Schiffelers, R.M. Exosome mimetics: A novel class of drug delivery systems. Int. J. Nanomed. 2012, 7, 1525–1541. [Google Scholar] [CrossRef] [Green Version]
- Rosso, G.; Cauda, V. Biomimicking Extracellular Vesicles with Fully Artificial Ones: A Rational Design of EV-BIOMIMETICS toward Effective Theranostic Tools in Nanomedicine. ACS Biomater. Sci. Eng. 2022. [Google Scholar] [CrossRef] [PubMed]
EVs’ Type | Size (Diameter) | Marker/s | Biogenetic Mechanism |
---|---|---|---|
Microvesicles | 100 nm to 1 µm | Tetraspanins | Microvesicles are generated by outward budding of the plasma membrane of the cell |
Exosomes | 30 nm to 150 nm | TSG101, Alix, HSP90β, HSC70 | Invagination of the early endosomal membrane produces exosomes which mature into multivesicular bodies (MVBs). MVBs fuse with the plasma membrane to release the exosomes outside the cells. MVBs can also fuse with auto-phagosomes to form amphisomes which eventually fuse with the plasma membrane to release the exosomes from the cells |
Apoptotic bodies | 50 nm to 5 µm | HSP60, GRP78, Histones | Increased hydrodynamic forces, generated during apoptosis-induced cell contraction, segregate the plasma membrane from cytoskeleton to release such bodies |
EVs Isolation Technique | Purity | Recovery |
---|---|---|
Precipitation kits or polymer (PEG or others) | Low | High |
Low molecular weight cut off centrifugal filters with no further separation steps | Low | High |
High speed ultracentrifugation with no previous lower speed steps | Low | High |
Size-exclusion chromatography | Moderate | Moderate |
High molecular weight centrifugal filters | Moderate | Moderate |
Differential ultracentrifugation with intermediate time/speed with/without wash | Moderate | Moderate |
Tangential flow filtration | Moderate | Moderate |
Membrane affinity columns | Moderate | Moderate |
Filtration combined with size-exclusion chromatography | High | Low |
Immuno- or other affinity isolation with flow cytometry | High | Low |
Surface charge-based isolation techniques | High | Low |
Disease | Model | EVs’ Source | Function | Reference |
---|---|---|---|---|
Neuroinflammatory diseases | Ischemic stroke | MSC | MSC-EVs prevent activation of astrocytes, infiltrating leukocytes, and microglial cells | [187] |
LPS-induced brain inflammation | T-lymphoblast | Curcumin-laden EVs induce apoptosis of inflamed brain microglial cells | [188] | |
Cocaine-induced brain inflammation | DC | miR-124-laden EVs attenuate microglial activation and expression of pro-inflammatory mediators, TLR4, MYD88, STAT3, and NF-ĸB p65 | [189] | |
Cardiovascular inflammatory diseases | I/R-induced cardiac | CDC | CDC-EVs with Y-RNA fragments promote IL-10 release in the infarcted myocardium and trigger inflammation post-MI cardiac repair | [190] |
Cardiac MI inflammation | DC | DC-EVs promote IL-10 release from CD4+T-cells, reducing inflammation and improved cardiac functions | [191] | |
Ischemia-induced cardiac injury | ADSC | ADSC-EVs with miR-93-5p target Atg7 and TLR4, thereby attenuating autophagy and inflammation to protect against infarction-induced myocardial damage | [192] | |
Respiratory inflammatory diseases | Asthma and COPD | MSC | MSC-EVs’ miR-21-5p targets ROS-triggered apoptotic pathway in epithelial cells | [193,194] |
Lung inflammation | MSC | MSC-EVs promote the conversion of alveolar macrophages into M2 phenotypes, leading to anti-inflammation and would healing | [195] | |
ALI/ARDS | MSC | MSC-EVs inhibit proliferation and differentiation of B-cells and promote differentiation of TH-cells to Treg cells, leading to anti-inflammatory cytokines release while attenuating pro-inflammatory cytokines | [196] | |
Integumentary inflammatory disease | Diabetes | HK | miR-21+ HK-EVs promote angiogenesis and facilitate fibroblast function, leading to skin wound healing | [197] |
Sepsis-induced inflammation | - | srIĸB-EVs inhibit the NF-ĸB pathway in neutrophils and monocytes, alleviating sepsis-induced inflammatory responses | [198] | |
Autoimmune inflammatory diseases | Arthritis | DC | IL-10-treated DC-EVs not only inhibit the onset of arthritis but also lower the severity of already-established disease | [199] |
Collagen-induced RA | MSC | MSC-EVs exert anti-inflammatory effects on B- and T-lymphocytes | [200] |
NTA Number | Disease | Phase | EVs’ Source | Age and Sex | No. of Participants | Recruitment Status |
---|---|---|---|---|---|---|
NCT03384433 | Cerebrovascular disease | I and II | Allogenic MSCs | 40–80 years, both M and F | 5 | Unknown by 15 January 2021 |
NCT04602104 | ARDS | I and II | Allogenic human MSCs | 18–70 years, both M and F | 169 | Unknown by 2 November 2021 |
NCT04493242 | COVID-19, ARDS | II | BM-MSCs | 18–85 years | 102 | Completed by 11 April 2023 |
NCT04602442 | SARS-CoV-2 pneumonia | II | MSCs | 18–65 years both M and F | 90 | Unknown by 26 October 2020 |
NCT04276987 | Coronavirus | I | Allogenic adipose MSCs | 18–75 years both M and F | 24 | Completed by 7 September 2020 |
NCT02565264 | Ulcer | Early I | Platelets | Child, adult, Older adult | 5 | Unknown by 9 September 2020 |
NCT04664738 | Skin graft | I | Platelets | 18–75 years both M and F | 37 | Enrolling by invitation by 27 June 2023 |
NCT02138331 | Diabetes Mellitus type 1 | II and III | MSCs | 18–60 years both M and F | 20 | Unknown by 14 May 2014 |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Das, K.; Paul, S.; Mukherjee, T.; Ghosh, A.; Sharma, A.; Shankar, P.; Gupta, S.; Keshava, S.; Parashar, D. Beyond Macromolecules: Extracellular Vesicles as Regulators of Inflammatory Diseases. Cells 2023, 12, 1963. https://doi.org/10.3390/cells12151963
Das K, Paul S, Mukherjee T, Ghosh A, Sharma A, Shankar P, Gupta S, Keshava S, Parashar D. Beyond Macromolecules: Extracellular Vesicles as Regulators of Inflammatory Diseases. Cells. 2023; 12(15):1963. https://doi.org/10.3390/cells12151963
Chicago/Turabian StyleDas, Kaushik, Subhojit Paul, Tanmoy Mukherjee, Arnab Ghosh, Anshul Sharma, Prem Shankar, Saurabh Gupta, Shiva Keshava, and Deepak Parashar. 2023. "Beyond Macromolecules: Extracellular Vesicles as Regulators of Inflammatory Diseases" Cells 12, no. 15: 1963. https://doi.org/10.3390/cells12151963
APA StyleDas, K., Paul, S., Mukherjee, T., Ghosh, A., Sharma, A., Shankar, P., Gupta, S., Keshava, S., & Parashar, D. (2023). Beyond Macromolecules: Extracellular Vesicles as Regulators of Inflammatory Diseases. Cells, 12(15), 1963. https://doi.org/10.3390/cells12151963