Next Article in Journal
Genetics and Epigenetics of Spontaneous Intracerebral Hemorrhage
Next Article in Special Issue
Lim Domain Binding 3 (Ldb3) Identified as a Potential Marker of Cardiac Extracellular Vesicles
Previous Article in Journal
Effects of Tearing Conditions on the Crack Propagation in a Monolayer Graphene Sheet
Previous Article in Special Issue
Bone Marrow MSC Secretome Increases Equine Articular Chondrocyte Collagen Accumulation and Their Migratory Capacities
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Strategy of Mesenchymal-Stem-Cell-Derived Extracellular Vesicles as Regenerative Medicine

by
Yasunari Matsuzaka
1,2,* and
Ryu Yashiro
2,3
1
Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, University of Tokyo, Minato-ku 108-8639, Tokyo, Japan
2
Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira 187-8551, Tokyo, Japan
3
Department of Infectious Diseases, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi 181-8611, Tokyo, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(12), 6480; https://doi.org/10.3390/ijms23126480
Submission received: 30 April 2022 / Revised: 7 June 2022 / Accepted: 8 June 2022 / Published: 9 June 2022

Abstract

:
Extracellular vesicles (EVs) are lipid bilayer membrane particles that play critical roles in intracellular communication through EV-encapsulated informative content, including proteins, lipids, and nucleic acids. Mesenchymal stem cells (MSCs) are pluripotent stem cells with self-renewal ability derived from bone marrow, fat, umbilical cord, menstruation blood, pulp, etc., which they use to induce tissue regeneration by their direct recruitment into injured tissues, including the heart, liver, lung, kidney, etc., or secreting factors, such as vascular endothelial growth factor or insulin-like growth factor. Recently, MSC-derived EVs have been shown to have regenerative effects against various diseases, partially due to the post-transcriptional regulation of target genes by miRNAs. Furthermore, EVs have garnered attention as novel drug delivery systems, because they can specially encapsulate various target molecules. In this review, we summarize the regenerative effects and molecular mechanisms of MSC-derived EVs.

1. Introduction

Extracellular vesicles (EVs), which are nano- to micro-sized lipid bilayer membrane particles secreted by host cells, play critical roles in novel intercellular communication mechanisms, mediating the transduction of functional molecules with physiological activity, such as microRNAs, mRNAs, proteins, and lipids (Figure 1) [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60]. The level of EVs in humans has gained attention for the early diagnosis of various diseases using liquid biopsy owing to their distribution in various body fluids, including blood, urine, saliva, spinal fluid, and tears, as well as their stability [61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107]. As the amount and type of these functional molecules present within or on the surface of EVs vary depending on the disease, they could be used for disease diagnosis, prognosis, and therapeutic targets [108,109,110,111,112,113,114]. EVs are classified into exosomes, microvesicles, and apoptotic bodies, based on differences in particle size and formation mechanisms [115,116,117,118,119,120,121,122,123,124,125,126]. Since exosomes can be regarded as a natural drug delivery system (DDS) that exists in the living body, they are widely used in drug discovery technologies [127,128,129,130,131]. Furthermore, since exosomes are abundant in numerous species and play a role in the transduction of molecular information between different species, research has focused on their application in various fields and elucidation of their mechanisms in various life phenomena and health and medical care. However, exosome analysis and sample preparation techniques, which form the basis of research, are still immature. Therefore, there is a need for the development of new technologies that can facilitate a breakthrough in the research of EVs as a therapeutic strategy. In this review, we summarize the latest studies on EVs and MSCs as novel therapeutic materials.

2. Mesenchymal Stem Cells (MSCs) for Regeneration

MSCs are pluripotent stem cells with the ability to self-renew, regenerate, and repair deficient cells and the plasticity ability to differentiate into bone, cartilage, blood vessels, and cardiomyocytes, which are derived from the mesoderm [132,133,134,135,136,137,138]. Unlike embryonic stem cells and induced pluripotent stem cells, general stem cells, which are more abundant during early childhood than adulthood, support human growth. Stem cells, also called tissue stem cells, such as adult stem cells or somatic stem cells, are still present at maturity when apparent growth ceases and serve to replenish cells in damaged tissue throughout life [139,140]. Hematopoietic stem cells (HSCs) present in the bone marrow have been studied for more than half a century and are being actively applied clinically [141]. The establishment of a treatment method using HSCs transplantation has expanded the possibilities of transplantation using other tissue stem cells [142,143,144,145]. However, depending on the tissue, such as the brain or heart, it is difficult to separate stem cells from the living tissue and for use as treatment. In recent years, MSCs have been the focus of attention because they are relatively easy to extract from various tissues, including the bone marrow, adipose tissue, placenta, umbilical cord, synovium, and pulp [146,147,148,149,150]. Furthermore, they can also differentiate into ectoderm-derived nerve cells and glial cells that perform functions such as supporting nerve cells and endoderm-derived hepatocytes [151,152,153,154]. Thus, MSCs have garnered attention as cell sources in regenerative medicine because they grow almost indefinitely in a culture dish and perform various functions, such as wound healing, immune regulation, and nerve regeneration; additionally, the therapeutic effects of MSCs against diseases are through their paracrine action rather than differentiation into specific cells. This paracrine action—namely, immune system control, angiogenesis, anti-inflammatory effect, antioxidant action, antiapoptotic action, and tissue repair action, in which cell secretions act on neighboring cells through direct diffusion and not on endocrine cells that act on distant cells via the general circulation—involves various cytokines, including tumor necrosis factor-α (TNF-α), interferon-gamma (IFN-g), interleukin 6 (IL-6), interleukin 10 (IL-10), and transforming growth factor-β (TGF-β), and growth factors secreted by MSCs [155,156,157,158,159]. In particular, in MSC transplantation in cardiomyopathy, MSCs regulate the activation of matrix metalloproteinases (MMPs), leading to the attenuation of cardiac remodeling [160]. In addition, MSCs produce vascular endothelial growth factor (VEGF), insulin-like growth factor-l (IGF-1), adrenomedullin, and hepatocyte growth factor (HGF), which stimulate myogenesis and angiogenesis in the injured myocardium [161,162,163,164,165]. Thus, MSCs improve myocardial perfusion and regeneration by differentiating into cardiomyocytes.
The minimum criteria for defining human MSCs are (1) adherence to plastics under standard culture conditions; (2) positive cell surface markers CD73, CD90, CD105, and negative CD11b or CD14, CD19 or CD79a, CD34, CD45, and HLA-DR; and (3) ability to differentiate into osteoblasts, chondrocytes, and adipocytes [166]. Since MSCs express major histocompatibility complex (MHC) class I, but not MHC class II, they are less likely to be attacked by natural killer (NK) cells and exhibit difficulty in developing humoral immunity. The MSCs used for clinical purposes are collected from various tissues such as bone marrow, umbilical cord, umbilical cord blood, and fat, and have important biological activities related to tissue repair, such as anti-inflammatory effect, proliferative factor secretion, and angiogenesis-promoting effects without the risk of tumorigenesis [167,168,169]. Furthermore, it is clear that the properties of MSCs differ depending on the organ from which they are collected. Adipose-derived MSCs, as well as bone-marrow-derived MSCs, have received a great deal of attention because they can be collected more easily and abundantly throughout the body, are less invasive, and have excellent organ repair and immunomodulatory abilities, compared with those of bone-marrow-derived MSCs [170,171]. Bone-marrow-derived MSCs comprise only approximately 0.01% of the cells in the bone marrow, whereas the number of adipose-derived MSCs in adipose tissue is 500 times that of MSCs in the bone marrow. Additionally, adipose-derived MSCs produce more growth factors, such as HGF and VEGF, that contribute to organ repair than those derived from bone marrow [172]. Furthermore, in addition to the ability to differentiate into fat, bone, and cartilage, similar to bone-marrow-derived MSCs, they have the ability to differentiate into the muscle, which is not derived from bone marrow. Although their cell morphology and differentiation potential are not different from those of bone-marrow-derived MSCs, they are characterized by a strong proliferative capacity, little effect of aging, and a small decrease in bone differentiation ability [173,174]. The number and growth of bone-marrow-derived MSCs decrease with age. Adipose-derived MSCs can grow sufficiently even if they are obtained from the adipose tissue of elderly individuals. General anesthesia is used to collect MSCs derived from the bone marrow, which puts a heavy burden on the patient. In contrast, when adipose-derived MSCs are collected, the burden on the patient is light because adipose tissue is close to the surface of the body. In addition, adipose-derived MSCs are characterized by a higher immunosuppressive capacity than that of bone-marrow-derived MSCs [175]. Animal studies have shown that adipose-derived MSCs can dramatically improve nephritis [176]. Moreover, MSCs accumulate at the treatment site because of the “homing phenomenon”, which includes MSCs recognizing the lesion-induced signals such as cytokines and adhesion factors. Therefore, when MSCs are injected into the blood circulation, they naturally accumulate at the desired site and exert a therapeutic effect. In particular, after intramuscular injection, MSCs deposit in the interstices of muscle fibers through the production of basic fibroblast growth factor (bFGF) and VEGF, and induce angiogenesis and support nerve cell regeneration, leading to amelioration of neuropathy [177,178,179]. These results suggest that MSCs are excellent for clinical use due to having a wide range of applications and sufficient supply, in addition to fewer safety-related and ethical issues. Allogenic MSCs are expected to have a wide range of therapeutic effects, and clinical trials are currently underway in various diseases, such as osteochondral disease, decompensated liver cirrhosis, systemic erythematosus, acute transplant-to-host disease, Crohn’s disease, myocardial infarction, cerebral infarction, and Parkinson’s disease [180,181,182,183,184,185,186,187,188,189,190,191,192]. In safety evaluation studies, mild-to-moderate abnormalities, such as fever, chills, headache, fatigue, increased anxiety, redness of administered skin, edema, weight loss, cold, and cough, were frequently observed, but no serious acute adverse events were reported even in elderly patients. Currently, regenerative medicinal products have been approved for the treatment of spinal cord injury and acute graft-versus-host disease after HSC transplantation. However, some issues still exist when using MSC for regenerative medicine.
Since MSCs, compared with autologous cells, have a higher risk of transmitting infectious factors such as viruses to patients undergoing transplantation, it is necessary to exert stringent control on the quality of the cells used as raw materials [193]. Additionally, a risk of immune rejection and low engraftment compared with that in autologous cells has been suggested [194,195,196,197,198,199]. It can be pointed out that the risk of tumorigenicity may be low because the engraftment is lower than that of autologous cells, and the immune system is easily activated. However, as the cells are amplified, the risk of accumulation of genomic mutation and chromosomal abnormalities also increases [200]. Therefore, appropriate evaluation is a critical factor because the risk of tumorigenesis varies greatly depending on the culture, proliferation period of used cells, and number of cells to be transplanted. With allogeneic cells, it is assumed that a single cell strain will be transplanted into multiple patients. Therefore, it is relatively easy to standardize and manage the timing of the processing and shipping, which leads to reduced costs. However, the risk of an intravascular embolism when administered transvascularly has been previously reported for both autologous and allogeneic cells in clinical use [201]. In addition, as an allogeneic bone-marrow-derived MSC preparation, Temcel® HS injection has already been approved for regenerative medicine [202]. However, the results of nonclinical studies in rats showed cell embolism in the brain, heart, lung, liver, kidney, spleen, bladder, etc., and thrombus in the lungs of some individuals [203]. Furthermore, the risk assessment in clinical studies did not rule out causality with the administered cells owing to one patient who died of gastrointestinal bleeding and one among 25 patients who exhibited a systemic rash after administration. However, no such adverse events have been confirmed in nonclinical studies to date. Another MSC product derived from allogeneic cord blood, CARTISTEM, is undergoing two clinical studies, including phase I/II and phase III, but no major adverse events have been reported yet [204].

3. MSC-EVs for Regeneration

MSC is a mesoderm-derived somatic stem cell that can be established from tissues such as bone marrow, fat, umbilical cord, and pulp, and has the ability to differentiate into fat, bone, and cartilage [205,206]. In addition to this differentiation capacity, MSC exerts secretory effects that induce anti-inflammatory, antifibrotic, or immunosuppressive effects. In recent years, it has been suggested that these effects are due to EVs secreted from bone marrow, fat, umbilical cord, menstruation blood, pulp, etc. [207,208,209,210]. MSC therapy is expected to be applied to various diseases including severe heart failure, but there are challenges such as individual differences and insufficient effects. Since T-cadherin, a receptor for adiponectin, is expressed in MSCs, it was revealed that adiponectin, which is secreted by adipocytes and is abundant in blood, promotes EVs production, thereby exerting a therapeutic effect on MSCs using a mouse model of heart failure (Figure 2) [211]. Thus, in MSC therapy, since the administrated MSCs produce a large amount of EVs by incorporating adiponectin into the cells via T-cadherin expressed on the membrane surface, the action of EVs on the heart improves the cardiac function of the heart failure model.

4. Therapy via MSC-Derived EVs as a Novel DDS System

Recent studies have shown that EVs secreted by MSCs have similar therapeutic effects on MSCs because some of the paracrine effects of MSCs are derived from exosomes, and most of the therapeutic effects of MSCs are responsible for the paracrine effects of EVs in certain diseases via miRNAs, mRNAs, and proteins as functional molecules [212,213,214,215].
As for the therapeutic effect of EVs secreted by MSCs, first, studies using animals modeled for acute renal disease showed that MSCs acted paraclinically against living epithelial cells to support tissue regeneration, in which paracrine effect plays a vital role via EV-encapsulated mRNAs involved in transcriptional regulation, proliferation, and immune regulation to induce tissue regeneration [216,217,218]. Furthermore, the in vitro signaling pathway to induce apoptosis and suppress the proliferation of renal epithelial cells was inactivated in the presence of EVs released by MSCs, leading to protection against cellular damage [219,220,221]. However, since no such effect was observed in fibroblast-derived EVs, it was assumed that the cytoprotective action is a specific property of MSC-derived EVs. Additionally, MSCs, which are induced to differentiate from ES cells or established from various fetal tissues, MSC-derived EVs have been shown to have therapeutic effects on myocardial damage in a very short time in animal models with myocardial ischemia–reperfusion disorder [222,223]. This was achieved through the delivery of proteins retaining functionality within the EVs to cardiomyocytes efficiently and rapidly, leading to reduced oxidative stress and promoted phosphorylation of the PI3K/Akt pathway; where the equivalent therapeutic effect of the EVs was indicated as an amount of 1/10 or less of the EV-depleted supernatant from the MSC culture [224,225,226,227]. In addition, the increased expression of microRNAs secreted by MSCs had therapeutic effects via the secretion of neurotrophic factors and angiogenesis-promoting factors, and the EVs ameliorate neuropathy in stroke animal models through neurite outgrowth, where administration of MSC-derived EVs intravenously had the same effects as that of cell administration [228,229]. Furthermore, in the Alzheimer’s disease (AD) model, neutral endopeptidase (NEP), which is the enzyme responsible for the rate-determining process of amyloid-beta (Aβ) in the brain of patients with this disorder and excessive accumulation of Aβ is one of the major characteristics of the pathophysiology of AD, was higher in adipose-derived MSCs than in bone-marrow-derived MSCs and contributed to the higher efficiency of Aβ degradation [230,231,232]. In addition, the NEP protein is also present in EVs from adipose-derived MSCs and exhibits enzymatic activity, leading to intracellular Aβ degradation via uptake by neural cells [233]. Moreover, in a mouse model of hypoxia-induced pulmonary hypertension, MSC-derived EVs exerted therapeutic effects by suppressing inflammation through the inhibition of the signal transducer and activator of transcription 3 (STAT3) pathway in the lung [234]. Here, the EVs suppressed the upregulation of the hypoxia-inducible miR-17 superfamily and induced the upregulation of the growth-inhibitory miR-204, and no therapeutic effects were observed in the EV-depleted supernatant. Additionally, in a mouse model of acute lung injury, keratinocyte growth factor (KGF) mRNA, which is important for the therapeutic effect of EVs on lung disorders due to its abundance within the EVs and the paracrine effect of MSCs, is partially responsible for the healing effect on lung injury [235]. In addition, the administration of MSCs into the lungs of lipopolysaccharide-induced acute lung injury mice restored the proliferative capacity of alveolar epithelial cells and lung function through mitochondrial transmission [236]. Further, MSC-derived EVs have been reported to have opposite effects, i.e., tumor progression via tumor microenvironment remodeling and tumor suppression via regulation of immune responses and intercellular signaling. However, it has been suggested that they will be safe carriers of antitumor drugs [237,238,239].
Thus, although substantial evidence is available on the usefulness of the therapeutic effects of MSC-derived EVs, some important challenges remain. The basic molecular mechanisms of EVs, such as secretion, uptake by the receiving cell, sorting of their contents, and biogenesis, are still unclear. Various regulatory molecules in multiple molecular pathways have been identified as the mechanism of EV synthesis and secretion [240,241,242,243,244]. These pathways include the endosomal-sorting complex required for transport (ESCRT) involved in membrane vesicle formation, neutral sphingomyelinase 2/sphingomyelin phosphodiesterase 3 (nSMase2/Smpd3) that is the rate-determining enzyme for a membrane component ceramide synthesis, members of the Rab GTPase family involved in intracellular membrane vesicle transport, and heparanase that is a heparan sulfate degrading enzyme [245,246,247,248]. However, the degree of contribution of these regulatory molecules to EV biogenesis and secretion pathways varies greatly depending on the cell type. Therefore, to investigate the function of EVs in a cell, it is difficult to predict which pathways or molecules should be suppressed, even if an attempt is made to inhibit the secretion of EVs in target cells. Little is known about the mechanism of EV uptake into cells by the nonimmune cell system, except for the uptake of EVs in immune cells by phagocytosis. Another challenge is the unification of experimental techniques in EV research, especially in the isolation of EVs, including the most common techniques ultracentrifugation, separation based on molecular size via gel chromatography, and extraction reagents, which is the root of this research. It is unclear whether the EV fractions recovered by using different isolation methods show bioequivalence.
When considering the prospects for treatment strategies using MSC-derived EVs, first, a stable supply of MSC-derived EVs is necessary to identify suitable molecular pathways and appropriate management techniques to increase the yield of MSC-derived EVs while retaining their original therapeutic effects. Second, ensuring sufficient capacity for clinical application, if the therapeutic effect of MSC-derived EVs can be amplified, the therapeutic applicability will be greatly increased. A possible method to resolve this issue is to overexpress therapeutic molecules, such as mRNAs, miRNAs, or proteins, in MSC-derived EVs and produce a large number of EVs encapsulating these molecules. In fact, it has been shown that cells overexpressing certain miRNAs secrete EVs containing abundant miRNAs [249,250,251]. In addition, an approach that completely improves the therapeutic effect of MSC-derived EVs would be to produce a large number of EVs containing an excess of molecules by inducing a specific stimulus to the MSCs with the target therapeutic effects. Third, DDS techniques need to be developed to specifically deliver the MSC-derived EVs to target tissues. Since MSC-derived EVs have not been characterized completely, there is an unexpected risk from systemic administration via the intravenous route. For example, given that MSC-derived EVs can promote the repair of damaged tissues, the risk of carcinogenesis due to delivery to nontarget tissues cannot be ruled out. At present, modification of the surface of EVs seems promising for tissue-specific DDS through the expression of receptor proteins that exhibit tissue-specific expression using genetic recombination technology [252,253]. Thus, a new DDS with high specificity and delivery efficiency can be developed using MSC-derived EVs to introduce functional molecules with therapeutic effects. In contrast, since the complete biological action of EVs is not completely understood yet, the risk of side effects is a major concern. Bone-marrow-derived EVs induce dormancy of tumor cells and are involved in long-term recurrence, while EV-encapsulated miRNAs secreted by tumor cells cause intratumoral angiogenesis and promote metastasis in the brain through disruption of the blood–brain barrier [254,255,256].

5. Clinical Use of MSC-EVs

More than 500 clinical research on MSC is underway in the world for various target diseases, including rheumatoid arthritis, systemic lupus erythematosus, Crohn’s disease, myocardial infarction, Parkinson’s disease, spinal cord injury, osteoarthritis, and graft-versus-host disease (GVHD) [257,258,259,260,261,262,263,264,265,266]. MSCs other than those derived from bone marrow are also being clinically applied. Physician-led clinical trials such as amniotic-membrane- or umbilical-cord-derived MSC treatments for Crohn’s disease or acute GVHD are underway. In addition, a study of the induced pluripotent stem (iPS)-cell-derived MSCs for acute GVHD has begun. Further, bone-marrow-derived MSC Temcel® indication expanded to epidermolysis bullosa. Additionally, a clinical trial has begun in which pulp-derived MSCs are administrated for acute cerebral infarction. Since MSCs are slightly different in nature depending on the organization from which they are sourced, it is important to select a cell source suitable for specific diseases and therapeutic effects. Moreover, it has been clarified that the secreted EV plays an important role when MSC exerts various actions. Additionally, it has also been reported that administration of EVs secreted by cultured bone-marrow-derived MSC to patients with refractory GVHD resulted in improvement in symptoms. Furthermore, clinical trials of MSC-derived exosomes are currently underway for diabetes mellitus type 1, cerebrovascular disorders, coronavirus, Alzheimer's disease, and osteoarthritis (Table 1).

6. Conclusions

MSC-derived EVs treated with the cytokines upregulate the expression of the immunomodulatory molecules, including miRNAs and proteins, involved in the immunoregulatory pathways. This plays an important role in tissue repair of chronic damage through the concentration of active ingredients in the contents and efficient migration of the macrophages incorporated in the MSC-derived EVs to the damage site due to the removal of dead cells and improvement of fibrosis. These therapeutic effects are equal to or higher than those of the MSCs themselves. These reports suggest that the administration of MSC-derived EVs is a useful novel cell-free therapeutic strategy.

Author Contributions

Writing—review and editing, Y.M.; supervision, R.Y.; funding acquisition, Y.M. All authors have read and agreed to the published version of the manuscript.

Funding

This review was funded by Fukuda Foundation for Medical Technology, and the APC was funded by Fukuda Foundation for Medical Technology.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tan, Y.; Tang, F.; Li, J.; Yu, H.; Wu, M.; Wu, Y.; Zeng, H.; Hou, K.; Zhang, Q. Tumor-derived exosomes: The emerging orchestrators in melanoma. Biomed. Pharmacother. 2022, 149, 112832. [Google Scholar] [CrossRef] [PubMed]
  2. Dong, M.; Liu, Q.; Xu, Y.; Zhang, Q. Extracellular Vesicles: The Landscape in the Progression, Diagnosis, and Treatment of Triple-Negative Breast Cancer. Front. Cell Dev. Biol. 2022, 10, 842898. [Google Scholar] [CrossRef] [PubMed]
  3. Bond, S.T.; Calkin, A.C.; Drew, B.G. Adipose-Derived Extracellular Vesicles: Systemic Messengers and Metabolic Regulators in Health and Disease. Front. Physiol. 2022, 13, 837001. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, J.; Yue, B.L.; Huang, Y.Z.; Lan, X.Y.; Liu, W.J.; Chen, H. Exosomal RNAs: Novel Potential Biomarkers for Diseases—A Review. Int. J. Mol. Sci. 2022, 23, 2461. [Google Scholar] [CrossRef]
  5. Li, H.; Su, Y.; Wang, F.; Tao, F. Exosomes: A new way of protecting and regenerating optic nerve after injury. Hum. Cell 2022, 35, 771–778. [Google Scholar] [CrossRef]
  6. Kowalczyk, A.; Wrzecińska, M.; Czerniawska-Piątkowska, E.; Kupczyński, R. Exosomes—Spectacular role in reproduction. Biomed. Pharmacother. 2022, 148, 112752. [Google Scholar] [CrossRef]
  7. Rodríguez, D.A.; Vader, P. Extracellular Vesicle-Based Hybrid Systems for Advanced Drug Delivery. Pharmaceutics 2022, 14, 267. [Google Scholar] [CrossRef]
  8. Infante, A.; Alcorta-Sevillano, N.; Macías, I.; Rodríguez, C.I. Educating EVs to Improve Bone Regeneration: Getting Closer to the Clinic. Int. J. Mol. Sci. 2022, 23, 1865. [Google Scholar] [CrossRef]
  9. Chen, J.; Li, P.; Zhang, T.; Xu, Z.; Huang, X.; Wang, R.; Du, L. Review on Strategies and Technologies for Exosome Isolation and Purification. Front. Bioeng. Biotechnol. 2022, 9, 811971. [Google Scholar] [CrossRef]
  10. Bischoff, J.P.; Schulz, A.; Morrison, H. The role of exosomes in intercellular and inter-organ communication of the peripheral nervous system. FEBS Lett. 2022, 596, 655–664. [Google Scholar] [CrossRef]
  11. Cione, E.; Cannataro, R.; Gallelli, L.; De Sarro, G.; Caroleo, M.C. Exosome microRNAs in Metabolic Syndrome as Tools for the Early Monitoring of Diabetes and Possible Therapeutic Options. Pharmaceuticals 2021, 14, 1257. [Google Scholar] [CrossRef] [PubMed]
  12. Santos, A.; Domingues, C.; Jarak, I.; Veiga, F.; Figueiras, A. Osteosarcoma from the unknown to the use of exosomes as a versatile and dynamic therapeutic approach. Eur. J. Pharm. Biopharm. 2022, 170, 91–111. [Google Scholar] [CrossRef] [PubMed]
  13. Quadri, Z.; Elsherbini, A.; Bieberich, E. Extracellular vesicles in pharmacology: Novel approaches in diagnostics and therapy. Pharmacol. Res. 2022, 175, 105980. [Google Scholar] [CrossRef] [PubMed]
  14. Spellicy, S.E.; Stice, S.L. Tissue and Stem Cell Sourced Extracellular Vesicle Communications with Microglia. Stem Cell Rev. Rep. 2021, 17, 357–368. [Google Scholar] [CrossRef] [PubMed]
  15. Yang, D.; Liu, J. Targeting extracellular vesicles-mediated hepatic inflammation as a therapeutic strategy in liver diseases. Liver Int. 2020, 40, 2064–2073. [Google Scholar] [CrossRef] [PubMed]
  16. Beck, S.; Hochreiter, B.; Schmid, J.A. Extracellular Vesicles Linking Inflammation, Cancer and Thrombotic Risks. Front. Cell Dev. Biol. 2022, 10, 859863. [Google Scholar] [CrossRef]
  17. Han, C.; Yang, J.; Sun, J.; Qin, G. Extracellular vesicles in cardiovascular disease: Biological functions and therapeutic implications. Pharmacol. Ther. 2021, 108025. [Google Scholar] [CrossRef] [PubMed]
  18. Coly, P.M.; Boulanger, C.M. Role of extracellular vesicles in atherosclerosis: An update. J. Leukoc. Biol. 2022, 111, 51–62. [Google Scholar] [CrossRef]
  19. Sorop, A.; Constantinescu, D.; Cojocaru, F.; Dinischiotu, A.; Cucu, D.; Dima, S.O. Exosomal microRNAs as Biomarkers and Therapeutic Targets for Hepatocellular Carcinoma. Int. J. Mol. Sci. 2021, 22, 4997. [Google Scholar] [CrossRef]
  20. Velot, É.; Madry, H.; Venkatesan, J.K.; Bianchi, A.; Cucchiarini, M. Is Extracellular Vesicle-Based Therapy the Next Answer for Cartilage Regeneration? Front. Bioeng. Biotechnol. 2021, 9, 645039. [Google Scholar] [CrossRef]
  21. Lee, S.; Choi, C.; Yoo, T.H. Extracellular vesicles in kidneys and their clinical potential in renal diseases. Kidney Res. Clin. Pract. 2021, 40, 194–207. [Google Scholar] [CrossRef] [PubMed]
  22. Saheera, S.; Jani, V.P.; Witwer, K.W.; Kutty, S. Extracellular vesicle interplay in cardiovascular pathophysiology. Am. J. Physiol. Heart Circ. Physiol. 2021, 320, H1749–H1761. [Google Scholar] [CrossRef] [PubMed]
  23. Geng, T.; Pan, P.; Leung, E.; Chen, Q.; Chamley, L.; Wu, Z. Recent Advancement and Technical Challenges in Developing Small Extracellular Vesicles for Cancer Drug Delivery. Pharm. Res. 2021, 38, 179–197. [Google Scholar] [CrossRef] [PubMed]
  24. Jin, T.; Gu, J.; Li, Z.; Xu, Z.; Gui, Y. Recent Advances on Extracellular Vesicles in Central Nervous System Diseases. Clin. Interv. Aging 2021, 16, 257–274. [Google Scholar] [CrossRef] [PubMed]
  25. Bazzoni, R.; Tanasi, I.; Turazzi, N.; Krampera, M. Update on the role and utility of extracellular vesicles in hematological malignancies. Stem Cells 2022, sxac032. [Google Scholar] [CrossRef]
  26. Vahabi, A.; Rezaie, J.; Hassanpour, M.; Panahi, Y.; Nemati, M.; Rasmi, Y.; Nemati, M. Tumor Cells-derived exosomal CircRNAs: Novel cancer drivers, molecular mechanisms, and clinical opportunities. Biochem. Pharmacol. 2022, 200, 115038. [Google Scholar] [CrossRef]
  27. Kim, G.; Chen, X.; Yang, Y. Pathogenic Extracellular Vesicle (EV) Signaling in Amyotrophic Lateral Sclerosis (ALS). Neurotherapeutics 2022, in press. [CrossRef]
  28. Loch-Neckel, G.; Matos, A.T.; Vaz, A.R.; Brites, D. Challenges in the Development of Drug Delivery Systems Based on Small Extracellular Vesicles for Therapy of Brain Diseases. Front. Pharmacol. 2022, 13, 839790. [Google Scholar] [CrossRef]
  29. Lazana, I.; Anagnostopoulos, C. A Novel, Cell-Free Therapy to Enter Our Hearts: The Potential Role of Small EVs in Prevention and Treatment of CVD. Int. J. Mol. Sci. 2022, 23, 3662. [Google Scholar] [CrossRef]
  30. Goutas, D.; Pergaris, A.; Goutas, N.; Theocharis, S. Utilizing Exosomal-EPHs/Ephrins as Biomarkers and as a Potential Platform for Targeted Delivery of Therapeutic Exosomes. Int. J. Mol. Sci. 2022, 23, 3551. [Google Scholar] [CrossRef]
  31. Allegra, A.; Petrarca, C.; Di Gioacchino, M.; Casciaro, M.; Musolino, C.; Gangemi, S. Exosome-Mediated Therapeutic Strategies for Management of Solid and Hematological Malignancies. Cells 2022, 11, 1128. [Google Scholar] [CrossRef] [PubMed]
  32. Wu, Q.; Duan, W.Z.; Chen, J.B.; Zhao, X.P.; Li, X.J.; Liu, Y.Y.; Ma, Q.Y.; Xue, Z.; Chen, J.X. Extracellular Vesicles: Emerging Roles in Developing Therapeutic Approach and Delivery Tool of Chinese Herbal Medicine for the Treatment of Depressive Disorder. Front. Pharmacol. 2022, 13, 843412. [Google Scholar] [CrossRef] [PubMed]
  33. Khadka, A.; Spiers, J.G.; Cheng, L.; Hill, A.F. Extracellular vesicles with diagnostic and therapeutic potential for prion diseases. Cell Tissue Res. 2022, in press. [CrossRef] [PubMed]
  34. Bağcı, C.; Sever-Bahcekapili, M.; Belder, N.; Bennett, A.P.S.; Erdener, Ş.E.; Dalkara, T. Overview of extracellular vesicle characterization techniques and introduction to combined reflectance and fluorescence confocal microscopy to distinguish extracellular vesicle subpopulations. Neurophotonics 2022, 9, 021903. [Google Scholar] [CrossRef] [PubMed]
  35. Gomez, N.; James, V.; Onion, D.; Fairclough, L.C. Extracellular vesicles and chronic obstructive pulmonary disease (COPD): A systematic review. Respir Res. 2022, 23, 82. [Google Scholar] [CrossRef] [PubMed]
  36. Qian, D.; Xie, Y.; Huang, M.; Gu, J. Tumor-derived exosomes in hypoxic microenvironment: Release mechanism, biological function and clinical application. J. Cancer 2022, 13, 1685–1694. [Google Scholar] [CrossRef] [PubMed]
  37. Liu, K.; Gao, X.; Kang, B.; Liu, Y.; Wang, D.; Wang, Y. The Role of Tumor Stem Cell Exosomes in Cancer Invasion and Metastasis. Front. Oncol. 2022, 12, 836548. [Google Scholar] [CrossRef]
  38. Luxmi, R.; King, S.M. Cilia-derived vesicles: An ancient route for intercellular communication. Semin. Cell Dev. Biol. 2022, S1084-9521, 00081–00087. [Google Scholar] [CrossRef]
  39. Kumari, M.; Anji, A. Small but Mighty-Exosomes, Novel Intercellular Messengers in Neurodegeneration. Biology 2022, 11, 413. [Google Scholar] [CrossRef]
  40. Thompson, R.E.; Bouma, G.J.; Hollinshead, F.K. The Roles of Extracellular Vesicles and Organoid Models in Female Reproductive Physiology. Int. J. Mol. Sci. 2022, 23, 3186. [Google Scholar] [CrossRef]
  41. Hu, M.; Li, J.; Liu, C.G.; Goh, R.M.W.J.; Yu, F.; Ma, Z.; Wang, L. Noncoding RNAs of Extracellular Vesicles in Tumor Angiogenesis: From Biological Functions to Clinical Significance. Cells 2022, 11, 947. [Google Scholar] [CrossRef] [PubMed]
  42. Kim, H.Y.; Kwon, S.; Um, W.; Shin, S.; Kim, C.H.; Park, J.H.; Kim, B.S. Functional Extracellular Vesicles for Regenerative Medicine. Small 2022, e2106569. [Google Scholar] [CrossRef] [PubMed]
  43. Duggan, M.R.; Lu, A.; Foster, T.C.; Wimmer, M.; Parikh, V. Exosomes in Age-Related Cognitive Decline: Mechanistic Insights and Improving Outcomes. Front. Aging Neurosci. 2022, 14, 834775. [Google Scholar] [CrossRef]
  44. Miao, H.B.; Wang, F.; Lin, S.; Chen, Z. Update on the role of extracellular vesicles in rheumatoid arthritis. Expert Rev. Mol. Med. 2022, 24, e12. [Google Scholar] [CrossRef] [PubMed]
  45. Xu, K.; Jin, Y.; Li, Y.; Huang, Y.; Zhao, R. Recent Progress of Exosome Isolation and Peptide Recognition-Guided Strategies for Exosome Research. Front. Chem. 2022, 10, 844124. [Google Scholar] [CrossRef]
  46. Panvongsa, W.; Pegtel, D.M.; Voortman, J. More than a Bubble: Extracellular Vesicle microRNAs in Head and Neck Squamous Cell Carcinoma. Cancers 2022, 14, 1160. [Google Scholar] [CrossRef]
  47. van Niel, G.; Carter, D.R.F.; Clayton, A.; Lambert, D.W.; Raposo, G.; Vader, P. Challenges and directions in studying cell-cell communication by extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2022, in press. [CrossRef]
  48. Keshtkar, S.; Kaviani, M.; Soleimanian, S.; Azarpira, N.; Asvar, Z.; Pakbaz, S. Stem Cell-Derived Exosome as Potential Therapeutics for Microbial Diseases. Front. Microbiol. 2022, 12, 786111. [Google Scholar] [CrossRef]
  49. Lee, Y.; Kim, J.H. The emerging roles of extracellular vesicles as intercellular messengers in liver physiology and pathology. Clin. Mol. Hepatol. 2022, in press. [CrossRef]
  50. Pancholi, S.; Tripathi, A.; Bhan, A.; Acharya, M.M.; Pillai, P. Emerging Concepts on the Role of Extracellular Vesicles and Its Cargo Contents in Glioblastoma-Microglial Crosstalk. Mol. Neurobiol. 2022, 59, 2822–2837. [Google Scholar] [CrossRef]
  51. He, X.; Guan, F.; Lei, L. Structure and function of glycosphingolipids on small extracellular vesicles. Glycoconj. J. 2022, 39, 197–205. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, F.; Guo, J.; Zhang, Z.; Duan, M.; Wang, G.; Qian, Y.; Zhao, H.; Yang, Z.; Jiang, X. Application of engineered extracellular vesicles for targeted tumor therapy. J. Biomed. Sci. 2022, 29, 14. [Google Scholar] [CrossRef] [PubMed]
  53. Spiers, J.G.; Vassileff, N.; Hill, A.F. Neuroinflammatory Modulation of Extracellular Vesicle Biogenesis and Cargo Loading. Neuromol. Med. 2022, in press. [CrossRef] [PubMed]
  54. Yeung, C.L.S.; Yam, J.W.P. Therapy-induced modulation of extracellular vesicles in hepatocellular carcinoma. Semin. Cancer Biol. 2022, S1044-579X, 40–42. [Google Scholar] [CrossRef] [PubMed]
  55. Peng, J.; Liang, Q.; Xu, Z.; Cai, Y.; Peng, B.; Li, J.; Zhang, W.; Kang, F.; Hong, Q.; Yan, Y.; et al. Current Understanding of Exosomal MicroRNAs in Glioma Immune Regulation and Therapeutic Responses. Front. Immunol. 2022, 12, 813747. [Google Scholar] [CrossRef] [PubMed]
  56. Li, Q.C.; Li, C.; Zhang, W.; Pi, W.; Han, N. Potential Effects of Exosomes and Their MicroRNA Carrier on Osteoporosis. Curr. Pharm. Des. 2022, in press. [CrossRef]
  57. Zhang, H.; Xing, J.; Dai, Z.; Wang, D.; Tang, D. Exosomes: The key of sophisticated cell-cell communication and targeted metastasis in pancreatic cancer. Cell Commun. Signal 2022, 20, 9. [Google Scholar] [CrossRef]
  58. Zhou, Z.W.; Zheng, W.; Xiang, Z.; Ye, C.S.; Yin, Q.Q.; Wang, S.H.; Xu, C.A.; Wu, W.H.; Hui, T.C.; Wu, Q.Q.; et al. Clinical implications of exosome-derived noncoding RNAs in liver. Lab. Investig. 2022, in press. [CrossRef]
  59. Cong, M.; Tan, S.; Li, S.; Gao, L.; Huang, L.; Zhang, H.G.; Qiao, H. Technology insight: Plant-derived vesicles-How far from the clinical biotherapeutics and therapeutic drug carriers? Adv. Drug Deliv. Rev. 2022, 182, 114108. [Google Scholar] [CrossRef]
  60. Li, J.; Zhang, G.; Liu, C.G.; Xiang, X.; Le, M.T.N.; Sethi, G.; Wang, L.; Goh, B.C.; Ma, Z. The potential role of exosomal circRNAs in the tumor microenvironment: Insights into cancer diagnosis and therapy. Theranostics 2022, 12, 87–104. [Google Scholar] [CrossRef]
  61. Ye, D.; Gong, M.; Deng, Y.; Fang, S.; Cao, Y.; Xiang, Y.; Shen, Z. Roles and clinical application of exosomal circRNAs in the diagnosis and treatment of malignant tumors. J. Transl. Med. 2022, 20, 161. [Google Scholar] [CrossRef] [PubMed]
  62. Nesteruk, K.; Levink, I.J.M.; Dits, N.F.J.; Cahen, D.L.; Peppelenbosch, M.P.; Bruno, M.J.; Fuhler, G.M. Size and Concentration of Extracellular Vesicles in Pancreatic Juice From Patients With Pancreatic Ductal Adenocarcinoma. Clin. Transl. Gastroenterol. 2022, 13, e00465. [Google Scholar] [CrossRef] [PubMed]
  63. Pink, R.C.; Beaman, E.M.; Samuel, P.; Brooks, S.A. Carter DRF. Utilising extracellular vesicles for early cancer diagnostics: Benefits, challenges and recommendations for the future. Br. J. Cancer 2022, 126, 323–330. [Google Scholar] [CrossRef] [PubMed]
  64. Soltész, B.; Buglyó, G.; Németh, N.; Szilágyi, M.; Pös, O.; Szemes, T.; Balogh, I.; Nagy, B. The Role of Exosomes in Cancer Progression. Int. J. Mol. Sci. 2021, 23, 8. [Google Scholar] [CrossRef]
  65. Nimitrungtawee, N.; Inmutto, N.; Chattipakorn, S.C.; Chattipakorn, N. Extracellular vesicles as a new hope for diagnosis and therapeutic intervention for hepatocellular carcinoma. Cancer Med. 2021, 10, 8253–8271. [Google Scholar] [CrossRef]
  66. Liu, J.; Ren, L.; Li, S.; Li, W.; Zheng, X.; Yang, Y.; Fu, W.; Yi, J.; Wang, J.; Du, G. The biology, function, and applications of exosomes in cancer. Acta. Pharm. Sin. B. 2021, 11, 2783–2797. [Google Scholar] [CrossRef]
  67. Caruso Bavisotto, C.; Marino Gammazza, A.; Campanella, C.; Bucchieri, F.; Cappello, F. Extracellular heat shock proteins in cancer: From early diagnosis to new therapeutic approach. Semin. Cancer Biol. 2021, S1044-579X, 00244-3. [Google Scholar] [CrossRef]
  68. Zhu, L.; Zhao, L.; Wang, Q.; Zhong, S.; Guo, X.; Zhu, Y.; Bao, J.; Xu, K.; Liu, S. Circulating exosomal miRNAs and cancer early diagnosis. Clin. Transl. Oncol. 2022, 24, 393–406. [Google Scholar] [CrossRef]
  69. Jing, Z.; Chen, K.; Gong, L. The Significance of Exosomes in Pathogenesis, Diagnosis, and Treatment of Esophageal Cancer. Int. J. Nanomed. 2021, 16, 6115–6127. [Google Scholar] [CrossRef]
  70. Jiang, C.; Zhang, N.; Hu, X.; Wang, H. Tumor-associated exosomes promote lung cancer metastasis through multiple mechanisms. Mol. Cancer 2021, 20, 117. [Google Scholar] [CrossRef]
  71. Kumar, S.; Kumar, P.; Kodidela, S.; Duhart, B.; Cernasev, A.; Nookala, A.; Kumar, A.; Singh, U.; Bissler, J. Racial Health Disparity and COVID-19. J. Neuroimmune Pharmacol. 2021, 16, 729–742. [Google Scholar] [CrossRef] [PubMed]
  72. Testa, A.; Venturelli, E.; Brizzi, M.F. Extracellular Vesicles: New Tools for Early Diagnosis of Breast and Genitourinary Cancers. Int. J. Mol. Sci. 2021, 22, 8430. [Google Scholar] [CrossRef] [PubMed]
  73. Xiong, H.; Huang, Z.; Yang, Z.; Lin, Q.; Yang, B.; Fang, X.; Liu, B.; Chen, H.; Kong, J. Recent Progress in Detection and Profiling of Cancer Cell-Derived Exosomes. Small 2021, 17, e2007971. [Google Scholar] [CrossRef] [PubMed]
  74. Tatischeff, I. Current Search through Liquid Biopsy of Effective Biomarkers for Early Cancer Diagnosis into the Rich Cargoes of Extracellular Vesicles. Int. J. Mol. Sci. 2021, 22, 5674. [Google Scholar] [CrossRef]
  75. Liu, J.; Chen, Y.; Pei, F.; Zeng, C.; Yao, Y.; Liao, W.; Zhao, Z. Extracellular Vesicles in Liquid Biopsies: Potential for Disease Diagnosis. Biomed. Res. Int. 2021, 2021, 6611244. [Google Scholar] [CrossRef]
  76. Yousif, G.; Qadri, S.; Haik, M.; Haik, Y.; Parray, A.S.; Shuaib, A. Circulating Exosomes of Neuronal Origin as Potential Early Biomarkers for Development of Stroke. Mol. Diagn. Ther. 2021, 25, 163–180. [Google Scholar] [CrossRef]
  77. Rastogi, S.; Sharma, V.; Bharti, P.S.; Rani, K.; Modi, G.P.; Nikolajeff, F.; Kumar, S. The Evolving Landscape of Exosomes in Neurodegenerative Diseases: Exosomes Characteristics and a Promising Role in Early Diagnosis. Int. J. Mol. Sci. 2021, 22, 440. [Google Scholar] [CrossRef]
  78. Yee, N.S.; Zhang, S.; He, H.Z.; Zheng, S.Y. Extracellular Vesicles as Potential Biomarkers for Early Detection and Diagnosis of Pancreatic Cancer. Biomedicines 2020, 8, 581. [Google Scholar] [CrossRef]
  79. Zhang, L.; Gu, C.; Wen, J.; Liu, G.; Liu, H.; Li, L. Recent advances in nanomaterial-based biosensors for the detection of exosomes. Anal. Bioanal. Chem. 2021, 413, 83–102. [Google Scholar] [CrossRef]
  80. Happel, C.; Ganguly, A.; Tagle, D.A. Extracellular RNAs as potential biomarkers for cancer. J. Cancer Metastasis Treat 2020, 6, 32. [Google Scholar] [CrossRef]
  81. Yu, D.; Li, Y.; Wang, M.; Gu, J.; Xu, W.; Cai, H.; Fang, X.; Zhang, X. Exosomes as a new frontier of cancer liquid biopsy. Mol. Cancer 2022, 21, 56. [Google Scholar] [CrossRef] [PubMed]
  82. Dow, R.; Ridger, V. Neutrophil microvesicles and their role in disease. Int. J. Biochem. Cell Biol. 2021, 141, 106097. [Google Scholar] [CrossRef] [PubMed]
  83. Kato, T.; Vykoukal, J.V.; Fahrmann, J.F.; Hanash, S. Extracellular Vesicles in Lung Cancer: Prospects for Diagnostic and Therapeutic Applications. Cancers 2021, 13, 4604. [Google Scholar] [CrossRef] [PubMed]
  84. Ayyar, K.K.; Moss, A.C. Exosomes in Intestinal Inflammation. Front. Pharmacol. 2021, 12, 658505. [Google Scholar] [CrossRef] [PubMed]
  85. Bhatt, S.; Kanoujia, J.; Dhar, A.K.; Arumugam, S.; Silva, A.K.A.; Mishra, N. Exosomes: A Novel Therapeutic Paradigm for the Treatment of Depression. Curr. Drug Targets 2021, 22, 183–191. [Google Scholar] [CrossRef]
  86. Nannan, L.; Oudart, J.B.; Monboisse, J.C.; Ramont, L.; Brassart-Pasco, S.; Brassart, B. Extracellular Vesicle-Dependent Cross-Talk in Cancer-Focus on Pancreatic Cancer. Front. Oncol. 2020, 10, 1456. [Google Scholar] [CrossRef]
  87. Kandimalla, R.; Aqil, F.; Tyagi, N.; Gupta, R. Milk exosomes: A biogenic nanocarrier for small molecules and macromolecules to combat cancer. Am. J. Reprod. Immunol. 2021, 85, e13349. [Google Scholar] [CrossRef]
  88. Bagheri Hashkavayi, A.; Cha, B.S.; Lee, E.S.; Kim, S.; Park, K.S. Advances in Exosome Analysis Methods with an Emphasis on Electrochemistry. Anal. Chem. 2020, 92, 12733–12740. [Google Scholar] [CrossRef]
  89. Xu, Y.; Hu, Y.; Xu, S.; Liu, F.; Gao, Y. Exosomal microRNAs as Potential Biomarkers and Therapeutic Agents for Acute Ischemic Stroke: New Expectations. Front. Neurol. 2022, 12, 747380. [Google Scholar] [CrossRef]
  90. Shetgaonkar, G.G.; Marques, S.M.; DCruz, C.E.M.; Vibhavari, R.J.A.; Kumar, L.; Shirodkar, R.K. Exosomes as cell-derivative carriers in the diagnosis and treatment of central nervous system diseases. Drug Deliv. Transl. Res. 2022, 12, 1047–1079. [Google Scholar] [CrossRef]
  91. Jelski, W.; Mroczko, B. Molecular and Circulating Biomarkers of Brain Tumors. Int. J. Mol. Sci. 2021, 22, 7039. [Google Scholar] [CrossRef] [PubMed]
  92. Miao, C.; Wang, X.; Zhou, W.; Huang, J. The emerging roles of exosomes in autoimmune diseases, with special emphasis on microRNAs in exosomes. Pharmacol. Res. 2021, 169, 105680. [Google Scholar] [CrossRef] [PubMed]
  93. Bunda, S.; Zuccato, J.A.; Voisin, M.R.; Wang, J.Z.; Nassiri, F.; Patil, V.; Mansouri, S.; Zadeh, G. Liquid Biomarkers for Improved Diagnosis and Classification of CNS Tumors. Int. J. Mol. Sci. 2021, 22, 4548. [Google Scholar] [CrossRef] [PubMed]
  94. Jones, J.; Nguyen, H.; Drummond, K.; Morokoff, A. Circulating Biomarkers for Glioma: A Review. Neurosurgery 2021, 88, E221–E230. [Google Scholar] [CrossRef]
  95. Cao, J.; Zhang, M.; Xie, F.; Lou, J.; Zhou, X.; Zhang, L.; Fang, M.; Zhou, F. Exosomes in head and neck cancer: Roles, mechanisms and applications. Cancer Lett. 2020, 494, 7–16. [Google Scholar] [CrossRef]
  96. Chen, A.; Wang, H.; Su, Y.; Zhang, C.; Qiu, Y.; Zhou, Y.; Wan, Y.; Hu, B.; Li, Y. Exosomes: Biomarkers and Therapeutic Targets of Diabetic Vascular Complications. Front. Endocrinol. 2021, 12, 720466. [Google Scholar] [CrossRef]
  97. Li, J.; Guan, X.; Fan, Z.; Ching, L.M.; Li, Y.; Wang, X.; Cao, W.M.; Liu, D.X. Non-Invasive Biomarkers for Early Detection of Breast Cancer. Cancers 2020, 12, 2767. [Google Scholar] [CrossRef]
  98. Sun, F.; Xu, W.; Qian, H. The emerging role of extracellular vesicles in retinal diseases. Am. J. Transl. Res. 2021, 13, 13227–13245. [Google Scholar]
  99. Chen, H.; Wang, L.; Zeng, X.; Schwarz, H.; Nanda, H.S.; Peng, X.; Zhou, Y. Exosomes, a New Star for Targeted Delivery. Front. Cell Dev. Biol. 2021, 9, 751079. [Google Scholar] [CrossRef]
  100. Hur, J.Y.; Lee, K.Y. Characteristics and Clinical Application of Extracellular Vesicle-Derived DNA. Cancers 2021, 13, 3827. [Google Scholar] [CrossRef]
  101. Tang, X.H.; Guo, T.; Gao, X.Y.; Wu, X.L.; Xing, X.F.; Ji, J.F.; Li, Z.Y. Exosome-derived noncoding RNAs in gastric cancer: Functions and clinical applications. Mol. Cancer 2021, 20, 99. [Google Scholar] [CrossRef] [PubMed]
  102. Yuan, F.; Li, Y.M.; Wang, Z. Preserving extracellular vesicles for biomedical applications: Consideration of storage stability before and after isolation. Drug Deliv. 2021, 28, 1501–1509. [Google Scholar] [CrossRef] [PubMed]
  103. Buschmann, D.; Mussack, V.; Byrd, J.B. Separation, characterization, and standardization of extracellular vesicles for drug delivery applications. Adv. Drug Deliv. Rev. 2021, 174, 348–368. [Google Scholar] [CrossRef] [PubMed]
  104. Xue, D.; Han, J.; Liu, Y.; Tuo, H.; Peng, Y. Current perspectives on exosomes in the diagnosis and treatment of hepatocellular carcinoma (review). Cancer Biol. Ther. 2021, 22, 279–290. [Google Scholar] [CrossRef] [PubMed]
  105. Xi, X.M.; Xia, S.J.; Lu, R. Drug loading techniques for exosome-based drug delivery systems. Pharmazie 2021, 76, 61–67. [Google Scholar] [CrossRef] [PubMed]
  106. Modani, S.; Tomar, D.; Tangirala, S.; Sriram, A.; Mehra, N.K.; Kumar, R.; Khatri, D.K.; Singh, P.K. An updated review on exosomes: Biosynthesis to clinical applications. J. Drug Target 2021, 29, 925–940. [Google Scholar] [CrossRef] [PubMed]
  107. Pi, Y.N.; Xia, B.R.; Jin, M.Z.; Jin, W.L.; Lou, G. Exosomes: Powerful weapon for cancer nano-immunoengineering. Biochem. Pharmacol. 2021, 186, 114487. [Google Scholar] [CrossRef]
  108. Ghafourian, M.; Mahdavi, R.; Akbari Jonoush, Z.; Sadeghi, M.; Ghadiri, N.; Farzaneh, M.; Mousavi Salehi, A. The implications of exosomes in pregnancy: Emerging as new diagnostic markers and therapeutics targets. Cell Commun. Signal 2022, 20, 51. [Google Scholar] [CrossRef]
  109. Li, J.; Li, Y.; Li, P.; Zhang, Y.; Du, L.; Wang, Y.; Zhang, C.; Wang, C. Exosome detection via surface-enhanced Raman spectroscopy for cancer diagnosis. Acta. Biomater. 2022, S1742-7061, 00174. [Google Scholar] [CrossRef]
  110. Weng, Q.; Wang, Y.; Xie, Y.; Yu, X.; Zhang, S.; Ge, J.; Li, Z.; Ye, G.; Guo, J. Extracellular vesicles-associated tRNA-derived fragments (tRFs): Biogenesis, biological functions, and their role as potential biomarkers in human diseases. J. Mol. Med. 2022, in press. [CrossRef]
  111. Boussadia, Z.; Gambardella, A.R.; Mattei, F.; Parolini, I. Acidic and Hypoxic Microenvironment in Melanoma: Impact of Tumour Exosomes on Disease Progression. Cells 2021, 10, 3311. [Google Scholar] [CrossRef] [PubMed]
  112. Zhang, N.; He, F.; Li, T.; Chen, J.; Jiang, L.; Ouyang, X.P.; Zuo, L. Role of Exosomes in Brain Diseases. Front. Cell Neurosci. 2021, 15, 743353. [Google Scholar] [CrossRef] [PubMed]
  113. Szwedowicz, U.; Łapińska, Z.; Gajewska-Naryniecka, A.; Choromańska, A. Exosomes and Other Extracellular Vesicles with High Therapeutic Potential: Their Applications in Oncology, Neurology, and Dermatology. Molecules 2022, 27, 1303. [Google Scholar] [CrossRef] [PubMed]
  114. Ramírez-Hernández, A.A.; Velázquez-Enríquez, J.M.; Santos-Álvarez, J.C.; López-Martínez, A.; Reyes-Jiménez, E.; Carrasco-Torres, G.; González-García, K.; Vásquez-Garzón, V.R.; Baltierrez-Hoyos, R. The Role of Extracellular Vesicles in Idiopathic Pulmonary Fibrosis Progression: An Approach on Their Therapeutics Potential. Cells 2022, 11, 630. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, W.; Huang, P.; Lin, J.; Zeng, H. The Role of Extracellular Vesicles in Osteoporosis: A Scoping Review. Membranes 2022, 12, 324. [Google Scholar] [CrossRef]
  116. González-Félix, M.A.; Mejía-Manzano, L.A.; González-Valdez, J. Biological nanoparticles: Relevance as novel target drug delivery systems and leading chromatographic isolation approaches. Electrophoresis 2022, 43, 109–118. [Google Scholar] [CrossRef]
  117. Prieto-Vila, M.; Yoshioka, Y.; Ochiya, T. Biological Functions Driven by mRNAs Carried by Extracellular Vesicles in Cancer. Front. Cell Dev. Biol. 2021, 9, 620498. [Google Scholar] [CrossRef]
  118. Dai, J.; Shupp, A.B.; Bussard, K.M.; Keller, E.T. Extracellular Vesicles and Bone-Associated Cancer. Curr. Osteoporos. Rep. 2021, 19, 223–229. [Google Scholar] [CrossRef]
  119. Saheera, S.; Potnuri, A.G.; Krishnamurthy, P. Nano-Vesicle (Mis)Communication in Senescence-Related Pathologies. Cells 2020, 9, 1974. [Google Scholar] [CrossRef]
  120. Sun, H.; Burrola, S.; Wu, J.; Ding, W.Q. Extracellular Vesicles in the Development of Cancer Therapeutics. Int J Mol Sci. 2020, 21, 6097. [Google Scholar] [CrossRef]
  121. Battistelli, M.; Falcieri, E. Apoptotic Bodies: Particular Extracellular Vesicles Involved in Intercellular Communication. Biology 2020, 9, 21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Zifkos, K.; Dubois, C.; Schäfer, K. Extracellular Vesicles and Thrombosis: Update on the Clinical and Experimental Evidence. Int. J. Mol. Sci. 2021, 22, 9317. [Google Scholar] [CrossRef] [PubMed]
  123. Aires, I.D.; Ribeiro-Rodrigues, T.; Boia, R.; Ferreira-Rodrigues, M.; Girão, H.; Ambrósio, A.F.; Santiago, A.R. Microglial Extracellular Vesicles as Vehicles for Neurodegeneration Spreading. Biomolecules 2021, 11, 770. [Google Scholar] [CrossRef] [PubMed]
  124. Tesfaye, D.; Menjivar, N.; Gebremedhn, S. Current knowledge and the future potential of extracellular vesicles in mammalian reproduction. Reprod. Fertil. Dev. 2021, 34, 174–189. [Google Scholar] [CrossRef]
  125. de Freitas, R.C.C.; Hirata, R.D.C.; Hirata, M.H.; Aikawa, E. Circulating Extracellular Vesicles As Biomarkers and Drug Delivery Vehicles in Cardiovascular Diseases. Biomolecules 2021, 11, 388. [Google Scholar] [CrossRef]
  126. Liu, T.; Hooda, J.; Atkinson, J.M.; Whiteside, T.L.; Oesterreich, S.; Lee, A.V. Exosomes in Breast Cancer—Mechanisms of Action and Clinical Potential. Mol. Cancer Res. 2021, 19, 935–945. [Google Scholar] [CrossRef]
  127. Mukherjee, A.; Bisht, B.; Dutta, S.; Paul, M.K. Current advances in the use of exosomes, liposomes, and bioengineered hybrid nanovesicles in cancer detection and therapy. Acta. Pharmacol. Sin. 2022, in press. [CrossRef]
  128. Chaudhari, P.; Ghate, V.; Nampoothiri, M.; Lewis, S. Multifunctional role of exosomes in viral diseases: From transmission to diagnosis and therapy. Cell Signal 2022, 94, 110325. [Google Scholar] [CrossRef]
  129. Ngu, A.; Wang, S.; Wang, H.; Khanam, A.; Zempleni, J. Milk exosomes in nutrition and drug delivery. Am. J. Physiol. Cell Physiol. 2022, in press. [CrossRef]
  130. Chew, B.C.; Liew, F.F.; Tan, H.W.; Chung, I. Chemical Advances in Therapeutic Application of Exosomes and Liposomes. Curr. Med. Chem. 2022, in press. [CrossRef]
  131. Ferreira, D.; Moreira, J.N.; Rodrigues, L.R. New advances in exosome-based targeted drug delivery systems. Crit. Rev. Oncol. Hematol. 2022, 172, 103628. [Google Scholar] [CrossRef] [PubMed]
  132. Guo, Y.; Zhai, Y.; Wu, L.; Wang, Y.; Wu, P.; Xiong, L. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Pleiotropic Impacts on Breast Cancer Occurrence, Development, and Therapy. Int. J. Mol. Sci. 2022, 23, 2927. [Google Scholar] [CrossRef] [PubMed]
  133. Jasim, S.A.; Yumashev, A.V.; Abdelbasset, W.K.; Margiana, R.; Markov, A.; Suksatan, W.; Pineda, B.; Thangavelu, L.; Ahmadi, S.H. Shining the light on clinical application of mesenchymal stem cell therapy in autoimmune diseases. Stem Cell Res. Ther. 2022, 13, 101. [Google Scholar] [CrossRef] [PubMed]
  134. Zha, K.; Tian, Y.; Panayi, A.C.; Mi, B.; Liu, G. Recent Advances in Enhancement Strategies for Osteogenic Differentiation of Mesenchymal Stem Cells in Bone Tissue Engineering. Front. Cell Dev. Biol. 2022, 10, 824812. [Google Scholar] [CrossRef]
  135. Luby, A.O.; Ranganathan, K.; Lynn, J.V.; Nelson, N.S.; Donneys, A.; Buchman, S.R. Stem Cells for Bone Regeneration: Current State and Future Directions. J. Craniofac. Surg. 2019, 30, 730–735. [Google Scholar] [CrossRef]
  136. Xie, J.; Li, X.; Zhang, Y.; Tang, T.; Chen, G.; Mao, H.; Gu, Z.; Yang, J. VE-cadherin-based matrix promoting the self-reconstruction of pro-vascularization microenvironments and endothelial differentiation of human mesenchymal stem cells. J. Mater. Chem. B. 2021, 9, 3357–3370. [Google Scholar] [CrossRef]
  137. Yamada, Y.; Minatoguchi, S.; Kanamori, H.; Mikami, A.; Okura, H.; Dezawa, M.; Minatoguchi, S. Stem cell therapy for acute myocardial infarction—focusing on the comparison between Muse cells and mesenchymal stem cells. J. Cardiol. 2021, S0914-5087, 00309-9. [Google Scholar] [CrossRef]
  138. Koliaraki, V.; Prados, A.; Armaka, M.; Kollias, G. The mesenchymal context in inflammation, immunity and cancer. Nat. Immunol. 2020, 21, 974–982. [Google Scholar] [CrossRef]
  139. Tao, J.; Cao, X.; Yu, B.; Qu, A. Vascular Stem/Progenitor Cells in Vessel Injury and Repair. Front. Cardiovasc. Med. 2022, 9, 845070. [Google Scholar] [CrossRef]
  140. Sameri, S.; Samadi, P.; Dehghan, R.; Salem, E.; Fayazi, N.; Amini, R. Stem Cell Aging in Lifespan and Disease: A State-of-the-Art Review. Curr. Stem Cell Res. Ther. 2020, 15, 362–378. [Google Scholar] [CrossRef]
  141. Chatterjee, C.; Schertl, P.; Frommer, M.; Ludwig-Husemann, A.; Mohra, A.; Dilger, N.; Naolou, T.; Meermeyer, S.; Bergmann, T.C.; Alonso Calleja, A.; et al. Rebuilding the hematopoietic stem cell niche: Recent developments and future prospects. Acta. Biomater. 2021, 132, 129–148. [Google Scholar] [CrossRef] [PubMed]
  142. Sophie, S.; Yves, B.; Frédéric, B. Current Status and Perspectives of Allogeneic Hematopoietic Stem Cell Transplantation in Elderly Patients with Acute Myeloid Leukemia. Stem Cells Transl. Med. 2022, 11, 461–477. [Google Scholar] [CrossRef] [PubMed]
  143. Barisic, S.; Childs, R.W. Graft-Versus-Solid-Tumor Effect: From Hematopoietic Stem Cell Transplantation to Adoptive Cell Therapies. Stem Cells 2022, sxac021. [Google Scholar] [CrossRef] [PubMed]
  144. Tucci, F.; Galimberti, S.; Naldini, L.; Valsecchi, M.G.; Aiuti, A. A systematic review and meta-analysis of gene therapy with hematopoietic stem and progenitor cells for monogenic disorders. Nat. Commun. 2022, 13, 1315. [Google Scholar] [CrossRef]
  145. Huang, X.; Guo, B. Update on preclinical and clinical efforts on ex-vivo expansion of hematopoietic stem and progenitor cells. Curr. Opin. Hematol. 2022, in press. [CrossRef]
  146. Akkawi, I.; Draghetti, M.; Zmerly, H. Minimally manipulated adipose derived mesenchymal stromal cells and osteoarthritis: A narrative review. Acta. Biomed. 2022, 93, e2022135. [Google Scholar] [CrossRef]
  147. Gorodetsky, R.; Aicher, W.K. Allogenic Use of Human Placenta-Derived Stromal Cells as a Highly Active Subtype of Mesenchymal Stromal Cells for Cell-Based Therapies. Int. J. Mol. Sci. 2021, 22, 5302. [Google Scholar] [CrossRef]
  148. Ahani-Nahayati, M.; Niazi, V.; Moradi, A.; Pourjabbar, B.; Roozafzoon, R.; Keshel, S.H.; Baradaran-Rafii, A. Umbilical Cord Mesenchymal Stem/Stromal Cells Potential to Treat Organ Disorders; An Emerging Strategy. Curr. Stem Cell Res. Ther. 2022, 17, 126–146. [Google Scholar] [CrossRef]
  149. Li, N.; Gao, J.; Mi, L.; Zhang, G.; Zhang, L.; Zhang, N.; Huo, R.; Hu, J.; Xu, K. Synovial membrane mesenchymal stem cells: Past life, current situation, and application in bone and joint diseases. Stem Cell Res. Ther. 2020, 11, 381. [Google Scholar] [CrossRef]
  150. Ledesma-Martínez, E.; Mendoza-Núñez, V.M.; Santiago-Osorio, E. Mesenchymal Stem Cells Derived from Dental Pulp: A Review. Stem Cells Int. 2016, 2016, 4709572. [Google Scholar] [CrossRef] [Green Version]
  151. Khazaei, S.; Keshavarz, G.; Bozorgi, A.; Nazari, H.; Khazaei, M. Adipose tissue-derived stem cells: A comparative review on isolation, culture, and differentiation methods. Cell Tissue Bank 2022, 23, 1–16. [Google Scholar] [CrossRef] [PubMed]
  152. Sasaki, R.; Watanabe, Y.; Yamato, M.; Okamoto, T. Tissue-engineered nerve guides with mesenchymal stem cells in the facial nerve regeneration. Neurochem. Int. 2021, 148, 105062. [Google Scholar] [CrossRef] [PubMed]
  153. Wang, F.; Tang, H.; Zhu, J.; Zhang, J.H. Transplanting Mesenchymal Stem Cells for Treatment of Ischemic Stroke. Cell Transplant. 2018, 27, 1825–1834. [Google Scholar] [CrossRef]
  154. Ignat, S.R.N.; Gharbia, S.; Hermenean, A.; Dinescu, S.; Costache, M. Regenerative Potential of Mesenchymal Stem Cells’ (MSCs) Secretome for Liver Fibrosis Therapies. Int. J. Mol. Sci. 2021, 22, 13292. [Google Scholar] [CrossRef]
  155. Harrell, C.R.; Djonov, V.; Volarevic, V. The Cross-Talk between Mesenchymal Stem Cells and Immune Cells in Tissue Repair and Regeneration. Int. J. Mol. Sci. 2021, 22, 2472. [Google Scholar] [CrossRef]
  156. Krawczenko, A.; Klimczak, A. Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells and Their Contribution to Angiogenic Processes in Tissue Regeneration. Int. J. Mol. Sci. 2022, 23, 2425. [Google Scholar] [CrossRef]
  157. Cheng, X.; Jiang, M.; Long, L.; Meng, J. Potential roles of mesenchymal stem cells and their exosomes in the treatment of COVID-19. Front. Biosci. 2021, 26, 948–961. [Google Scholar] [CrossRef]
  158. Zhao, L.; Hu, C.; Zhang, P.; Jiang, H.; Chen, J. Preconditioning strategies for improving the survival rate and paracrine ability of mesenchymal stem cells in acute kidney injury. J. Cell Mol. Med. 2019, 23, 720–730. [Google Scholar] [CrossRef]
  159. Ortiz, A.C.; Fideles, S.O.M.; Pomini, K.T.; Bellini, M.Z.; Pereira, E.S.B.M.; Reis, C.H.B.; Pilon, J.P.G.; de Marchi, M.Â.; Trazzi, B.F.M.; da Silva, W.S.; et al. Potential of Fibrin Glue and Mesenchymal Stem Cells (MSCs) to Regenerate Nerve Injuries: A Systematic Review. Cells 2022, 11, 221. [Google Scholar] [CrossRef]
  160. Nagaya, N.; Kangawa, K.; Itoh, T.; Iwase, T.; Murakami, S.; Miyahara, Y.; Fujii, T.; Uematsu, M.; Ohgushi, H.; Yamagishi, M.; et al. Transplantation of Mesenchymal Stem Cells Improves Cardiac Function in a Rat Model of Dilated Cardiomyopathy. Circulation 2005, 112, 1128–1135. [Google Scholar] [CrossRef] [Green Version]
  161. Boccardo, S.; Gaudiello, E.; Melly, L.; Cerino, G.; Ricci, D.; Martin, I.; Eckstein, F.; Banfi, A.; Marsano, A. Engineered mesenchymal cell-based patches as controlled VEGF delivery systems to induce extrinsic angiogenesis. Acta. Biomater. 2016, 42, 127–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Tang, J.; Wang, J.; Yang, J.; Kong, X.; Zheng, F.; Guo, L.; Zhang, L.; Huang, Y. Mesenchymal stem cells over-expressing SDF-1 promote angiogenesis and improve heart function in experimental myocardial infarction in rats. Eur. J. Cardiothorac. Surg. 2009, 36, 644–650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Gao, F.; He, T.; Wang, H.; Yu, S.; Yi, D.; Liu, W.; Cai, Z. A promising strategy for the treatment of ischemic heart disease: Mesenchymal stem cell-mediated vascular endothelial growth factor gene transfer in rats. Can. J. Cardiol. 2007, 23, 891–898. [Google Scholar] [CrossRef] [Green Version]
  164. Aboalola, D.; Han, V.K.M. Different Effects of Insulin-Like Growth Factor-1 and Insulin-Like Growth Factor-2 on Myogenic Differentiation of Human Mesenchymal Stem Cells. Stem Cells Int. 2017, 2017, 8286248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Witt, R.; Weigand, A.; Boos, A.M.; Cai, A.; Dippold, D.; Boccaccini, A.R.; Schubert, D.W.; Hardt, M.; Lange, C.; Arkudas, A.; et al. Mesenchymal stem cells and myoblast differentiation under HGF and IGF-1 stimulation for 3D skeletal muscle tissue engineering. BMC Cell Biol. 2017, 18, 15. [Google Scholar] [CrossRef] [Green Version]
  166. Choudhery, M.S.; Mahmood, R.; Harris, D.T.; Ahmad, F.J. Minimum criteria for defining induced mesenchymal stem cells. Cell Biol Int. 2022, in press. [CrossRef]
  167. Hatakeyama, M.; Ninomiya, I.; Otsu, Y.; Omae, K.; Kimura, Y.; Onodera, O.; Fukushima, M.; Shimohata, T.; Kanazawa, M. Cell Therapies under Clinical Trials and Polarized Cell Therapies in Pre-Clinical Studies to Treat Ischemic Stroke and Neurological Diseases: A Literature Review. Int. J. Mol. Sci. 2020, 21, 6194. [Google Scholar] [CrossRef]
  168. Gomes, A.; Coelho, P.; Soares, R.; Costa, R. Human umbilical cord mesenchymal stem cells in type 2 diabetes mellitus: The emerging therapeutic approach. Cell Tissue Res. 2021, 385, 497–518. [Google Scholar] [CrossRef]
  169. Rautiainen, S.; Laaksonen, T.; Koivuniemi, R. Angiogenic Effects and Crosstalk of Adipose-Derived Mesenchymal Stem/Stromal Cells and Their Extracellular Vesicles with Endothelial Cells. Int. J. Mol. Sci. 2021, 22, 10890. [Google Scholar] [CrossRef]
  170. Oliva, J. Therapeutic Properties of Mesenchymal Stem Cell on Organ Ischemia-Reperfusion Injury. Int. J. Mol. Sci. 2019, 20, 5511. [Google Scholar] [CrossRef] [Green Version]
  171. Kwon, D.G.; Kim, M.K.; Jeon, Y.S.; Nam, Y.C.; Park, J.S.; Ryu, D.J. State of the Art: The Immunomodulatory Role of MSCs for Osteoarthritis. Int. J. Mol. Sci. 2022, 23, 1618. [Google Scholar] [CrossRef] [PubMed]
  172. Trzyna, A.; Banaś-Ząbczyk, A. Adipose-Derived Stem Cells Secretome and Its Potential Application in “Stem Cell-Free Therapy”. Biomolecules 2021, 11, 878. [Google Scholar] [CrossRef] [PubMed]
  173. Beane, O.S.; Fonseca, V.C.; Cooper, L.L.; Koren, G.; Darling, E.M. Impact of aging on the regenerative properties of bone marrow-, muscle-, and adipose-derived mesenchymal stem/stromal cells. PLoS ONE 2014, 9, e115963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Tanimoto, K.; Matsumoto, T.; Nagaoka, Y.; Kazama, T.; Yamamoto, C.; Kano, K.; Nagaoka, M.; Saito, S.; Tokuhashi, Y.; Nakanishi, K. Phenotypic and functional properties of dedifferentiated fat cells derived from infrapatellar fat pad. Regen. Ther. 2022, 19, 35–46. [Google Scholar] [CrossRef]
  175. Ock, S.A.; Baregundi Subbarao, R.; Lee, Y.M.; Lee, J.H.; Jeon, R.H.; Lee, S.L.; Park, J.K.; Hwang, S.C.; Rho, G.J. Comparison of Immunomodulation Properties of Porcine Mesenchymal Stromal/Stem Cells Derived from the Bone Marrow, Adipose Tissue, and Dermal Skin Tissue. Stem Cells Int. 2016, 2016, 9581350. [Google Scholar] [CrossRef]
  176. Eirin, A.; Zhu, X.Y.; Puranik, A.S.; Tang, H.; McGurren, K.A.; van Wijnen, A.J.; Lerman, A.; Lerman, L.O. Mesenchymal stem cell-derived extracellular vesicles attenuate kidney inflammation. Kidney Int. 2017, 92, 114–124. [Google Scholar] [CrossRef]
  177. Han, J.W.; Choi, D.; Lee, M.Y.; Huh, Y.H.; Yoon, Y.S. Bone Marrow-Derived Mesenchymal Stem Cells Improve Diabetic Neuropathy by Direct Modulation of Both Angiogenesis and Myelination in Peripheral Nerves. Cell Transplant. 2016, 25, 313–326. [Google Scholar] [CrossRef] [Green Version]
  178. Shibata, T.; Naruse, K.; Kamiya, H.; Kozakae, M.; Kondo, M.; Yasuda, Y.; Nakamura, N.; Ota, K.; Tosaki, T.; Matsuki, T.; et al. Transplantation of bone marrow-derived mesenchymal stem cells improves diabetic polyneuropathy in rats. Diabetes 2008, 57, 3099–3107. [Google Scholar] [CrossRef] [Green Version]
  179. Zhou, L.N.; Wang, J.C.; Zilundu, P.L.M.; Wang, Y.Q.; Guo, W.P.; Zhang, S.X.; Luo, H.; Zhou, J.H.; Deng, R.D.; Chen, D.F. A comparison of the use of adipose-derived and bone marrow-derived stem cells for peripheral nerve regeneration In Vitro and In Vivo. Stem Cell Res. Ther. 2020, 11, 153. [Google Scholar] [CrossRef] [Green Version]
  180. Shariati, A.; Nemati, R.; Sadeghipour, Y.; Yaghoubi, Y.; Baghbani, R.; Javidi, K.; Zamani, M.; Hassanzadeh, A. Mesenchymal stromal cells (MSCs) for neurodegenerative disease: A promising frontier. Eur. J. Cell Biol. 2020, 99, 151097. [Google Scholar] [CrossRef] [PubMed]
  181. Ulpiano, C.; da Silva, C.; Monteiro, G.A. Mesenchymal Stromal Cells (MSCs): A Promising Tool for Cell-Based Angiogenic Therapy. Curr. Gene Ther. 2021, 21, 382–405. [Google Scholar] [CrossRef] [PubMed]
  182. Yang, C.; Sun, J.; Tian, Y.; Li, H.; Zhang, L.; Yang, J.; Wang, J.; Zhang, J.; Yan, S.; Xu, D. Immunomodulatory Effect of MSCs and MSCs-Derived Extracellular Vesicles in Systemic Lupus Erythematosus. Front. Immunol. 2021, 12, 714832. [Google Scholar] [CrossRef] [PubMed]
  183. Hu, C.; Wu, Z.; Li, L. Pre-treatments enhance the therapeutic effects of mesenchymal stem cells in liver diseases. J. Cell Mol. Med. 2020, 24, 40–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Gonzalez-Vilchis, R.A.; Piedra-Ramirez, A.; Patiño-Morales, C.C.; Sanchez-Gomez, C.; Beltran-Vargas, N.E. Sources, Characteristics, and Therapeutic Applications of Mesenchymal Cells in Tissue Engineering. Tissue Eng. Regen. Med. 2022, 19, 325–361. [Google Scholar] [CrossRef]
  185. Freitag, J.; Wickham, J.; Shah, K.; Li, D.; Norsworthy, C.; Tenen, A. Mesenchymal stem cell therapy combined with arthroscopic abrasion arthroplasty regenerates cartilage in patients with severe knee osteoarthritis: A case series. Regen. Med. 2020, 15, 1957–1977. [Google Scholar] [CrossRef]
  186. Tsuchiya, A.; Takeuchi, S.; Watanabe, T.; Yoshida, T.; Nojiri, S.; Ogawa, M.; Terai, S. Mesenchymal stem cell therapies for liver cirrhosis: MSCs as “conducting cells” for improvement of liver fibrosis and regeneration. Inflamm. Regen. 2019, 39, 18. [Google Scholar] [CrossRef] [Green Version]
  187. Wang, D.; Zhang, H.; Liang, J.; Wang, H.; Hua, B.; Feng, X.; Gilkeson, G.S.; Farge, D.; Shi, S.; Sun, L. A Long-Term Follow-Up Study of Allogeneic Mesenchymal Stem/Stromal Cell Transplantation in Patients with Drug-Resistant Systemic Lupus Erythematosus. Stem Cell Rep. 2018, 10, 933–941. [Google Scholar] [CrossRef] [Green Version]
  188. Zhou, X.; Jin, N.; Wang, F.; Chen, B. Mesenchymal stem cells: A promising way in therapies of graft-versus-host disease. Cancer Cell Int. 2020, 20, 114. [Google Scholar] [CrossRef] [Green Version]
  189. Guo, G.; Tan, Z.; Liu, Y.; Shi, F.; She, J. The therapeutic potential of stem cell-derived exosomes in the ulcerative colitis and colorectal cancer. Stem Cell Res. Ther. 2022, 13, 138. [Google Scholar] [CrossRef]
  190. Kobayashi, K.; Suzuki, K. Mesenchymal Stem/Stromal Cell-Based Therapy for Heart Failure—What Is the Best Source? Circ. J. 2018, 82, 2222–2232. [Google Scholar] [CrossRef] [Green Version]
  191. Guo, Y.; Peng, Y.; Zeng, H.; Chen, G. Progress in Mesenchymal Stem Cell Therapy for Ischemic Stroke. Stem Cells Int. 2021, 2021, 9923566. [Google Scholar] [CrossRef] [PubMed]
  192. Joyce, N.; Annett, G.; Wirthlin, L.; Olson, S.; Bauer, G.; Nolta, J.A. Mesenchymal stem cells for the treatment of neurodegenerative disease. Regen. Med. 2010, 5, 933–946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Thanunchai, M.; Hongeng, S.; Thitithanyanont, A. Mesenchymal Stromal Cells and Viral Infection. Stem Cells Int. 2015, 2015, 860950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Li, C.; Zhao, H.; Cheng, L.; Wang, B. Allogeneic vs. autologous mesenchymal stem/stromal cells in their medication practice. Cell Biosci. 2021, 11, 187. [Google Scholar] [CrossRef]
  195. Hare, J.M.; Fishman, J.E.; Gerstenblith, G.; DiFede Velazquez, D.L.; Zambrano, J.P.; Suncion, V.Y.; Tracy, M.; Ghersin, E.; Johnston, P.V.; Brinker, J.A.; et al. Comparison of allogeneic vs autologous bone marrow–derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: The POSEIDON randomized trial. JAMA 2012, 308, 2369–2379. [Google Scholar] [CrossRef]
  196. Shadmani, A.; Razmkhah, M.; Jalalpoor, M.H.; Lari, S.Y.; Eghtedari, M. Autologous Activated Omental versus Allogeneic Adipose Tissue-Derived Mesenchymal Stem Cells in Corneal Alkaline Injury: An Experimental Study. J. Curr. Ophthalmol. 2021, 33, 136–142. [Google Scholar] [CrossRef]
  197. Pan, Q.; Li, Y.; Li, Y.; Wang, H.; Kong, L.; Yang, Z.; Zhang, X.; Bai, S.; Zong, Z.; Chen, G.; et al. Local administration of allogeneic or autologous bone marrow-derived mesenchymal stromal cells enhances bone formation similarly in distraction osteogenesis. Cytotherapy 2021, 23, 590–598. [Google Scholar] [CrossRef]
  198. Bertoni, L.; Branly, T.; Jacquet, S.; Desancé, M.; Desquilbet, L.; Rivory, P.; Hartmann, D.J.; Denoix, J.M.; Audigié, F.; Galéra, P.; et al. Intra-Articular Injection of 2 Different Dosages of Autologous and Allogeneic Bone Marrow- and Umbilical Cord-Derived Mesenchymal Stem Cells Triggers a Variable Inflammatory Response of the Fetlock Joint on 12 Sound Experimental Horses. Stem Cells Int. 2019, 2019, 9431894. [Google Scholar] [CrossRef]
  199. Joswig, A.J.; Mitchell, A.; Cummings, K.J.; Levine, G.J.; Gregory, C.A.; Smith, R., 3rd; Watts, A.E. Repeated intra-articular injection of allogeneic mesenchymal stem cells causes an adverse response compared to autologous cells in the equine model. Stem Cell Res. Ther. 2017, 8, 42. [Google Scholar] [CrossRef] [Green Version]
  200. Nikitina, V.; Astrelina, T.; Nugis, V.; Ostashkin, A.; Karaseva, T.; Dobrovolskaya, E.; Usupzhanova, D.; Suchkova, Y.; Lomonosova, E.; Rodin, S.; et al. Clonal chromosomal and genomic instability during human multipotent mesenchymal stromal cells long-term culture. PLoS ONE 2018, 13, e0192445. [Google Scholar] [CrossRef] [Green Version]
  201. Coppin, L.; Sokal, E.; Stéphenne, X. Thrombogenic Risk Induced by Intravascular Mesenchymal Stem Cell Therapy: Current Status and Future Perspectives. Cells 2019, 8, 1160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Okada, K.; Miyata, T.; Sawa, Y. Insurance systems and reimbursement concerning research and development of regenerative medicine in Japan. Regen. Med. 2017, 12, 179–186. [Google Scholar] [CrossRef] [PubMed]
  203. Dilli, D.; Kılıç, E.; Yumuşak, N.; Beken, S.; Uçkan Çetinkaya, D.; Karabulut, R.; Zenciroğlu, A.L. Additive effect of mesenchymal stem cells and defibrotide in an arterial rat thrombosis model. Arch. Argent. Pediatr. 2017, 115, 249–256. [Google Scholar] [CrossRef]
  204. Park, Y.B.; Ha, C.W.; Lee, C.H.; Yoon, Y.C.; Park, Y.G. Cartilage Regeneration in Osteoarthritic Patients by a Composite of Allogeneic Umbilical Cord Blood-Derived Mesenchymal Stem Cells and Hyaluronate Hydrogel: Results from a Clinical Trial for Safety and Proof-of-Concept with 7 Years of Extended Follow-Up. Stem Cells Transl. Med. 2017, 6, 613–621. [Google Scholar] [CrossRef] [PubMed]
  205. Yuan, J.; Wei, Z.; Xu, X.; Ocansey, D.K.W.; Cai, X.; Mao, F. The Effects of Mesenchymal Stem Cell on Colorectal Cancer. Stem Cells Int. 2021, 2021, 9136583. [Google Scholar] [CrossRef]
  206. Li, Y.; Mao, A.S.; Seo, B.R.; Zhao, X.; Gupta, S.K.; Chen, M.; Han, Y.L.; Shih, T.Y.; Mooney, D.J.; Guo, M. Generation of the Compression-induced Dedifferentiated Adipocytes (CiDAs) Using Hypertonic Medium. Bio. Protoc. 2021, 11, e3920. [Google Scholar] [CrossRef] [PubMed]
  207. Weng, Z.; Zhang, B.; Wu, C.; Yu, F.; Han, B.; Li, B.; Li, L. Therapeutic roles of mesenchymal stem cell-derived extracellular vesicles in cancer. J. Hematol. Oncol. 2021, 14, 136. [Google Scholar] [CrossRef] [PubMed]
  208. Chen, L.; Qu, J.; Mei, Q.; Chen, X.; Fang, Y.; Chen, L.; Li, Y.; Xiang, C. Small extracellular vesicles from menstrual blood-derived mesenchymal stem cells (MenSCs) as a novel therapeutic impetus in regenerative medicine. Stem Cell Res. Ther. 2021, 12, 433. [Google Scholar] [CrossRef]
  209. Psaraki, A.; Ntari, L.; Karakostas, C.; Korrou-Karava, D.; Roubelakis, M.G. Extracellular vesicles derived from mesenchymal stem/stromal cells: The regenerative impact in liver diseases. Hepatology 2022, 75, 1590–1603. [Google Scholar] [CrossRef]
  210. Zhao, A.G.; Shah, K.; Cromer, B.; Sumer, H. Comparative analysis of extracellular vesicles isolated from human mesenchymal stem cells by different isolation methods and visualisation of their uptake. Exp. Cell Res. 2022, 414, 113097. [Google Scholar] [CrossRef]
  211. Nakamura, Y.; Kita, S.; Tanaka, Y.; Fukuda, S.; Obata, Y.; Okita, T.; Nishida, H.; Takahashi, Y.; Kawachi, Y.; Tsugawa-Shimizu, Y.; et al. Adiponectin Stimulates Exosome Release to Enhance Mesenchymal Stem-Cell-Driven Therapy of Heart Failure in Mice. Mol. Ther. 2020, 28, 2203–2219. [Google Scholar] [CrossRef] [PubMed]
  212. Cao, Q.; Huang, C.; Chen, X.M.; Pollock, C.A. Mesenchymal Stem Cell-Derived Exosomes: Toward Cell-Free Therapeutic Strategies in Chronic Kidney Disease. Front. Med. 2022, 9, 816656. [Google Scholar] [CrossRef] [PubMed]
  213. Sarhadi, V.K.; Daddali, R.; Seppänen-Kaijansinkko, R. Mesenchymal Stem Cells and Extracellular Vesicles in Osteosarcoma Pathogenesis and Therapy. Int. J. Mol. Sci. 2021, 22, 11035. [Google Scholar] [CrossRef] [PubMed]
  214. Joo, H.S.; Suh, J.H.; Lee, H.J.; Bang, E.S.; Lee, J.M. Current Knowledge and Future Perspectives on Mesenchymal Stem Cell-Derived Exosomes as a New Therapeutic Agent. Int. J. Mol. Sci. 2020, 21, 727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Huldani, H.; Abdalkareem Jasim, S.; Olegovich Bokov, D.; Abdelbasset, W.K.; Nader Shalaby, M.; Thangavelu, L.; Margiana, R.; Qasim, M.T. Application of extracellular vesicles derived from mesenchymal stem cells as potential therapeutic tools in autoimmune and rheumatic diseases. Int. Immunopharmacol. 2022, 106, 108634. [Google Scholar] [CrossRef]
  216. Birtwistle, L.; Chen, X.M.; Pollock, C. Mesenchymal Stem Cell-Derived Extracellular Vesicles to the Rescue of Renal Injury. Mesenchymal Stem Cell-Derived Extracellular Vesicles to the Rescue of Renal Injury. Int. J. Mol. Sci. 2021, 22, 6596. [Google Scholar] [CrossRef]
  217. Quaglia, M.; Dellepiane, S.; Guglielmetti, G.; Merlotti, G.; Castellano, G.; Cantaluppi, V. Extracellular Vesicles as Mediators of Cellular Crosstalk Between Immune System and Kidney Graft. Front. Immunol. 2020, 11, 74. [Google Scholar] [CrossRef]
  218. Ranghino, A.; Bruno, S.; Bussolati, B.; Moggio, A.; Dimuccio, V.; Tapparo, M.; Biancone, L.; Gontero, P.; Frea, B.; Camussi, G. The effects of glomerular and tubular renal progenitors and derived extracellular vesicles on recovery from acute kidney injury. Stem Cell Res. Ther. 2017, 8, 24. [Google Scholar] [CrossRef] [Green Version]
  219. Du, T.; Zhou, J.; Chen, W.X.; Zhang, X.L.; Ji, T.Y.; Liu, J.; Rong, L.; Wang, L.D.; Zhou, R.J.; Ding, D.G. Microvesicles derived from human umbilical cord mesenchymal stem cells ameliorate renal ischemia-reperfusion injury via delivery of miR-21. Cell Cycle 2020, 19, 1285–1297. [Google Scholar] [CrossRef]
  220. Collino, F.; Pomatto, M.; Bruno, S.; Lindoso, R.S.; Tapparo, M.; Sicheng, W.; Quesenberry, P.; Camussi, G. Exosome and Microvesicle-Enriched Fractions Isolated from Mesenchymal Stem Cells by Gradient Separation Showed Different Molecular Signatures and Functions on Renal Tubular Epithelial Cells. Stem Cell Rev. Rep. 2017, 13, 226–243. [Google Scholar] [CrossRef] [Green Version]
  221. Zhou, Y.; Xu, H.; Xu, W.; Wang, B.; Wu, H.; Tao, Y.; Zhang, B.; Wang, M.; Mao, F.; Yan, Y.; et al. Exosomes released by human umbilical cord mesenchymal stem cells protect against cisplatin-induced renal oxidative stress and apoptosis in vivo and in vitro. Stem Cell Res. Ther. 2013, 4, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Charles, C.J.; Li, R.R.; Yeung, T.; Mazlan, S.M.I.; Lai, R.C.; de Kleijn, D.P.V.; Lim, S.K.; Richards, A.M. Systemic Mesenchymal Stem Cell-Derived Exosomes Reduce Myocardial Infarct Size: Characterization With MRI in a Porcine Model. Front. Cardiovasc. Med. 2020, 7, 601990. [Google Scholar] [CrossRef] [PubMed]
  223. Keshtkar, S.; Azarpira, N.; Ghahremani, M.H. Mesenchymal stem cell-derived extracellular vesicles: Novel frontiers in regenerative medicine. Stem Cell Res. Ther. 2018, 9, 63. [Google Scholar] [CrossRef] [PubMed]
  224. Shi, B.; Wang, Y.; Zhao, R.; Long, X.; Deng, W.; Wang, Z. Bone marrow mesenchymal stem cell-derived exosomal miR-21 protects C-kit+ cardiac stem cells from oxidative injury through the PTEN/PI3K/Akt axis. PLoS ONE 2018, 13, e0191616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Ning, W.; Li, S.; Yang, W.; Yang, B.; Xin, C.; Ping, X.; Huang, C.; Gu, Y.; Guo, L. Blocking exosomal miRNA-153-3p derived from bone marrow mesenchymal stem cells ameliorates hypoxia-induced myocardial and microvascular damage by targeting the ANGPT1-mediated VEGF/PI3k/Akt/eNOS pathway. Cell Signal 2021, 77, 109812. [Google Scholar] [CrossRef] [PubMed]
  226. Wiest, E.F.; Zubair, A.C. Challenges of manufacturing mesenchymal stromal cell-derived extracellular vesicles in regenerative medicine. Cytotherapy 2020, 22, 606–612. [Google Scholar] [CrossRef] [PubMed]
  227. Wendt, S.; Goetzenich, A.; Goettsch, C.; Stoppe, C.; Bleilevens, C.; Kraemer, S.; Benstoem, C. Evaluation of the cardioprotective potential of extracellular vesicles—A systematic review and meta-analysis. Sci. Rep. 2018, 8, 15702. [Google Scholar] [CrossRef]
  228. Fan, B.; Chopp, M.; Zhang, Z.G.; Liu, X.S. Emerging Roles of microRNAs as Biomarkers and Therapeutic Targets for Diabetic Neuropathy. Front. Neurol. 2020, 11, 558758. [Google Scholar] [CrossRef]
  229. Xin, H.; Li, Y.; Liu, Z.; Wang, X.; Shang, X.; Cui, Y.; Zhang, Z.G.; Chopp, M. 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. [Google Scholar] [CrossRef] [Green Version]
  230. Katsuda, T.; Oki, K.; Ochiya, T. Potential application of extracellular vesicles of human adipose tissue-derived mesenchymal stem cells in Alzheimer's disease therapeutics. Methods Mol. Biol. 2015, 1212, 171–181. [Google Scholar] [CrossRef]
  231. Jeong, H.; Kim, O.J.; Oh, S.H.; Lee, S.; Lee, H.A.R.; Lee, K.O.; Lee, B.Y.; Kim, N.K. Extracellular Vesicles Released from Neprilysin Gene-Modified Human Umbilical Cord-Derived Mesenchymal Stem Cell Enhance Therapeutic Effects in an Alzheimer's Disease Animal Model. Stem Cells Int. 2021, 2021, 5548630. [Google Scholar] [CrossRef] [PubMed]
  232. Izadpanah, M.; Dargahi, L.; Ai, J.; Asgari Taei, A.; Barough, S.E.; Mowla, S.J.; Dana, G.T.; Farahmandfar, M. Extracellular Vesicles as a Neprilysin Delivery System Memory Improvement in Alzheimer's Disease. Iran. J. Pharm. Res. 2020, 19, 45–60. [Google Scholar] [CrossRef] [PubMed]
  233. Habisch, H.J.; Schmid, B.; von Arnim, C.A.; Ludolph, A.C.; Brenner, R.; Storch, A. Efficient processing of Alzheimer's disease amyloid-Beta peptides by neuroectodermally converted mesenchymal stem cells. Stem Cells Dev. 2010, 19, 629–633. [Google Scholar] [CrossRef] [PubMed]
  234. Lee, C.; Mitsialis, S.A.; Aslam, M.; Vitali, S.H.; Vergadi, E.; Konstantinou, G.; Sdrimas, K.; Fernandez-Gonzalez, A. Exosomes mediate the cytoprotective action of mesenchymal stromal cells on hypoxia-induced pulmonary hypertension. Circulation 2012, 126, 2601–2611. [Google Scholar] [CrossRef] [Green Version]
  235. Zhu, Y.G.; Feng, X.M.; Abbott, J.; Fang, X.H.; Hao, Q.; Monsel, A.; Qu, J.M.; Matthay, M.A.; Lee, J.W. Human mesenchymal stem cell microvesicles for treatment of Escherichia coli endotoxin-induced acute lung injury in mice. Stem Cells 2014, 32, 116–125. [Google Scholar] [CrossRef] [Green Version]
  236. Dutra Silva, J.; Su, Y.; Calfee, C.S.; Delucchi, K.L.; Weiss, D.; McAuley, D.F.; O’Kane, C.; Krasnodembskaya, A.D. Mesenchymal stromal cell extracellular vesicles rescue mitochondrial dysfunction and improve barrier integrity in clinically relevant models of ARDS. Eur. Respir. J. 2021, 58, 2002978. [Google Scholar] [CrossRef]
  237. Zhang, F.; Guo, J.; Zhang, Z.; Qian, Y.; Wang, G.; Duan, M.; Zhao, H.; Yang, Z.; Jiang, X. Mesenchymal stem cell-derived exosome: A tumor regulator and carrier for targeted tumor therapy. Cancer Lett. 2022, 526, 29–40. [Google Scholar] [CrossRef]
  238. Sun, Z.; Zhang, J.; Li, J.; Li, M.; Ge, J.; Wu, P.; You, B.; Qian, H. Roles of Mesenchymal Stem Cell-Derived Exosomes in Cancer Development and Targeted Therapy. Stem Cells Int. 2021, 2021, 9962194. [Google Scholar] [CrossRef]
  239. Jafarinia, M.; Alsahebfosoul, F.; Salehi, H.; Eskandari, N.; Ganjalikhani-Hakemi, M. Mesenchymal Stem Cell-Derived Extracellular Vesicles: A Novel Cell-Free Therapy. Immunol. Investig. 2020, 49, 758–780. [Google Scholar] [CrossRef]
  240. Hu, Y.; Sun, Y.; Wan, C.; Dai, X.; Wu, S.; Lo, P.C.; Huang, J.; Lovell, J.F.; Jin, H.; Yang, K. Microparticles: Biogenesis, characteristics and intervention therapy for cancers in preclinical and clinical research. J. Nanobiotechnol. 2022, 20, 189. [Google Scholar] [CrossRef]
  241. Lima, T.S.M.; Souza, W.; Geaquinto, L.R.O.; Sanches, P.L.; Stepień, E.L.; Meneses, J.; Fernández-de Gortari, E.; Meisner-Kober, N.; Himly, M.; Granjeiro, J.M.; et al. Nanomaterial Exposure, Extracellular Vesicle Biogenesis and Adverse Cellular Outcomes: A Scoping Review. Nanomaterials 2022, 12, 1231. [Google Scholar] [CrossRef] [PubMed]
  242. Rezaie, J.; Akbari, A.; Rahbarghazi, R. Inhibition of extracellular vesicle biogenesis in tumor cells: A possible way to reduce tumorigenesis. Cell Biochem. Funct. 2022, 40, 248–262. [Google Scholar] [CrossRef] [PubMed]
  243. Wang, W.; Li, M.; Chen, Z.; Xu, L.; Chang, M.; Wang, K.; Deng, C.; Gu, Y.; Zhou, S.; Shen, Y.; et al. Biogenesis and function of extracellular vesicles in pathophysiological processes of skeletal muscle atrophy. Biochem. Pharmacol. 2022, 198, 114954. [Google Scholar] [CrossRef] [PubMed]
  244. Hussain, S.; Fatima, A.; Fan, X.X.; Malik, S.I. REVIEW-The Biological importance of cells secreted Exosomes. Pak. J. Pharm. Sci. 2021, 34, 2273–2279. [Google Scholar] [PubMed]
  245. Ju, Y.; Bai, H.; Ren, L.; Zhang, L. The Role of Exosome and the ESCRT Pathway on Enveloped Virus Infection. Int. J. Mol. Sci. 2021, 22, 9060. [Google Scholar] [CrossRef] [PubMed]
  246. Tallon, C.; Hollinger, K.R.; Pal, A.; Bell, B.J.; Rais, R.; Tsukamoto, T.; Witwer, K.W.; Haughey, N.J.; Slusher, B.S. Nipping disease in the bud: nSMase2 inhibitors as therapeutics in extracellular vesicle-mediated diseases. Drug Discov. Today 2021, 26, 1656–1668. [Google Scholar] [CrossRef]
  247. Blanc, L.; Vidal, M. New insights into the function of Rab GTPases in the context of exosomal secretion. Small GTPases 2018, 9, 95–106. [Google Scholar] [CrossRef] [Green Version]
  248. David, G.; Zimmermann, P. Heparanase Involvement in Exosome Formation. Adv. Exp. Med. Biol. 2020, 1221, 285–307. [Google Scholar] [CrossRef]
  249. Lara-Barba, E.; Araya, M.J.; Hill, C.N.; Bustamante-Barrientos, F.A.; Ortloff, A.; García, C.; Galvez-Jiron, F.; Pradenas, C.; Luque-Campos, N.; Maita, G.; et al. Role of microRNA Shuttled in Small Extracellular Vesicles Derived From Mesenchymal Stem/Stromal Cells for Osteoarticular Disease Treatment. Front. Immunol. 2021, 12, 768771. [Google Scholar] [CrossRef]
  250. Liu, H.; Chen, Y.; Yin, G.; Xie, Q. Therapeutic prospects of MicroRNAs carried by mesenchymal stem cells-derived extracellular vesicles in autoimmune diseases. Life Sci. 2021, 277, 119458. [Google Scholar] [CrossRef]
  251. Loussouarn, C.; Pers, Y.M.; Bony, C.; Jorgensen, C.; Noël, D. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Regulate the Mitochondrial Metabolism via Transfer of miRNAs. Front. Immunol. 2021, 12, 623973. [Google Scholar] [CrossRef] [PubMed]
  252. Man, K.; Brunet, M.Y.; Jones, M.C.; Cox, S.C. Engineered Extracellular Vesicles: Tailored-Made Nanomaterials for Medical Applications. Nanomaterials 2020, 10, 1838. [Google Scholar] [CrossRef] [PubMed]
  253. Luan, X.; Sansanaphongpricha, K.; Myers, I.; Chen, H.; Yuan, H.; Sun, D. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta. Pharmacol. Sin. 2017, 38, 754–763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Zhao, L.; Zhang, K.; He, H.; Yang, Y.; Li, W.; Liu, T.; Li, J. The Relationship Between Mesenchymal Stem Cells and Tumor Dormancy. Front. Cell Dev. Biol. 2021, 9, 731393. [Google Scholar] [CrossRef] [PubMed]
  255. Wang, Y.; Lu, J.; Chen, L.; Bian, H.; Hu, J.; Li, D.; Xia, C.; Xu, H. Tumor-Derived EV-Encapsulated miR-181b-5p Induces Angiogenesis to Foster Tumorigenesis and Metastasis of ESCC. Mol. Ther. Nucleic Acids. 2020, 20, 421–437. [Google Scholar] [CrossRef]
  256. Tominaga, N.; Kosaka, N.; Ono, M.; Katsuda, T.; Yoshioka, Y.; Tamura, K.; Lötvall, J.; Nakagama, H.; Ochiya, T. Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood-brain barrier. Nat. Commun. 2015, 6, 6716. [Google Scholar] [CrossRef] [Green Version]
  257. Li, Y.; Hao, J.; Hu, Z.; Yang, Y.G.; Zhou, Q.; Sun, L.; Wu, J. Current status of clinical trials assessing mesenchymal stem cell therapy for graft versus host disease: A systematic review. Stem Cell Res. Ther. 2022, 13, 93. [Google Scholar] [CrossRef]
  258. Murata, M.; Teshima, T. Treatment of Steroid-Refractory Acute Graft-Versus-Host Disease Using Commercial Mesenchymal Stem Cell Products. Front. Immunol. 2021, 12, 724380. [Google Scholar] [CrossRef]
  259. Hwang, J.J.; Rim, Y.A.; Nam, Y.; Ju, J.H. Recent Developments in Clinical Applications of Mesenchymal Stem Cells in the Treatment of Rheumatoid Arthritis and Osteoarthritis. Front. Immunol. 2021, 12, 631291. [Google Scholar] [CrossRef]
  260. Lopez-Santalla, M.; Fernandez-Perez, R.; Garin, M.I. Mesenchymal Stem/Stromal Cells for Rheumatoid Arthritis Treatment: An Update on Clinical Applications. Cells 2020, 9, 1852. [Google Scholar] [CrossRef]
  261. Karamini, A.; Bakopoulou, A.; Andreadis, D.; Gkiouras, K.; Kritis, A. Therapeutic Potential of Mesenchymal Stromal Stem Cells in Rheumatoid Arthritis: A Systematic Review of In Vivo Studies. Stem Cell Rev. Rep. 2020, 16, 276–287. [Google Scholar] [CrossRef] [PubMed]
  262. El-Jawhari, J.J.; El-Sherbiny, Y.; McGonagle, D.; Jones, E. Multipotent Mesenchymal Stromal Cells in Rheumatoid Arthritis and Systemic Lupus Erythematosus; From a Leading Role in Pathogenesis to Potential Therapeutic Saviors? Front. Immunol. 2021, 12, 643170. [Google Scholar] [CrossRef] [PubMed]
  263. Buscail, E.; Le Cosquer, G.; Gross, F.; Lebrin, M.; Bugarel, L.; Deraison, C.; Vergnolle, N.; Bournet, B.; Gilletta, C.; Buscail, L. Adipose-Derived Stem Cells in the Treatment of Perianal Fistulas in Crohn’s Disease: Rationale, Clinical Results and Perspectives. Int. J. Mol. Sci. 2021, 22, 9967. [Google Scholar] [CrossRef] [PubMed]
  264. Tan, S.J.O.; Floriano, J.F.; Nicastro, L.; Emanueli, C.; Catapano, F. Novel Applications of Mesenchymal Stem Cell-derived Exosomes for Myocardial Infarction Therapeutics. Biomolecules 2020, 10, 707. [Google Scholar] [CrossRef]
  265. Chen, Y.; Shen, J.; Ke, K.; Gu, X. Clinical potential and current progress of mesenchymal stem cells for Parkinson's disease: A systematic review. Neurol. Sci. 2020, 41, 1051–1061. [Google Scholar] [CrossRef]
  266. de Araújo, L.T.; Macêdo, C.T.; Damasceno, P.K.F.; das Neves, Í.G.C.; de Lima, C.S.; Santos, G.C.; de Santana, T.A.; Sampaio, G.L.A.; Silva, D.N.; Villarreal, C.F.; et al. Clinical Trials Using Mesenchymal Stem Cells for Spinal Cord Injury: Challenges in Generating Evidence. Cells 2022, 11, 1019. [Google Scholar] [CrossRef]
Figure 1. Extracellular vesicle structure. Exosome consists of lipid bilayer membrane, including proteins such as cytoskeletal proteins (actin, myosin, vimentin, tubulin, etc.), heat shock proteins (HSP60, HSP70, HSP90, etc.), tetraspanins (CD9, CD63, CD81, etc.), cytokines (IL-1β, TNF-α, IL-6, etc.), Lamp, nucleic acids such as microRNAs, circRNAs, IncRNAs, lipid rafts such as cholesterol, ceramide, sphingomyelin, phosphatidylserine, and protein receptor such as transferrin receptor.
Figure 1. Extracellular vesicle structure. Exosome consists of lipid bilayer membrane, including proteins such as cytoskeletal proteins (actin, myosin, vimentin, tubulin, etc.), heat shock proteins (HSP60, HSP70, HSP90, etc.), tetraspanins (CD9, CD63, CD81, etc.), cytokines (IL-1β, TNF-α, IL-6, etc.), Lamp, nucleic acids such as microRNAs, circRNAs, IncRNAs, lipid rafts such as cholesterol, ceramide, sphingomyelin, phosphatidylserine, and protein receptor such as transferrin receptor.
Ijms 23 06480 g001
Figure 2. Regeneration effects of MSC-derived extracellular vesicles (MSC-EVs). MSC-EVs have different sources, including adipocytes, bone marrow, umbilical cord, pulp, etc. These MSC-EVs represent regenerative effects for heart, lung, kidney, neuron, etc. Further, MSC promotes secretion of EVs via interaction between T-cadherin receptor on the MSC and adiponectin derived from adipocytes, leading to regeneration effects of injured tissues.
Figure 2. Regeneration effects of MSC-derived extracellular vesicles (MSC-EVs). MSC-EVs have different sources, including adipocytes, bone marrow, umbilical cord, pulp, etc. These MSC-EVs represent regenerative effects for heart, lung, kidney, neuron, etc. Further, MSC promotes secretion of EVs via interaction between T-cadherin receptor on the MSC and adiponectin derived from adipocytes, leading to regeneration effects of injured tissues.
Ijms 23 06480 g002
Table 1. Clinical trials of MSC-derived Exosomes.
Table 1. Clinical trials of MSC-derived Exosomes.
#NCT NumberCondition or Disease PhaseSponsorBrief Summary
1NCT02138331Diabetes Mellitus Type 1Phase 2, Phase 3General Committee of Teaching Hospitals and Institutes, EgyptEffect of Microvesicles and Exosomes Therapy on β-cell Mass in Type I Diabetes Mellitus
2NCT03384433Cerebrovascular DisordersPhase 1, Phase 2Isfahan University of Medical Sciences, IranAllogenic Mesenchymal Stem Cell Derived Exosome in Patients With Acute Ischemic Stroke
3NCT03437759Macular HolesEarly Phase 1Tianjin Medical University, China To assess the safety and efficacy of mesenchymal stem cells (MSCs) and MSC-derived exosomes (MSC-Exos) for promoting healing of large and refractory macular holes (MHs).
4NCT03608631Metastatic Pancreatic AdenocarcinomaPhase 1M.D. Anderson Cancer Center, USiExosomes in Treating Participants With Metastatic Pancreas Cancer With KrasG12D Mutation
5NCT04173650Dystrophic Epidermolysis BullosaPhase 1, Phase 2Aegle Therapeutics, USMSC EVs in Dystrophic Epidermolysis Bullosa
6NCT04276987CoronavirusPhase 1Ruijin Hospital, ChinaA Pilot Clinical Study on Inhalation of Mesenchymal Stem Cells Exosomes Treating Severe Novel Coronavirus Pneumonia
7NCT04313647HealthyPhase 1Ruijin Hospital, ChinaA Tolerance Clinical Study on Aerosol Inhalation of Mesenchymal Stem Cells Exosomes In Healthy Volunteers
8NCT04388982Alzheimer DiseasePhase 1, Phase 2Ruijin Hospital, ChinaSafety and the Efficacy Evaluation of Allogenic Adipose MSC-Exos in Patients With Alzheimer's Disease
9NCT04491240SARS-CoV-2 PNEUMONIAPhase 2State-Financed Health Facility, Russia Evaluation of Safety and Efficiency of Method of Exosome Inhalation in SARS-CoV-2 Associated Pneumonia
10NCT04602442SARS-CoV-2 PNEUMONIAPhase 2State-Financed Health Facility, Russia Safety and Efficiency of Method of Exosome Inhalation in COVID-19 Associated Pneumonia
11NCT04747574SARS-CoV-2Phase 1Tel-Aviv Sourasky Medical Center, IsraelEvaluation of the Safety of CD24-Exosomes in Patients With COVID-19 Infection
12NCT05060107Osteoarthritis, KneePhase 1 Universidad de los Andes, ChileIntra-articular Injection of MSC-derived Exosomes in Knee Osteoarthritis
13NCT05216562SARS-CoV2 InfectionPhase 2Dermama Bioteknologi Laboratorium, IndonesiaEfficacy and Safety of EXOSOME-MSC Therapy to Reduce Hyper-inflammation In Moderate COVID-19 Patients
14NCT05261360Knee; Injury, MeniscusPhase 2Eskisehir Osmangazi University, TurkeyClinical Efficacy of Exosome in Degenerative Meniscal Injury
15NCT05402748Fistula PerianalPhase 1, Phase 2Tehran University of Medical Sciences, IranSafety and Efficacy of Injection of Human Placenta Mesenchymal Stem Cells Derived Exosomes for Treatment of Complex Anal Fistula
Searched by ClinicalTrials.gov (https://clinicaltrials.gov/ct2/home, accessed on 1 June 2022).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Matsuzaka, Y.; Yashiro, R. Therapeutic Strategy of Mesenchymal-Stem-Cell-Derived Extracellular Vesicles as Regenerative Medicine. Int. J. Mol. Sci. 2022, 23, 6480. https://doi.org/10.3390/ijms23126480

AMA Style

Matsuzaka Y, Yashiro R. Therapeutic Strategy of Mesenchymal-Stem-Cell-Derived Extracellular Vesicles as Regenerative Medicine. International Journal of Molecular Sciences. 2022; 23(12):6480. https://doi.org/10.3390/ijms23126480

Chicago/Turabian Style

Matsuzaka, Yasunari, and Ryu Yashiro. 2022. "Therapeutic Strategy of Mesenchymal-Stem-Cell-Derived Extracellular Vesicles as Regenerative Medicine" International Journal of Molecular Sciences 23, no. 12: 6480. https://doi.org/10.3390/ijms23126480

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop