Review Article
Austin Neurosurg Open Access. 2022; 8(1): 1069.
The Enigmatic Regulators of Central Nervous System Diseases- Extracellular Vesicles Derived from Mesenchymal Stem Cell
Sun Y1,2, Zhang Y1,2, Zhang W1,2,3* and Yang Z1,2,3*
¹Department of Neurosurgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
²Guangdong Provincial Engineering and Technology Research Center of Stem Cell Therapy for Pituitary Disease, Guangzhou, China
³Critical Disease Stem Cell Therapy Innovation Team, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
*Corresponding author: Wei Zhang, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, China
Zhiqian Yang, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, China
Received: March 07, 2022; Accepted: March 30, 2022; Published: April 06, 2022
Abstract
Stem cells can spontaneously secret extracellular vesicle (EVs), which containing proteins, lipids, and nucleic acids. EVs have a broad prospect as a treatment of central nervous system (CNS) diseases, the release and uptake of EVs has important physiological functions and may also contribute to the development and propagation of inflammation, vascular, malignant, trauma and neurodegenerative diseases. This review will detail and discuss the characteristics of EVs and the potential challenges and strategies in the treatment of EVs-based therapies for CNS diseases in the future.
Keywords: Stem cells; Central nervous system; Extracellular vesicles; miRNA; Therapy
Introduction
CNS has complex anatomy and communicated with diverse cellular populations to respond to environmental stimuli and their metabolic requests. The diversity of CNS diseases can be caused by multifactor, and the existence of the blood-brain barrier has brought great difficulties to the treatment of CNS diseases. Currently, numerous of research developed to promote neurological recovery, but no drugs on the market is available. Increasing evidence is demonstrating that Mesenchymal stem cells (MSCs) derived EVs (MSCs-EVs) shown the positive effects and that the therapeutic value mainly attributed to the miRNA enriched EVs. This review summarized the characteristics of different components of EVs and the main mechanisms involved therapeutic approaches in CNS diseases, focusing on miRNA enriched MSCs-EVs, elucidating how and why miRNA enriched EVs could provide a unique opportunities in CNS diseases therapy, and discussed their clinical potential, neuro restoration could be a viable treatment strategy.
MSC Derived EVs
MSC
MSCs are usually obtained from adult bone marrow, peripheral blood, adipose tissue, and placenta, bone marrow is the most frequently used source [1,2]. They can promote tissue repair and remodeling through and secretion of cytokines [3-8], which has become a promising alternative strategy in treatment of CNS diseases [9]. However, the application of MSCs in clinical has some drawbacks, such as unstable phenotype, high costs of generation and processing, ectopic tissue formation [10]. In addition, lodged in the pulmonary microvasculature and caused infusion toxicity [11,12], and cellular rejection or unwanted implantation also have been reported recently [13]. In recent years, some scholars have found that MSCs affects tissue repair rather than cell replacement by stimulating tissue cells through paracrine factors [14]. The main aspect of MSCs response to the disease is the secretion of differ functional molecules stored in EVs and play a significant role in cell-to-cell communication, which generate important actions in development, regeneration, angiogenesis, homeostasis, et al. [15-19].
EVs
EVs comprise a large variety of membranous structures released from almost all types of cells. The accumulated historical data and recent research have indicated that the contents, EVs are heterogeneous and dynamic existed in EV’s size, content, membrane composition and function, according to their cellular source, physiological status, and most importantly environmental conditions. Increasing research on EVs has increased understanding of their variety and complexity. EVs are lipid bilayer and comprise a heterogeneous population of membrane vesicles, by fusion of multivesicular bodies and the plasma membrane or formed from the direct budding or microvesicles. At present, three main subgroups of EVs have been defined by The International Society of Extracellular Vesicles, and they can be broadly classified based on size or origin: microvesicles (MVs, and exosomes) and apoptotic bodies [20,21]. MVs, which are typically larger in size (ranging from 100 to 1,000 nm) and are formed as the result of the outward budding of the plasma membrane. Exosomes generally representing smallest EVs of 150nm or less. Exosomes generally representing smallest EVs of 150nm or less, and derived from early endosomes by invagination of the recruited membrane, inward budding, and scission. Apoptotic bodies characterized by a dimension ranging from 1,000 to 5,000 nm, and that released as blebs from cells undergoing programmed death cell [22-24]. EVs contain various specific molecules, such as, DNA, miRNA, mRNA, long noncoding RNA, lipids, proteins, and genetic materials from viruses or prions, depending on the different cellular origin and the function of putative target [25,26]. The best characterized of EVs were firstly described in the early ‘80s [27]. Later, a small vesicle was founded in reticulocytes, It has a circular or concave cup shape under the electron microscope with a lipid bilayer structure, small vesicles fuse with the plasma membrane and release their contents to the outside of the cell in the process of exocytosis [28]. This was coined for 40- 100 nm vesicles released during reticulocyte differentiation by fusion of multivesicular endosomes (MVEs) with the plasma membrane [29,30]. To unify the nomenclature throughout, we will, therefore, use the term EVs for all types of vesicles in this review.
In the past 20 years, EVs were demonstrated that be produced and released by B lymphocytes and dendritic cells through differential centrifugation and described the role of EVs in antigen presentation in vivo and able to induce T cell responses [31,32]. Recently research found that all cell types are able to secrete EVs, including mesenchymal stem cell (MSC) [33], hematopoietic stem cell [34], cardiac progenitor cells [35], embryonic stem cell [36], pancreatic cancer cell [37] and liver cancer cell [38] et al. EVs are also found in physiological and pathological fluids, including pleural effusions, plasma [39], ocular effluent and aqueous humor [40], breast milk [41], broncho-alveolar lavage [42], synovial fluid [43], bile [44], urine [45,46] and sputum [46], ascites [47], amniotic fluid [48], semen (“prostasomes” and “epididymosomes”) [49-51], nasal secretions [52], CSF [53].
EVs have recently gained much attention for their application to CNS diseases, EVs participate in the regulation of normal physiological processes and disease pathology, not only including tissue homeostasis, such as stem cell maintenance, development, repair, regeneration, as well as pathophysiology [54-57], but also modulate immune system and protect apoptosis via multiple pathways [58,59]. In addition, EVS plays a significant role in cancer and cardiovascular disease [23]. Increasing interest in the development of EVs (Figure 1). Its therapeutic effect for central nervous system diseases have been used in pre-clinical and clinical settings over the last decade, administration of MSCs-EVs has beneficial effects in numerous animal models of CNS diseases, including stroke, intracerebral hemorrhage (ICH), traumatic brain injury (TBI), glioblastoma (GBM), spinal cord injury (SCI) et al. The therapeutic effect of EVs in CNS diseases can be ascribed to the modulation of variety processes, including angiogenesis, neurogenesis, apoptosis, immune response, and reprogram in physiological and pathological status [60]. Compare with MSCs: EVs are smaller diameter and less complex content, so they are easier to produce and store, and will be more potential to address contentious regulatory issues [61]. Interestingly, purified EVs from MSCs exerted most, the key mechanism by which MSC contribute to tissue repair and regeneration is through their paracrine function, EVs are one of the major factors that are secreted [62-64]. Other studies have also showed that the treatment effects of MSCs are mostly attributed to cell-secreted paracrine factors rather than target and direct action of transplanted cells [65-67]. Moreover, paracrine factors can induce revascularization and promote proliferation of tissue cells [68,69].
Figure 1: Publication in exosome research annually since 2010 retrieved from a PubMed search of the term “exosome”, “exosome + RNA”, “exosome + Lipid”, “exosome + Proteomic”.
EV components
The database was developed based on published articles, such as, EVpedia (http://evpedia.info) and ExoCarta (http://exocarta. org) storing millions of the proteins, mRNAs, miRNAs, and lipids of mammalian EVs.
Nucleic acid content: Walking through the historical discovery of EVs mRNAs and miRNAs in humans and mice, 4,946 mRNA entries and 2,838 miRNAs from EVs from multi cells and body fluids have been identified (EVpedia, ExoCarta) [70,71]. Highly abundant small RNAs constituent of wide range of genetic materials in EVs, and many of them derived from ribosomal 18S and 28S rRNAs, tRNAs. By next-generation sequencing, diverse composition of small RNAs have been identified, not only contain commonly known RNA species, such as mRNAs, miRNAs, rRNAs, tRNA, vault RNA, long and short non-coding RNA, piwi-interacting RNA, and Y RNA also have been characterized [20,72-76]. Most of the RNA is generally shorter in size, around 200 nucleotides and smaller portion extending out to 4kb [77]. Interestingly, different types of RNA species can also be stably together with functional ribonucleoprotein (RNP) and depending on the isolation procedure, for example, high and lowdensity lipoproteins (HDLs and LDLs) participate in a mechanism of cell-sell communication involving the transport of miRNAs and enables their stability and delivery throughout the body, argonaute 2 (AGO2), which are essential to miRNA-induced silencing [78,79]. Multiple reports have implicated EVs serve as carriers of genetic information in cell-cell communication by carrying cell-specific mRNAs and miRNAs, especially in the treatment of CNS diseases. In some cases, genomic and mitochondrial DNA has been found, however, the therapeutic effects of not yet documented [80,81].
Protein content: Proteomic profiling of the EVs underwent a significant development in recent years, 41,860 protein entries from EVs have been identified, including annexin (Annexins I, II, IV, V and VII), cytoskeletal components (actin, alanine, tubulin, coenzyme), small GTPase family members Rab7 and Rab11, and EVS marker proteins Alix, TSG101, CD9, CD63 [82,83]. Alix, Syntenin-1, integrins, tetraspanins, cytoskeletal proteins, Tsg101, HSPs, annexins, Rab proteins, metabolic enzymes, and ribosomal proteins were categorized as common EVs protein [84,85]. As more EVs proteomes are identified, the inclusion or exclusion of cellular protein packaging is governed by protein-sorting mechanism during production of EVs [86].
During the last 20 years, extensive research has investigated in the protein composition of MSC-EVs (MSC derived EVs) by various proteomic analysis, including immunoelectron microscopy, fluorescence-activated cell sorting and Western blotting, there have been over 900 proteins identified in MSC-EVs [82,87]. Recent studies have shown that MSC-EVs can improve the recovery of damaged tissues [84], and identified 730 proteins by LC coupled MS/MS analysis, 43 surface receptors as well as signaling molecules are characterized, which is responsible for self-renewal and differentiation. Some other analysis indicated that MSCs-EVs proteins contributed to cell proliferation, migration, adhesion, and morphogenesis [88,89]. Self-renewal ability and differentiation potentiality of MSCs can be associated with the therapeutic effects, which is regulated by the integration of related genes, including signaling molecules (CDC42, VAV2); surface receptors (PDGFRB, EGFR, and PLAUR); antigens (CD109, CD248, CD151 and CD276) [84].
Lipid content: In addition to the abundance of proteins and nucleic acid within EVs, similarly, the lipid composition has also been investigated quite extensively [90-92]. A total of 1116 lipids have been identified (http://exocarta.org) in various settings. While there does not appear to similar lipid composition for EVs derived from different cells types, all of EVs lipid bilayer mainly contains the components plasma membrane lipids (sphingomyelin, phospholipids, ganglioside GM3, and cholesterol) [93,94]. Among them, glycerophospholipids accounted for 91.5% of the total lipid ion abundance, followed by sphingolipids (5.3%), sterols (1.9%) and glycerol (1.4%) [95]. Glycerol phospholipids mainly include phosphatidylserine and phosphatidylglycerol. The former is a recruiter of negative charge activators and signaling proteins [96], and the latter involved in transmembrane transport [97,98]; sphingomyelin is related to the construction of outer membrane [99]; sterols are associated with vesicles and plasma membranes [100].
MSC-EVs-miRNA in CNS Disease Therapy
Accumulating evidence reveals that modification of exosomal miRNAs content can be a promising tool in the development of CNS diseases therapeutic. In addition, exosomes can penetrate the blood-brain barrier to enhance the therapeutic effect of miRNAs. Evidence from extensive studies in the last decade has indicated that exosomes from MSCs carrying miRNAs were found to be effective against several CNS disease targets and were reported to enhance chemosensitivity while suppress angiogenesis. Moreover, Exosomes express miRNAs regulate the expression of related genes in receptor cells and promote the regeneration and repair of receptor cells. Exosomal miRNAs currently studied in CNS diseases mainly include miRNA-126, miRNA-21, miRNA-17-92, miRNA-133b, and miRNA-124 (Figure 2).
Figure 2: Schematic representation of possible routes of MSC-EVs-miRNA to improve CNS diseases.
Stroke
Compared to native MSC, MSC-EVs-miR-17-92 cluster can further enhanced axonal growth in stroke mice, which activated the PTEN/mTOR signaling pathway [101]. Similarly, Xin et al, suggest that MSC-EVs-miR-17-92 cluster also increased neural plasticity and improved functional recovery via the PI3K/Akt/mTOR/GSK-3β signaling pathway [102]. In another study, miR-21, which increased the expression in MSC-derived EVs, improved learning and memory capabilities via the PTEN/Akt pathway. Moreover, EVs have been shown to increase miR-21 level in AD mice, which restored the cognitive deficits in APP/PS1 mic [103]. Additionally, c(RGDyK)- conjugated EVs (cRGD-Exo) loaded with cholesterol-modified miR- 210 (RGD-exo-miR-210) targets the ischemic brain contributing to miR-210 increase in the lesion region, which increased vascular endothelial growth factor (VEGF) and CD34 and shown neural protection in middle cerebral artery occlusion (MCAO) mouse [104]. What’s more, obtained evidence indicated that, overexpression of miR-138-5p was observed to promotes the proliferation and migration of astrocytes injured because of hypoxia and glucose deprivation, while BMSCs-EVs-miR138-5p promote proliferation which was achieved by inhibiting the apoptosis of astrocytes via downregulate LCN2 [105]. After oxygen-glucose deprivation (OGD) treatment, BMSCs-derived miR-134 EVs suppressed oligodendrocytes (OLs) apoptosis through a caspase-8-dependent apoptosis pathway [106].
Traumatic brain injury (TBI)
Some studies demonstrated for the first time that EVs from MSCs improve nerve functional recovery, stimulate angiogenesis, and promote neurogenesis in traumatic brain injury (TBI) [107]. In the latter, MSCs-EVs have been found to reduce neurological impairment in a transient intraluminal MCAO. MSCs-EVs -miR133b could regulates the expression of connective tissue growth factor (CTGF), a major inhibitor of axonal growth at injury sites, in astrocytes and the Ras homolog gene family member A (RhoA) expression in the IBZ, it also increased the expression of von Willebrand factor (vWF) in stroke rat, which demonstrated that MSCs communicate with brain parenchymal cells by transfer miR-133b to neural cells via EVs [108,109]. EVs from MSCs cultured in 3D conditions showed better outcome than 2D, EVs derived from 3D scaffolds provided better spatial learning, hMSC-generated EVs promoted endogenous angiogenesis, neurogenesis, and improved functional recovery in rats after TBI [110]. Moreover, EVs-miR124 could play a crucial role in the microglial polarization, and move it towards to antiinflammatory phenotype (M2) state by inhibiting TLR4 pathway, thus improving neurogenesis and enhancing functional recovery in the hippocampus [111]. Recently research demonstrated that, MSCs- EVs-miR-17-92 improved functional recovery after TBI by reducing neuroinflammation and enhancing endogenous angiogenesis and neurogenesis, providing a novel therapeutic strategy for TBI.
Spinal cord injury (SCI)
A significant recovery of hindlimb function was observed in spinal cord injury (SCI) rats after MSCs-EVs-miR133b administration. Additionally, MSCs-EVs-miR133b promoted the regeneration of axons, protected neuronal cells, and reduced the volume of the lesion, which targeted RhoA and decreased the expression. Moreover, it also activated signaling pathway proteins, which involved in the survival of neurons and the regeneration of axons, such as ERK1/2, STAT3, and CREB, could be an effective strategy [112]. Other research reported that, hMSCs-EVs-miR-21 and miR-19b regulated PTEN mRNA and protein as well as the value of cell apoptosis index, involved in the apoptosis and differentiation of neuron cells in SCI [113]. Further studies reveal that, EVs with miR-21 deficiency would not exert protective effects against SCI [114]. Interestingly, miR-21- 5p was up-regulated after SCI, motor function and apoptosis were significantly increased in spinal cord post-SCI after miR-21-5p in MSCs-EVs transplantation therapy, an effect which was associated with increased the proportion of TUNEL-positive cells [115]. In vitro analysis indicated that, MSCs-EVs-miR-126, a key regulator, promoted the angiogenesis and migration of human umbilical venous endothelial cells (HUVECs), through inhibiting the expression of SPRED1 and PIK3R2 [116].
Intracerebral hemorrhage (ICH)
Interestingly, intracerebral hemorrhage (ICH) could induce an increase of EVs level in the brain, inhibition of EVs release augmented neurological deficits and brain edema [117]. Proteomics analysis of the EVs identified 2416 proteins that have been implicated in regulating number of cell functions, EVs derived from MSC therapy work in ICH as paracrine effectors responsible for brain repair processes, improved functional recovery, more axons, a higher expression of oligodendrocyte formation markers [118]. Regarding functional recovery, previous studies demonstrated that miR-21 is downregulated in blood and brain tissue after ICH, MSCs-EVsmiR- 21 can be transported to neurons and that it plays a crucial role in alleviate neuronal injury via targeting transient receptor potential melastatin 7 (TRPM7) [119]. Recent evidence indicated that improvement in functional outcome after miR-146a-5priched BMSCs-EVs administration following ICH, which could be associated with the inhibition of M1/M2 polarization transitions via downregulating the expression level of IRAK1 and NFAT5 [120]. In addition, it was also suggested that the expression of miR-133b levels were negatively correlated with RhoA expression and that the effect of MSCs-EVs-miR-133b play a significant role in neuroprotection via targeting RhoA and activating ERK1/2 [121].
Glioblastomas (GBM)
MSC-EVs are associated with mixed effects on tumor initiation and progression, being able either to favor angiogenesis and tumor initiation, or to inhibit metabolic signaling and progression of established tumors. These biological rationale for this response is likely attributable to their microenvironment. MSC-EVs, as the pivotal mediators of communication in the tumor microenvironment, play essential roles in cell-to-cell communication, and suppress angiogenesis by directly transferring anti-angiogenic molecules, MSC-EVs-miR-16 suppressed the expression of VEGF, to lead to the inhibition of angiogenesis and tumor progression [122]. Other research found that MSC-EVs-miR146b effectively reduced glioma xenograft growth in rat brain, it is likely that inhibition of factors EGFR and NF-κB protein in glioma cells underpins the anti-tumor [123]. When administered systemically, EVs-miR [124] is capable of downregulating FOXA2 and induced apoptotic cell death, which may correlate with FOXA2-mediated aberrant intracellular lipid accumulation [124]. In addition, EVs-miR9 was linked to temozolomide resistance in glioblastoma, when transfer this antimiRNA into co-cultured- GBM cells, it was be able to affect GBM cells at a considerable distance [125]. MSC-EVs- miR199a enhanced the chemosensitivity to temozolomide and inhibited the tumor growth, besides, it suppressed the proliferation, invasion, as well as migration of glioma cells by down-regulating the expression of AGAP2 [126]. In a separate study, further research showed that the known targets of the miR-302-367 cell-to-cell transfer resulted in the inhibition of cyclin A, SHH, cyclin Dand, E2F1 and CXCR4/SDF1, and prevented growth of GBM stem-like or progenitor cells [127]. Some studies have suggested that MSC-EVs promote tumorigenesis and metastasis, for example, it promoted the metastasis of the breast cancer, human renal cell carcinoma and bladder cancer [128-130].
Discussion
EVs, vectors of biological information, are involved in cell-cell communication in both physiological and pathological processes by delivering miRNAs to recipient cells that transferring beneficial mediators in CNS diseases. MSCs-EVs target housekeeping processes by their secreted EVs containing different miRNA to promote angiogenesis and neurogenesis, protect neurons, suppresses t neuron apoptosis, and increase neural plasticity. MSCs-EVs can also inhibit tumor proliferation, migration, and invasion, as well as enhance the chemosensitivity (Figure 2). The MSCs-EVs therapy, easier to cross the blood-brain barrier and avoiding the risk of iatrogenic tumor formation as well as intravenous administration-induced pulmonary embolisms, is a promising alternative to overcome the obstacles of cell-therapy. Despite numerous studies and literature reports, challenges are remaining for clinical application of MSCs-EVs-based therapy. Further studies of the cellular and molecular mechanisms of MSCs-EVs are necessary to maximize its clinical benefits in CNS diseases. Development and standardization of technologies in the manufacture, detection, and characterization of MSC-EVs are also indispensable for its clinical application. In conclusion, once these critical issues around MSC-EVs are settled, MSC-derived EVs can be harnessed as powerful therapeutic agents in CNS diseases.
Funding
Zhiqian Yang received Medical Science and Technology Foundation of Guangdong Province (A2020419). Wei Zhang supported by Department of Education of Guangdong Province (2018KCXT017). Yesheng Sun supported by “Climbing” Program of Guangdong Province (pdjh2021b0261).
References
- Murphy MB, Moncivais K, Caplan AI. Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp Mol Med. 2013; 45: e54.
- Riazifar M, Pone EJ, Lotvall J, Zhao W. Stem Cell Extracellular Vesicles: Extended Messages of Regeneration. Annu Rev Pharmacol Toxicol. 2017; 57: 125-154.
- Glennie S, Soeiro I, Dyson PJ, Lam EW, Dazzi F. Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells. Blood. 2005; 105: 2821-2827.
- da Silva Meirelles L, Caplan AI, Nardi NB. In search of the in vivo identity of mesenchymal stem cells. Stem Cells. 2008; 26: 2287-2299.
- Singer NG, Caplan AI. Mesenchymal stem cells: mechanisms of inflammation. Annu Rev Pathol. 2011; 6: 457-478.
- Li P, Gong Z, Shultz LD, Ren G. Mesenchymal stem cells: From regeneration to cancer. Pharmacol Ther. 2019; 200: 42-54.
- Manieri NA, Mack MR, Himmelrich MD, et al. Mucosally transplanted mesenchymal stem cells stimulate intestinal healing by promoting angiogenesis. J Clin Invest. 2015; 125: 3606-3618.
- D’Souza N, Rossignoli F, Golinelli G, et al. Mesenchymal stem/stromal cells as a delivery platform in cell and gene therapies. BMC Med. 2015; 13: 186.
- Sadan O, Melamed E, Offen D. Bone-marrow-derived mesenchymal stem cell therapy for neurodegenerative diseases. Expert Opin Biol Ther. 2009; 9: 1487-1497.
- AlMuraikhi N, Ali D, Vishnubalaji R, et al. Notch Signaling Inhibition by LY411575 Attenuates Osteoblast Differentiation and Decreased Ectopic Bone Formation Capacity of Human Skeletal (Mesenchymal) Stem Cells. Stem Cells Int. 2019; 2019: 3041262.
- Gonsalves A, Carrier M, Wells PS, McDiarmid SA, Huebsch LB, Allan DS. Incidence of symptomatic venous thromboembolism following hematopoietic stem cell transplantation. J Thromb Haemost. 2008; 6: 1468-1473.
- Kekre N, Kim HT, Ho VT, et al. Venous thromboembolism is associated with graft-versus-host disease and increased non-relapse mortality after allogeneic hematopoietic stem cell transplantation. Haematologica. 2017; 102: 1185-1191.
- Maffini E, Festuccia M, Brunello L, Boccadoro M, Giaccone L, Bruno B. Neurologic Complications after Allogeneic Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant. 2017; 23: 388-397.
- Nagaishi K, Mizue Y, Chikenji T, et al. Mesenchymal stem cell therapy ameliorates diabetic nephropathy via the paracrine effect of renal trophic factors including exosomes. Sci Rep. 2016; 6: 34842.
- Caccia D, Dugo M, Callari M, Bongarzone I. Bioinformatics tools for secretome analysis. Biochim Biophys Acta. 2013; 1834: 2442-2453.
- Golpanian S, Wolf A, Hatzistergos KE, Hare JM. Rebuilding the Damaged Heart: Mesenchymal Stem Cells, Cell-Based Therapy, and Engineered Heart Tissue. Physiol Rev. 2016; 96: 1127-1168.
- Madrigal M, Rao KS, Riordan NH. A review of therapeutic effects of mesenchymal stem cell secretions and induction of secretory modification by different culture methods. J Transl Med. 2014; 12: 260.
- Maumus M, Jorgensen C, Noel D. Mesenchymal stem cells in regenerative medicine applied to rheumatic diseases: role of secretome and exosomes. Biochimie. 2013; 95: 2229-2234.
- Paul G, Anisimov SV. The secretome of mesenchymal stem cells: potential implications for neuroregeneration. Biochimie. 2013; 95: 2246-2256.
- Crescitelli R, Lasser C, Szabo TG, et al. Distinct RNA profiles in subpopulations of extracellular vesicles: apoptotic bodies, microvesicles and exosomes. J Extracell Vesicles. 2013; 2.
- Gill S, Catchpole R, Forterre P. Extracellular membrane vesicles in the three domains of life and beyond. FEMS Microbiol Rev. 2019; 43: 273-303.
- Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013; 200: 373-383.
- Yanez-Mo M, Siljander PR, Andreu Z, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015; 4: 27066.
- Ban LA, Shackel NA, McLennan SV. Extracellular Vesicles: A New Frontier in Biomarker Discovery for Non-Alcoholic Fatty Liver Disease. Int J Mol Sci. 2016; 17: 376.
- Maas SLN, Breakefield XO, Weaver AM. Extracellular Vesicles: Unique Intercellular Delivery Vehicles. Trends Cell Biol. 2017; 27: 172-188.
- Balusu S, Van Wonterghem E, De Rycke R, et al. Identification of a novel mechanism of blood-brain communication during peripheral inflammation via choroid plexus-derived extracellular vesicles. EMBO Mol Med. 2016; 8: 1162-1183.
- Trams EG, Lauter CJ, Salem N Jr., Heine U. Exfoliation of membrane ectoenzymes in the form of micro-vesicles. Biochim Biophys Acta. 1981; 645: 63-70.
- Pan BT, Johnstone RM. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor. Cell. 1983; 33: 967-978.
- Harding C, Heuser J, Stahl P. Endocytosis and intracellular processing of transferrin and colloidal gold-transferrin in rat reticulocytes: demonstration of a pathway for receptor shedding. Eur J Cell Biol. 1984; 35: 256-263.
- Pan BT, Teng K, Wu C, Adam M, Johnstone RM. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J Cell Biol. 1985; 101: 942-948.
- Raposo G, Nijman HW, Stoorvogel W, et al. B lymphocytes secrete antigenpresenting vesicles. J Exp Med. 1996; 183: 1161-1172.
- Zitvogel L, Regnault A, Lozier A, et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nature medicine. 1998; 4: 594-600.
- Kang J, Li Z, Zhi Z, Wang S, Xu G. MiR-21 derived from the exosomes of MSCs regulates the death and differentiation of neurons in patients with spinal cord injury. Gene Ther. 2019; 26.
- Liao FL, Tan L, Liu H, et al. Hematopoietic stem cell-derived exosomes promote hematopoietic differentiation of mouse embryonic stem cells in vitro via inhibiting the miR126/Notch1 pathway. Acta Pharmacol Sin. 2018; 39: 552-560.
- Li X, Yang Z, Nie W, et al. Exosomes derived from cardiac progenitor cells attenuate CVB3-induced apoptosis via abrogating the proliferation of CVB3 and modulating the mTOR signaling pathways. Cell Death Dis. 2019; 10: 691.
- Tavakoli Dargani Z, Singla DK. Embryonic stem cell-derived exosomes inhibit doxorubicin-induced TLR4-NLRP3-mediated cell death-pyroptosis. Am J Physiol Heart Circ Physiol. 2019; 317: H460-H471.
- Chiba M, Kubota S, Sato K, Monzen S. Exosomes released from pancreatic cancer cells enhance angiogenic activities via dynamin-dependent endocytosis in endothelial cells in vitro. Sci Rep. 2018; 8: 11972.
- Wang H, Lu Z, Zhao X. Tumorigenesis, diagnosis, and therapeutic potential of exosomes in liver cancer. J Hematol Oncol. 2019; 12: 133.
- Ibsen SD, Wright J, Lewis JM, et al. Rapid Isolation and Detection of Exosomes and Associated Biomarkers from Plasma. ACS Nano. 2017; 11: 6641-6651.
- Perkumas KM, Hoffman EA, McKay BS, Allingham RR, Stamer WD. Myocilin-associated exosomes in human ocular samples. Experimental eye research. 2007; 84: 209-212.
- Kim K-U, Kim W-H, Jeong CH, Yi DY, Min H. More than Nutrition: Therapeutic Potential of Breast Milk-Derived Exosomes in Cancer. International journal of molecular sciences. 2020; 21.
- Carnino JM, 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.
- Qu X, Li Q, Yang J, et al. Double-Stranded DNA in Exosomes of Malignant Pleural Effusions as a Novel DNA Source for EGFR Mutation Detection in Lung Adenocarcinoma. Frontiers in oncology. 2019; 9: 931.
- Yao Y, Jiao D, Li Z, et al. Roles of Bile-Derived Exosomes in Hepatobiliary Disease. Biomed Res Int. 2021; 2021: 8743409.
- Merchant ML, Rood IM, Deegens JKJ, Klein JB. Isolation and characterization of urinary extracellular vesicles: implications for biomarker discovery. Nat Rev Nephrol. 2017; 13: 731-749.
- Musante L, Tataruch-Weinert D, Kerjaschki D, Henry M, Meleady P, Holthofer H. Residual urinary extracellular vesicles in ultracentrifugation supernatants after hydrostatic filtration dialysis enrichment. J Extracell Vesicles. 2017; 6: 1267896.
- Hu Y, Qi C, Liu X, et al. Malignant ascites-derived exosomes promote peritoneal tumor cell dissemination and reveal a distinct miRNA signature in advanced gastric cancer. Cancer letters. 2019; 457: 142-150.
- Zavatti M, Beretti F, Casciaro F, Bertucci E, Maraldi T. Comparison of the therapeutic effect of amniotic fluid stem cells and their exosomes on monoiodoacetate-induced animal model of osteoarthritis. Biofactors. 2020; 46: 106-117.
- Asea A, Jean-Pierre C, Kaur P, et al. Heat shock protein-containing exosomes in mid-trimester amniotic fluids. Journal of reproductive immunology. 2008; 79: 12-17.
- Poliakov A, Spilman M, Dokland T, Amling CL, Mobley JA. Structural heterogeneity and protein composition of exosome-like vesicles (prostasomes) in human semen. The Prostate. 2009; 69: 159-167.
- Ronquist G, Brody I. The prostasome: its secretion and function in man. Biochim Biophys Acta. 1985; 822: 203-218.
- Lasser C, O’Neil SE, Ekerljung L, Ekstrom K, Sjostrand M, Lotvall J. RNAcontaining exosomes in human nasal secretions. American journal of rhinology & allergy. 2011; 25: 89-93.
- Street JM, Barran PE, Mackay CL, et al. Identification and proteomic profiling of exosomes in human cerebrospinal fluid. J Transl Med. 2012; 10: 5.
- 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.
- Andrzejewska A, Lukomska B, Janowski M. Concise Review: Mesenchymal Stem Cells: From Roots to Boost. Stem Cells. 2019; 37: 855-864.
- Prada I, Gabrielli M, Turola E, et al. Glia-to-neuron transfer of miRNAs via extracellular vesicles: a new mechanism underlying inflammation-induced synaptic alterations. Acta Neuropathol. 2018; 135: 529-550.
- Zappulli V, Friis KP, Fitzpatrick Z, Maguire CA, Breakefield XO. Extracellular vesicles and intercellular communication within the nervous system. J Clin Invest. 2016; 126: 1198-1207.
- Greening DW, Gopal SK, Xu R, Simpson RJ, Chen W. Exosomes and their roles in immune regulation and cancer. Semin Cell Dev Biol. 2015; 40: 72- 81.
- Zhang HG, Grizzle WE. Exosomes and cancer: a newly described pathway of immune suppression. Clin Cancer Res. 2011; 17: 959-964.
- Chen J, Chopp M. Exosome Therapy for Stroke. Stroke. 2018; 49: 1083- 1090.
- Katsuda T, Kosaka N, Takeshita F, Ochiya T. The therapeutic potential of mesenchymal stem cell-derived extracellular vesicles. Proteomics. 2013; 13: 1637-1653.
- Shi M, Sheng L, Stewart T, Zabetian CP, Zhang J. New windows into the brain: Central nervous system-derived extracellular vesicles in blood. Prog Neurobiol. 2019; 175: 96-106.
- Robbins PD, Dorronsoro A, Booker CN. Regulation of chronic inflammatory and immune processes by extracellular vesicles. J Clin Invest. 2016; 126: 1173-1180.
- Kang J, Li Z, Zhi Z, Wang S, Xu G. MiR-21 derived from the exosomes of MSCs regulates the death and differentiation of neurons in patients with spinal cord injury. Gene Therapy. 2019.
- Szabo G, Momen-Heravi F. Extracellular vesicles in liver disease and potential as biomarkers and therapeutic targets. Nat Rev Gastroenterol Hepatol. 2017; 14: 455-466.
- Momen-Heravi F, Balaj L, Alian S, et al. Current methods for the isolation of extracellular vesicles. Biol Chem. 2013; 394: 1253-1262.
- Montecalvo A, Larregina AT, Shufesky WJ, et al. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood. 2012; 119: 756-766.
- Bruno S, Grange C, Deregibus MC, et al. Mesenchymal stem cell-derived microvesicles protect against acute tubular injury. J Am Soc Nephrol. 2009; 20: 1053-1067.
- You Y, Ikezu T. Emerging roles of extracellular vesicles in neurodegenerative disorders. Neurobiol Dis. 2019; 130: 104512.
- Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosomemediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature cell biology. 2007; 9: 654-659.
- Skog J, Wurdinger T, van Rijn S, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nature cell biology. 2008; 10: 1470-1476.
- Cheng L, Sun X, Scicluna BJ, Coleman BM, Hill AF. Characterization and deep sequencing analysis of exosomal and non-exosomal miRNA in human urine. Kidney international. 2014; 86: 433-444.
- Li CC, Eaton SA, Young PE, et al. Glioma microvesicles carry selectively packaged coding and non-coding RNAs which alter gene expression in recipient cells. RNA biology. 2013; 10: 1333-1344.
- Liu Y-M, Tseng C-H, Chen Y-C, et al. Exosome-delivered and Y RNAderived small RNA suppresses influenza virus replication. J Biomed Sci. 2019; 26: 58.
- Dai J, Su Y, Zhong S, et al. Exosomes: key players in cancer and potential therapeutic strategy. Signal Transduct Target Ther. 2020; 5: 145.
- Jeppesen DK, Fenix AM, Franklin JL, et al. Reassessment of Exosome Composition. Cell. 2019; 177.
- Batagov AO, Kurochkin IV. Exosomes secreted by human cells transport largely mRNA fragments that are enriched in the 3’-untranslated regions. Biology direct. 2013; 8: 12.
- Arroyo JD, Chevillet JR, Kroh EM, et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci USA. 2011; 108: 5003-5008.
- Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nature cell biology. 2011; 13: 423-433.
- Balaj L, Lessard R, Dai L, et al. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nat Commun. 2011; 2: 180.
- Guo Y, Ji X, Liu J, et al. Effects of exosomes on pre-metastatic niche formation in tumors. Mol Cancer. 2019; 18: 39.
- Keerthikumar S, Chisanga D, Ariyaratne D, et al. ExoCarta: A Web-Based Compendium of Exosomal Cargo. Journal of molecular biology. 2016; 428: 688-692.
- Gao M, Gao W, Papadimitriou JM, Zhang C, Gao J, Zheng M. Exosomes-the enigmatic regulators of bone homeostasis. Bone Res. 2018; 6: 36.
- Kim HS, Choi DY, Yun SJ, et al. Proteomic analysis of microvesicles derived from human mesenchymal stem cells. J Proteome Res. 2012; 11: 839-849.
- Choi DS, Kim DK, Kim YK, Gho YS. Proteomics, transcriptomics and lipidomics of exosomes and ectosomes. Proteomics. 2013; 13: 1554-1571.
- Raimondo F, Morosi L, Chinello C, Magni F, Pitto M. Advances in membranous vesicle and exosome proteomics improving biological understanding and biomarker discovery. Proteomics. 2011; 11: 709-720.
- Kalra H, Simpson RJ, Ji H, et al. Vesiclepedia: a compendium for extracellular vesicles with continuous community annotation. PLoS biology. 2012; 10: e1001450.
- da Costa Goncalves F, Paz AH. Cell membrane and bioactive factors derived from mesenchymal stromal cells: Cell-free based therapy for inflammatory bowel diseases. World J Stem Cells. 2019; 11: 618-633.
- Todorova D, Simoncini S, Lacroix R, Sabatier F, Dignat-George F. Extracellular Vesicles in Angiogenesis. Circ Res. 2017; 120: 1658-1673.
- Van Blitterswijk WJ, De Veer G, Krol JH, Emmelot P. Comparative lipid analysis of purified plasma membranes and shed extracellular membrane vesicles from normal murine thymocytes and leukemic GRSL cells. Biochim Biophys Acta. 1982; 688: 495-504.
- Carayon K, Chaoui K, Ronzier E, et al. Proteolipidic composition of exosomes changes during reticulocyte maturation. J Biol Chem. 2011; 286: 34426-34439.
- Llorente A, Skotland T, Sylvanne T, et al. Molecular lipidomics of exosomes released by PC-3 prostate cancer cells. Biochim Biophys Acta. 2013; 1831: 1302-1309.
- Gurunathan S, Kang MH, Jeyaraj M, Qasim M, Kim JH. Review of the Isolation, Characterization, Biological Function, and Multifarious Therapeutic Approaches of Exosomes. Cells. 2019; 8.
- Haraszti RA, Didiot MC, Sapp E, et al. High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources. J Extracell Vesicles. 2016; 5: 32570.
- Lydic TA, Townsend S, Adda CG, Collins C, Mathivanan S, Reid GE. Rapid and comprehensive ‘shotgun’ lipidome profiling of colorectal cancer cell derived exosomes. Methods. 2015; 87: 83-95.
- Moser von Filseck J, Copic A, Delfosse V, et al. INTRACELLULAR TRANSPORT. Phosphatidylserine transport by ORP/Osh proteins is driven by phosphatidylinositol 4-phosphate. Science. 2015; 349: 432-436.
- Record M, Carayon K, Poirot M, Silvente-Poirot S. Exosomes as new vesicular lipid transporters involved in cell-cell communication and various pathophysiologies. Biochim Biophys Acta. 2014; 1841: 108-120.
- Chung J, Torta F, Masai K, et al. INTRACELLULAR TRANSPORT. PI4P/ phosphatidylserine countertransport at ORP5- and ORP8-mediated ERplasma membrane contacts. Science. 2015; 349: 428-432.
- Neumann EK, Comi TJ, Rubakhin SS, Sweedler JV. Lipid Heterogeneity between Astrocytes and Neurons Revealed by Single-Cell MALDI-MS Combined with Immunocytochemical Classification. Angew Chem Int Ed Engl. 2019; 58: 5910-5914.
- Valastyan JS, Termine DJ, Lindquist S. Splice isoform and pharmacological studies reveal that sterol depletion relocalizes alpha-synuclein and enhances its toxicity. Proc Natl Acad Sci U S A. 2014; 111: 3014-3019.
- Zhang Y, Chopp M, Liu XS, et al. Exosomes Derived from Mesenchymal Stromal Cells Promote Axonal Growth of Cortical Neurons. Mol Neurobiol. 2017; 54: 2659-2673.
- Xin H, Katakowski M, Wang F, et al. MicroRNA cluster miR-17-92 Cluster in Exosomes Enhance Neuroplasticity and Functional Recovery after Stroke in Rats. Stroke. 2017; 48: 747-753.
- Cui GH, Wu J, Mou FF, et al. Exosomes derived from hypoxia-preconditioned mesenchymal stromal cells ameliorate cognitive decline by rescuing synaptic dysfunction and regulating inflammatory responses in APP/PS1 mice. FASEB J. 2018; 32: 654-668.
- Zhang H, Wu J, Wu J, et al. Exosome-mediated targeted delivery of miR- 210 for angiogenic therapy after cerebral ischemia in mice. Journal of nanobiotechnology. 2019; 17: 29.
- Deng Y, Chen D, Gao F, et al. Exosomes derived from microRNA-138- 5p-overexpressing bone marrow-derived mesenchymal stem cells confer neuroprotection to astrocytes following ischemic stroke via inhibition of LCN2. Journal of biological engineering. 2019; 13: 71.
- Xiao Y, Geng F, Wang G, Li X, Zhu J, Zhu W. Bone marrow-derived mesenchymal stem cells-derived exosomes prevent oligodendrocyte apoptosis through exosomal miR-134 by targeting caspase-8. J Cell Biochem. 2018.
- Zhang Y, Chopp M, Meng Y, et al. Effect of exosomes derived from multipluripotent mesenchymal stromal cells on functional recovery and neurovascular plasticity in rats after traumatic brain injury. J Neurosurg. 2015; 122: 856-867.
- Xin H, Li Y, Liu Z, et al. MiR-133b promotes neural plasticity and functional recovery after treatment of stroke with multipotent mesenchymal stromal cells in rats via transfer of exosome-enriched extracellular particles. Stem Cells. 2013; 31: 2737-2746.
- Xin H, Li Y, Buller B, et al. Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells. 2012; 30: 1556-1564.
- Zhang Y, Chopp M, Zhang ZG, et al. Systemic administration of cell-free exosomes generated by human bone marrow derived mesenchymal stem cells cultured under 2D and 3D conditions improves functional recovery in rats after traumatic brain injury. Neurochem Int. 2017; 111: 69-81.
- Yang Y, Ye Y, Kong C, et al. MiR-124 Enriched Exosomes Promoted the M2 Polarization of Microglia and Enhanced Hippocampus Neurogenesis After Traumatic Brain Injury by Inhibiting TLR4 Pathway. 2019; 44: 811-828.
- Li D, Zhang P, Yao X, et al. Exosomes Derived From miR-133b-Modified Mesenchymal Stem Cells Promote Recovery After Spinal Cord Injury. Frontiers in neuroscience. 2018; 12: 845.
- Xu G, Ao R, Zhi Z, Jia J, Yu B. miR-21 and miR-19b delivered by hMSCderived EVs regulate the apoptosis and differentiation of neurons in patients with spinal cord injury. J Cell Physiol. 2019; 234: 10205-10217.
- Ji W, Jiang W, Li M, Li J, Li Z. miR-21 deficiency contributes to the impaired protective effects of obese rat mesenchymal stem cell-derived exosomes against spinal cord injury. Biochimie. 2019; 167: 171-178.
- Zhou X, Chu X, Yuan H, et al. Mesenchymal stem cell derived EVs mediate neuroprotection after spinal cord injury in rats via the microRNA- 21-5p/FasL gene axis. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. 2019; 115: 108818.
- Huang JH, Xu Y, Yin XM, Lin FY. Exosomes Derived from miR-126-modified MSCs Promote Angiogenesis and Neurogenesis and Attenuate Apoptosis after Spinal Cord Injury in Rats. Neuroscience. 2019.
- Li M, Li X, Wang D, et al. Inhibition of exosome release augments neuroinflammation following intracerebral hemorrhage. FASEB journal: Official publication of the Federation of American Societies for Experimental Biology. 2021; 35: e21617.
- Otero-Ortega L, Gómez de Frutos MC, Laso-García F, et al. Exosomes promotes restoration after an experimental animal model of intracerebral hemorrhage. J Cereb Blood Flow Metab. 2018; 38: 767-779.
- Zhang H, Wang Y, Lv Q, Gao J, Hu L, He Z. MicroRNA-21 Overexpression Promotes the Neuroprotective Efficacy of Mesenchymal Stem Cells for Treatment of Intracerebral Hemorrhage. Frontiers in neurology. 2018; 9: 931.
- Duan S, Wang F, Cao J, Wang C. Exosomes Derived from MicroRNA-146a- 5p-Enriched Bone Marrow Mesenchymal Stem Cells Alleviate Intracerebral Hemorrhage by Inhibiting Neuronal Apoptosis and Microglial M1 Polarization. Drug design, development and therapy. 2020; 14: 3143-3158.
- Shen H, Yao X, Li H, et al. Role of Exosomes Derived from miR-133b Modified MSCs in an Experimental Rat Model of Intracerebral Hemorrhage. J Mol Neurosci. 2018; 64: 421-430.
- Lee JK, Park SR, Jung BK, et al. Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS One. 2013; 8: e84256.
- Katakowski M, Buller B, Zheng X, et al. Exosomes from marrow stromal cells expressing miR-146b inhibit glioma growth. Cancer Lett. 2013; 335: 201-204.
- Lang FM, Hossain A, Gumin J, et al. Mesenchymal stem cells as natural biofactories for exosomes carrying miR-124a in the treatment of gliomas. Neuro-oncology. 2018; 20: 380-390.
- Munoz JL, Bliss SA, Greco SJ, Ramkissoon SH, Ligon KL, Rameshwar P. Delivery of Functional Anti-miR-9 by Mesenchymal Stem Cell-derived Exosomes to Glioblastoma Multiforme Cells Conferred Chemosensitivity. Mol Ther Nucleic Acids. 2013; 2: e126.
- Yu L, Gui S, Liu Y, et al. Exosomes derived from microRNA-199aoverexpressing mesenchymal stem cells inhibit glioma progression by down-regulating AGAP2. Aging. 2019; 11: 5300-5318.
- Fareh M, Almairac F, Turchi L, et al. Cell-based therapy using miR-302-367 expressing cells represses glioblastoma growth. Cell Death Dis. 2017; 8: e2713.
- Du T, Ju G, Wu S, et al. Microvesicles derived from human Wharton’s jelly mesenchymal stem cells promote human renal cancer cell growth and aggressiveness through induction of hepatocyte growth factor. PloS one. 2014; 9: e96836.
- Feng Y, Huang W, Wani M, Yu X, Ashraf M. Ischemic preconditioning potentiates the protective effect of stem cells through secretion of exosomes by targeting Mecp2 via miR-22. PloS one. 2014; 9: e88685.
- Wang J, Hendrix A, Hernot S, et al. Bone marrow stromal cell-derived exosomes as communicators in drug resistance in multiple myeloma cells. Blood. 2014; 124: 555-566.