Next Article in Journal
Co-Encapsulation of Curcumin and α-Tocopherol in Bicosome Systems: Physicochemical Properties and Biological Activity
Previous Article in Journal
Application of Nanomedicine in Immunotherapy: Recent Advances and Prospects
Previous Article in Special Issue
Enhancing Anticancer Efficacy of Chemotherapeutics Using Targeting Ligand-Functionalized Synthetic Antigen Receptor-Mesenchymal Stem Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Clinical Prospect of Mesenchymal Stromal/Stem Cell-Derived Extracellular Vesicles in Kidney Disease: Challenges and the Way Forward

1
Institute for the Application of Nuclear Energy (INEP), University of Belgrade, 11 000 Belgrade, Serbia
2
Department of Neurosurgery, MD Anderson Cancer Center, University of Texas, Houston, TX 770302, USA
3
Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, 45355 Essen, North Rhine-Westhpalia, Germany
4
Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work as first authors.
Pharmaceutics 2023, 15(7), 1911; https://doi.org/10.3390/pharmaceutics15071911
Submission received: 26 May 2023 / Revised: 23 June 2023 / Accepted: 7 July 2023 / Published: 9 July 2023

Abstract

:
Kidney disease is a growing public health problem worldwide, including both acute and chronic forms. Existing therapies for kidney disease target various pathogenic mechanisms; however, these therapies only slow down the progression of the disease rather than offering a cure. One of the potential and emerging approaches for the treatment of kidney disease is mesenchymal stromal/stem cell (MSC) therapy, shown to have beneficial effects in preclinical studies. In addition, extracellular vesicles (EVs) released by MSCs became a potent cell-free therapy option in various preclinical models of kidney disease due to their regenerative, anti-inflammatory, and immunomodulatory properties. However, there are scarce clinical data available regarding the use of MSC-EVs in kidney pathologies. This review article provides an outline of the renoprotective effects of MSC-EVs in different preclinical models of kidney disease. It offers a comprehensive analysis of possible mechanisms of action of MSC-EVs with an emphasis on kidney disease. Finally, on the journey toward the implementation of MSC-EVs into clinical practice, we highlight the need to establish standardized methods for the characterization of an EV-based product and investigate the adequate dosing, safety, and efficacy of MSC-EVs application, as well as the development of suitable potency assays.

1. Introduction

Kidney disease is a serious health concern worldwide with an increasing prevalence [1]. Growing evidence suggests that even minor types of kidney disease are linked to an elevated risk of developing kidney failure, as well as a higher likelihood of cardiovascular disease (CVD) and death [1]. While there have been some improvements in understanding the pathophysiology of kidney disease, current treatments are limited in their ability to halt or reverse disease progression. Therefore, the development of new strategies for treating kidney disease is a critical area of research.
Stem cell therapies have shown potential in the treatment of different diseases and have been tested in preclinical models and clinical trials. In particular, the use of extracellular vesicles (EVs) derived from stem cells has emerged as a promising new treatment option for promoting recovery from various types of injuries. In this context, EVs derived from mesenchymal stromal/stem cells (MSCs), known as MSC-EVs, have become a potent cell-free therapy for several disease states, including kidney disease [2]. MSC-EVs can modulate immune responses, promote tissue repair, and inhibit cell death [3]. Due to their therapeutic potential and the ease of harvesting, MSC-EVs are emerging as a promising new drug for the treatment of kidney disease and several other disease states.
This review article provides a summary of the renoprotective effects of MSC-EVs in various preclinical models of kidney disease and offers details on the possible mechanisms of action (MoA) of MSC-EVs. As we progress towards the clinical use of MSC-EVs, the article underscores the importance of creating standardized methods for the characterization of EV-based products, studying the appropriate dosing, safety, and efficacy of MSC-EVs administration, and developing suitable potency assays.

2. Kidney Disease: Definition and Classification

Kidney disease is described as a heterogeneous group of disorders that affect kidney structure and function [1]. An increasing amount of evidence implies that even moderate forms of kidney disease are linked to a higher risk of kidney failure, as well as the risk of cardiovascular morbidity and death [1,4]. Based on the progression of the injury, kidney diseases are stratified into acute kidney injury (AKI), chronic kidney disease (CKD), and end-stage kidney disease (ESKD).
AKI is defined as an abrupt decrease in kidney function [5] and represents a serious clinical problem. It has been reported that around 22% of adults and 34% of children across the world experience AKI during hospitalization [6]. Despite major advancements in preventive measures and supportive care, AKI continues to be linked to higher rates of morbidity and mortality [3], where mortality rates for intensive care unit patients can exceed 50% [5]. Numerous stressors including ischemia, nephrotoxic medications, environmental pollutants, nephritis, and urinary tract obstruction [5] can cause an injury of cells within the nephron, specifically kidney proximal tubular cells, leading to cell damage, inflammation, oxidative stress, apoptosis, and vascular dysfunction [7]. Depending on the degree of kidney damage, AKI can lead to regeneration and recovery of the injured tissue, or the CKD, known as AKI-to-CKD transition [8,9]. Many patients who survive their acute illness fail to recover normal kidney function and develop CKD and ESKD [10]. The relationship between AKI and CKD is very complex: AKI can lead to CKD, and CKD increases the risk of AKI [4].
CKD is known as one of the major noncommunicable diseases worldwide, and up to 10% of the world’s adult population is assumed to be affected by this condition [4], with increasing prevalence in the aging population [11,12]. According to the National Kidney Foundation’s Kidney Disease Outcomes Quality Initiative (NKF-KDOQI), CKD is classified into stages 1–5 [13]. In the early stages of CKD, 1 and 2, kidney function is still in the compensatory period with mild changes in proteinuria and glomerular filtration rate (GFR) [14]. As the disease progresses, the kidney function deteriorates and the patient enters stages 3 and 4 of CKD, with the loss of about 50% of normal kidney function [14]. Stage 5 is the last stage of CKD and is characterized by a GFR of less than 15 mL/min/1.73 m2 or the requirement for maintenance dialysis [13,14]. The main pathological changes associated with CKD include kidney interstitial fibrosis and glomerulosclerosis; while kidney fibrosis is considered to be the final manifestation of all CKDs regardless of their etiology [15,16,17].
ESKD sets up when CKD reaches an advanced stage and kidneys lose their filtering abilities (GFR < 15 mL/min/1.73 m2). Patients with ESKD have a high mortality risk, with CVD mortality accounting for 53% of deaths [18]. At this stage, renal replacement therapies (mainly hemodialysis and peritoneal dialysis) or a kidney transplant are needed to prolong patients’ lives.
Current therapeutic strategies for AKI and CKD focus on supportive care to manage complications and slow disease progression; however, they are insufficient to reverse disease progression. The development of new approaches that enable the restoration of damaged kidney tissue or prevent the advancement to CKD is crucial in improving short-term and long-term outcomes.

3. Extracellular Vesicles—A New Paradigm Shift in the MSC Therapy

Numerous studies have confirmed the beneficial effects of MSCs for the treatment of a variety of pathologies, including kidney diseases [19,20,21,22,23,24]. Nevertheless, in the MSC field, it became obvious that MSCs exert therapeutic functions via their secretome [25,26,27,28], and that the biologically active component of MSC secretome, i.e., extracellular vesicles (EVs), can be used in place of MSCs as potential therapeutics.
EVs comprise a heterogeneous group of membrane-enveloped vesicles, that are secreted by cells into the extracellular space via different routes. According to classical nomenclature, EVs are divided into three main types: (a) exosomes, formed by the invagination of the limiting membrane of the early endosome, i.e., multivesicular body (MVB) and released into the extracellular space upon fusion of MVB with the plasma membrane; (b) microvesicles (MVs), formed by outward budding and detachment from the plasma membrane; and (c) apoptotic bodies, released during programmed cell death. However, in recent years, several other types of EVs released from plasma membranes have been discovered such as migrasomes, ciliary ectosomes, secreted midbody remnants, exophers, etc., which E.I. Buzas, in her recently published paper, classified into two major categories: exosomes (originating from endosomal compartment) and ectosomes (originating from plasma membrane) [29].
EVs contain luminal, transmembrane, and membrane-associated molecules, including proteins, nucleic acids, lipids, and metabolites. The diameter of EVs falls in the nanometer range (from 30 nm to several hundreds of nanometers), with exosomes being considered as generally smaller than some classes of ectosomes (i.e., microvesicles or apoptotic bodies). The size of EVs considerably overlaps between types, rendering size an unreliable criterion for distinguishing them [29,30]. Furthermore, there are no specific markers of EV type, although some tetraspanins (e.g., CD9, CD63, CD81) are used as EV markers, with CD81 being more enriched in small EVs (including exosomes) and CD9 in larger EVs (including MVs) [31]. The lack of differential markers, as well as overlap in other physical or biochemical properties, cause a shortfall of techniques able to isolate EVs of just one type. Consequently, the International Society of Extracellular Vesicles (ISEV) recommends using the general term “extracellular vesicles” for any vesicles isolated from body fluids or cell culture media, with guidelines for the application of biomarkers for in-depth characterization [30,32,33]. However, it is important to notice that nomenclature varies throughout the literature, especially for the one published before the release of ISEV guidelines [30,34], as well as in some recent publications. The term “exosomes” is still used by some researchers and also in the industry to designate all EVs. Since this inaccuracy in the utilization of the nomenclature can lead to misinterpretation of results, it is crucial for every reader to critically evaluate methods applied for separation of EVs and biochemical characterization used in the literature sources, in order to compare results. In this review, we will use the term “extracellular vesicles” and EVs regardless of the literature source, in order to better align original data with the current understanding of EVs biology.
EVs have the ability to transfer complex information or signals from one cell to another in a targeted manner, affecting the biology and function of the recipient cell. Theoretically, EVs can transmit the information through (a) internalization of EVs by target cells through endocytosis; (b) fusion of EVs with the target cell membrane; or (c) interaction of EV´s surface molecules with the recipient cell. The mode of information transfer, the type of the EV interaction with the recipient cell, and the type of active molecules depend on the EV type and likely specific subpopulation, i.e., on their content and characteristics of the recipient cell. However, we still lack in-depth knowledge about subpopulation heterogeneity to precisely attribute specific mechanisms to each subpopulation. Thus, although there are a plethora of information about the importance of both proteins and RNA [35,36,37,38,39] in the transfer of information by EVs, there are some studies that reveal subpopulations of MSC-EVs without RNA [40] or re-evaluate the influence of RNA [41,42], emphasizing the need for further exploration of the heterogeneity of EVs and elucidation of their mechanisms using advanced methodological approaches.
Nevertheless, the transfer of information by EVs is a universal mechanism by which cells communicate and they are involved in all physiological and pathological processes investigated so far [43]. Hence, the characteristics of EVs reflect the physiological state of the originating cells, and EVs can be used as a potential novel class of biomarkers [44,45,46]. Also, EVs are both targets and tools of novel therapeutic approaches, such as anti-tumor therapy, vaccination, modulation of the immune system, and drug delivery [2,25]. A proof of concept showing that MSC-EVs can be used therapeutically in humans was first demonstrated in 2014 when they were used as a treatment for a refractory acute Graft-versus-Host disease (GvHD) patient, whose symptoms improved significantly during a 2-week-long treatment period and remained stable for more than four months [47].
MSC-EVs are also considered a good therapeutic option due to several important advantages in comparison to MSC cellular therapy. Therapeutic MSCs need to be expanded in the culture, often under different conditions, conferring phenotypic differences to the final product [48]. Possible cellular senescence may lead to a loss of therapeutic properties [49]. Certain concerns are related to the probability of cellular rejection, formation of ectopic tissue, or embolism in small lung blood vessels [50]. It is accepted that MSC-EVs can be produced in large quantities and stored for prolonged periods without significant changes in biological properties. Additionally, MSC-EVs are not self-replicating, and their small size eliminates the risk of embolism. In addition to the clinical application of EVs in GvHD patient, results have been reported for the treatment of CKD patients, a patient obtaining a cochlea implant, and severe COVID-19 patients [51,52,53]. None of these studies reported side effects, while positive impacts of the MSC-EVs application were described in all studies. MSC-EVs have been shown to mediate immunomodulatory, anti-inflammatory, and pro-regenerative activities [48], qualifying them as a potential therapeutic option for a variety of diseases.

4. Demonstrable Renoprotective Effects of MSC-EVs in Preclinical Studies of Kidney Disease

The beneficial effects of MSC-EVs in ameliorating kidney damage have been shown in various preclinical models of AKI and CKD, thus demonstrating their potential value in the treatment of kidney failure. In these studies, EVs derived from MSCs isolated from different adult and neonatal sources such as bone marrow, adipose tissue, liver, kidney, umbilical cord, and placenta, have been used (Table 1).
Bone marrow (BM)-derived MSC-EVs (BM-MSC-EVs) exert beneficial effects in the renal ischemia-reperfusion (I/R) injury (IRI) model of AKI [54,55,56] and CKD [57]. The treatment of mice or rats with BM-MSC-EVs reduced tubular lesions and fibrosis; decreased lymphocyte infiltration; and enhanced tubular cell proliferation. Meanwhile, at the functional level, they lowered blood urea nitrogen (BUN), creatinine, and proteinuria. In AKI caused by glycerol, cisplatin, or gentamicin, similar effects were noted [27,58,59,60]. Namely, BM-MSC-EVs were able to restore the brush border and increase mitochondria, inhibit necrosis, apoptosis, and inflammation, enhance proliferation and survival, as well as to reduce the BUN and creatinine levels. Beneficial effects of BM-MSC-EVs have also been demonstrated in models of CKD such as unilateral ureteral obstruction (UUO) [61,62,63,64], nephropathy induced by aristolochic acid (AAN) [65], or diabetic nephropathy (DN) induced by streptozotocin [66]. In all these models, EVs ameliorated kidney damage. In the UOO model, BM-MSC-EVs reduced tubular dilation and interstitial inflammatory cell infiltration; preserved intact cortical tubules; reduced the disruption of the tubular basement membrane and ECM deposition; preserved E-cadherin levels; and decreased serum creatinine, BUN, and proteinuria. In the AAN model, BM-MSC-EVs showed regenerative and anti-fibrotic effects, as well as the anti-inflammatory effect seen as a decrease of the infiltration of CD45 positive immune cells, fibroblasts, and pericytes, similar to in the STZ-DN model [65,66].
Table 1. MSC-EVs sources and administration routes in various preclinical models of kidney disease.
Table 1. MSC-EVs sources and administration routes in various preclinical models of kidney disease.
EV SourceIn Vivo
Model
Type of Kidney DiseaseAdministrationReference
BM-MSCsGlycerolAKIIntravenous[27]
I/R AKIIntravenous[54]
I/R AKIIntravenous[55]
CisplatinAKIIntravenous[58]
GentamicinAKIIntravenous[59]
GlycerolAKIIntravenous[60]
I/R AKIIntravenous[56]
UUOCKDIntravenous[61]
I/R CKDIntrarenal[57]
AANCKDIntravenous[65]
UUOCKDIntravenous[62]
UUOCKDIntravenous[63]
STZ-DNCKDIntravenous[66]
UUOCKDIntravenous[64]
AD-MSCsI/R AKIIntravenous[67]
SepsisAKIIntravenous[68]
MS+ASCKDIntrarenal[69,70]
I/RCKDIntravenous[71]
UUOCKDIntravenous[72]
GlycerolAKIIntravenous[73]
L-MSCsSTZ-DNCKDIntravenous[66]
AANCKDIntravenous[74]
K-MSCsI/R AKIIntravenous[75]
I/R AKIIntravenous[76]
UUOCKDIntravenous[77]
UC-MSCsCisplatinAKIKidney capsule[78]
I/R AKIIntravenous[79]
I/R AKIIntravenous[80]
I/R AKIIntravenous[81]
I/R AKIIntravenous[82]
I/R AKIIntravenous[83]
I/R AKIIntravenous[84]
CisplatinAKIKidney capsule[85]
I/R AKIIntravenous[86]
SepsisAKIIntravenous[87]
I/RAKIIntravenous[88]
UUOCKDIntravenous[89]
I/RCKDIntravenous[86]
I/RCKDIntravenous[79]
I/RCKDIntravenous[84]
P-MSCsI/R AKIIntrarenal[90]
I/R AKIIntrarenal[91]
MSCs, mesenchymal stromal/stem cells; BM-MSCs, bone marrow MSCs; UC-MSCs, umbilical cord MSCs; AD-MSCs, adipose tissue MSCs; P-MSCs, placental MSCs; K-MSCs, kidney-derived MSCs; L-MSCs, liver MSCs; UUO, unilateral ureteral obstruction; I/R, ischemia-reperfusion; AAN, aristolochic acid-induced nephropathy; STZ-DN, streptozotocin-induced diabetic nephropathy; MS+AS, metabolic syndrome, and renal artery stenosis.
Adipose tissue-derived MSCs (AD-MSCs) are another type of MSCs recognized as a valuable source of EVs potentially therapeutic for kidney diseases. Thus, in the sepsis-induced AKI model, AD-MSC-EVs managed to abrogate inflammation, apoptosis, and microcirculation disorders, lowering overall mortality [68]. In CKD, AD-MSC-EVs were proven to have beneficial effects in models of metabolic renovascular disease and metabolic syndrome with renal artery stenosis [69,70], IRI [71], and UUO [72]. In these models, AD-MSC-EVs alleviated CKD by attenuating kidney inflammation, apoptosis, oxidative stress, and fibrosis, improving medullary oxygenation and GFR. They were also able to influence kidney blood flow by lowering renal artery stenosis or improving cortical microvascular peritubular capillary density.
EVs from liver stem-like cells (HLSCs) have also been investigated for their renoprotective potential in models of glycerol-induced AKI, DN in streptozotocin-induced diabetes and AAN [66,73,74], and they elicited similar effects as EVs from other types of MSCs.
MSCs derived from the kidney itself also produce EVs with the potential to induce beneficial changes in kidney diseases. Thus, in the IRI model of AKI [75,76], those EVs induced improvement in the proliferation of peritubular capillary endothelial cells, microvascular rarefaction, reduction of ischemic damage, and overall amelioration of kidney function. In the UUO model of CKD [77], they inhibited endothelial-to-mesenchymal transition of peritubular capillaries (PTC) endothelial cells, improved PTC rarefaction, and inhibited inflammatory cell infiltration and TIF.
Umbilical cord MSC-EVs (UC-MSC-EVs) have been used in various models of kidney disease. In IRI models [79,80,81,82,83,84,86,88], UC-MSC-EVs were found to mitigate renal cell apoptosis; enhance proliferation and tubular epithelial cell growth; increase capillary vessel density; decrease epithelial cell swelling, necrosis, and casts formation; and abrogate kidney fibrosis, inflammation, and oxidative stress, thus improving kidney function. UC-MSC-EVs have also shown renoprotective effects against cisplatin-induced nephrotoxicity and were able to prevent the development of AKI [78,85]. Their application led to an increase of PCNA-positive cells; reduction of oxidative stress, apoptosis, and necrosis of renal proximal tubules; and improvement of kidney function. In the sepsis model [87], UC-MSC-EVs alleviated sepsis-associated AKI improving the survival of affected mice. UC-MSC-EVs were also applied as treatment in CKD models. In IRI models of CKD [79,84,86], UC-MSC-EVs reduced apoptosis, inflammation, and fibrosis; enhanced proliferation and the number of PCNA-positive cells; increased capillary vessel density; and improved kidney function. Similar effects were observed in the UUO model of CKD [89].
Placental MSC-EVs were tested in IRI models of AKI [90,91] where they increased the proliferation of kidney tubular epithelial cells and angiogenesis, alleviated apoptosis and fibrosis, and improved levels of serum BUN and creatinine.
As seen from the evidence above, different types of MSC-EVs have the potential to modulate various key aspects of AKI and CKD pathogenesis, improving kidney damage in different preclinical models, which makes them good candidates for the development of novel therapy for kidney disease.

5. Utilization of MSC-EVs in Clinical Trials

5.1. MSC-EVs in Clinical Trials of Kidney Disease

As seen from a variety of preclinical models, MSC-EVs emerged as a promising agent in promoting recovery and regeneration after kidney injury. However, clinical studies testing the efficacy and safety of MSC-EVs in kidney pathologies are limited. To date, there is only one study assessing the therapeutic effect of MSC-EVs in patients with CKD. Nassar et al. (2016) conducted a randomized, placebo-controlled study investigating the safety and therapeutic efficacy of EVs derived from human cord blood MSCs (CB-MSCs) in alleviating the progression of CKD stages 3 and 4 [51]. Forty patients with diagnosed CKD stages 3 and 4 were enrolled in the study. They were randomly divided into a treatment group consisting of 20 CKD patients receiving CB-MSC-EVs and a control group composed of 20 CKD patients receiving saline. EVs were administered intravenously and intra-arterial with a one-week break between two injections. The authors reported a transient improvement of kidney function in stage 3 and 4 CKD patients, followed by the amelioration of inflammatory immune reaction [51]. Besides the beneficial effects, the administration of MSC-EVs were shown to be safe for the patients enrolled in the study.

5.2. What Can We Learn from MSC´s Clinical Trials?

The therapeutic potential of MSCs has long been recognized and numerous clinical trials are underway. In kidney disease, clinical trials are being conducted in AKI and CKD, DN, lupus-related nephropathy, and kidney transplants (ClinicalTrials.gov, accessed on 25 February 2023). Due to abundant experience in the production and application of clinical/therapeutic MSCs, some common conclusions can be made, and parallels can be drawn to better understand the future directions in the production of clinical-grade EVs. Furthermore, several bottlenecks associated with MSC-derived EVs need to be addressed in order to harness the full potential of MSC-derived EVs for therapeutic application, such as standardization of isolation methods for EVs, scalability and production efficiency, heterogeneity of EV populations, cargo loading methods, storage, and stability.

5.2.1. Characterization of Product for Clinical Trials

In the early 2000s, numerous reports on the therapeutic use of MSCs brought to our attention the fact that different methods of isolation and expansion of MSCs, as well as different approaches to characterize the cells, made it difficult to compare results of individual studies, underlining the need to define the minimum criteria for defining MSCs. In a position paper, the Mesenchymal and Tissue Stem Cell Committee of the International Society for Cellular Therapy proposed that MSCs must (1) be plastic-adherent under standard culture conditions; (2) express CD105, CD73, and CD90, and lack expression of CD45, CD34, CD14, or CD11b, CD79α or CD19, and HLA-DR surface molecules; and (3) differentiate into osteoblasts, adipocytes, and chondroblasts in vitro [92]. However, these criteria are insufficient for predicting the therapeutic potency of MSCs and do not consider factors such as cell source, culturing conditions, intrinsic donor variability, and effects of microenvironment after infusion (reviewed in [93]), contributing to the failure of some later stage clinical trials. Additionally, therapeutic MSCs are tested for viability, sterility, endotoxin, and mycoplasma before being released into the clinical setting [94,95,96,97].
In comparison, the characterization of EVs for clinical trials meets different challenges. Even though criteria for immunophenotypic characterization are defined [30], characterization at the single-vesicle level requires the use of advanced-flow cytometry [98]. Ensuring high purity of preparation is very important: both exogenous EVs (from fetal bovine serum (FBS) or human platelet lysate (hPL)) and non-EVs components (including protein, RNA, and lipid) may confound the therapeutic potential of MSC-EVs. An additional characterization of MSC-EVs based on specific lipid content may improve the reliability of data [30]. Implementation of rigorous criteria for phenotypic characterization of EVs using flow cytometry and the development of nanoflow technologies will increase the standard of production of clinical-grade EVs [98].

5.2.2. Dosing and Safety

The therapeutic dose of MSCs is commonly established as the number of cells per body mass [94,95,96,97]. In assessment of safety, MSC clinical study design estimates the increase of the treatment dose, while simultaneously monitoring for adverse effects (CTCAE) and dose-limiting toxicity (DLT). The study proceeds with the highest dose in which the adverse effects were not observed.
In comparison to MSCs, parallel attempts to define MSC-EVs for clinical trials fall short, due to limitations of technology used to quantify and characterize EVs. Quantification of EVs using nanoparticle tracking analysis (NTA), dynamic light scattering (DLS), or tunable resistive pulse sensor (TRPS) is hampered by the lack of discrimination between small EV and non-EV particles, including lipoproteins and protein aggregates. A recent position paper on the development of EV-based medicinal products, recommends the use of these technologies in combination with immune-chemical characterization [99].
Safety concerns in the application of MSCs and MSC-EVs are somewhat different. While in the case of MSC-based therapies, phenotypic stability (and related loss of therapeutic potential) present major concern, in the case of EVs, these concerns are related to the mechanism of action (MoA) and the target identity, which are often insufficiently understood [99].
Clinical trials of MSCs, performed to treat kidney disease, demonstrated that MSCs can be safely given to humans and are generally well tolerated [94,95,96,97] with no difference in the rate of adverse effects between groups treated with MSCs and placebo [100]. Taking this into account and considering (native) EVs are physiologically produced and taken up in different tissues, there is little expectation of adverse events. However, adverse effects may arise from events related to anticipated MoA, and they should be closely monitored and reported [99].

5.2.3. Efficacy

In terms of efficacy, available data require careful consideration with respect to pathology, endpoints used to monitor patient outcomes and differences in study design. Several, early-phase clinical trials used MSCs to treat AKI associated with cardiac surgery. Studies that used MSCs concurrent with surgery in patients with increased risk of AKI, reported shorter hospital stays [101,102]. However, when AKI developed during cardiac surgery, subsequent MSCs treatment did not decrease the time necessary for the recovery of kidney function [100]. In CKD, studies reported a significant decrease in unfavorable outcomes compared to the placebo group [103,104]. However, when parameters of kidney function are monitored, results may be conflicting: the improvement may be recorded based on protein excretion rate [105], but the lack of effect was reported when GFR is monitored [105,106]. More consistent efficacy was reported in lupus, with studies reporting stabilization of kidney function in systemic lupus erythematosus (SLE) [107,108,109] and long-term stabilization of proteinuria in lupus nephritis (LN) [110]. However, at least one clinical trial showed no differences between MSCs treatment and standard immunosuppressive therapy [111].
These data show that a careful choice of endpoints is a prerequisite for the success of phase III clinical trials which are intended to show treatment efficacy. Combining several endpoints, based on the proposed MoA, may help improve the outcome of clinical trials. This also underlines the importance of an in-depth understanding of MoA, which is often lacking when the therapeutic effects of EVs are considered.

5.2.4. Heterogeneity of MSC-Derived EVs

A major bottleneck hampering the successful clinical application of MSC-EVs is related to their heterogeneity. First, MSC-EVs carry diverse molecules and varying amounts of bioactive components. As EVs composition is dictated by the parent cell, culturing conditions (cell source, donor variability, bioreactors vs. monolayer, presence of FBS or hPL, EVs isolation protocol) may heavily influence the biological activity of the final product. Due to an evident donor-to-donor variability of primary MSCs and its influence on the heterogeneity of MSC-EVs, the usage of immortalized MSCs for the production of EVs holds great promise in the field of overcoming various constraints linked with primary MSCs such as limited lifespan and proliferative capacity. In-process quality control that would identify changes in the EV composition and the integrity of the genome from the beginning to the end of EV production would assure the preservation of bioactive components during MSC-EVs production. Secondly, MSC-EVs comprise a heterogenous mixture of different EVs. This heterogeneity is closely related to the plethora of methods applied for their isolation/enrichment (Table 2). The composition, size, and morphology of EVs can influence their uptake, biodistribution, and functionality. Variability in surface markers expressed can also vary depending on the isolation method, culturing conditions, and donor variability, which can all influence the targeting, binding, and recognition of EVs by recipient cells and the mechanisms triggered in the target cell.
Overcoming the heterogeneity of MSC-derived EVs is essential for improving their therapeutic efficacy, reproducibility, and clinical translation. Efforts in the field are being made to establish standardized protocols for the growth of MSCs, as well as MSC-EV isolation and characterization.

5.3. MSC-EVs—Proposed Mechanisms of Action

Numerous studies have demonstrated the therapeutic effects of MSC-EVs in different disease models; nevertheless, only a limited number described their precise MoA. Here, we provide an overview of possible mechanisms of MSC-EV´s actions with an emphasis on kidney disease (Table 2).

5.3.1. Proposed MSC-EV’s Mechanisms of Action in Kidney Diseases

A growing body of evidence suggests the significance of particular proteins in transmitting information from MSC-EVs to their target cells. Thus, human UC-MSC-EVs attenuated renal tubulointerstitial fibrosis (TIF) by delivering casein kinase 1δ (CK1δ) and E3 ubiquitin ligase (β-TRCP) to kidney fibroblasts, which promoted nuclear-cytoplasmic shuttling of Yes-associated protein (YAP) and its ubiquitination and degradation [89]. EVs from GDNF-stimulated human AD-MSCs ameliorated the loss of peritubular capillaries in the TIF model and in HUVECs through the SIRT1/eNOS signaling pathway [72]. UC-MSC-derived EVs are protected from cisplatin-induced renal oxidative stress and apoptosis by promoting proliferation through the activation of the ERK1/2 pathway [78]. There are also indications that MSC-EVs can sequester soluble molecules, hindering their biological effect. Thus, one of the highly abundant proteins on MSC-EVs, the C-C motif chemokine receptor-2 (CCR2), was able to bind to its ligand CCL2 and reduce its concentration suppressing recruitment and activation of macrophages in renal IRI in mice [55].
The therapeutic effect of MSC-EVs may also be mediated by the RNA. The study by Collino et al. demonstrated that miRNA from MSC-EVs might exert their effects on genes involved in fatty acid metabolism, inflammation, matrix-receptor interaction, and cell adhesion [60]. MSC-EVs were shown to transfer miR-let7c to kidney epithelial cells and downregulate the expression of TGF-βR1, attenuating kidney injury and fibrosis [61]. EVs from erythropoietin-treated MSCs transferred miR-144 to fibroblasts, thus attenuating kidney fibrosis via a decrease of tissue plasminogen activator (tPA) and MMP9 [63]. BM-MSC-EVs were also shown to protect against renal IRI by transferring miR-199a-3p to kidney cells, which downregulated semaphorin 3A expression and activated the protein kinase B (AKT) and ERK pathways [56]. Transfer of miR-30b by UC-MSC-EVs was shown to inhibit mitochondrial fission, one of the main contributors to IRI [81]. miR-146b from MSC-EVs targeted interleukin (IL)-1 receptor-associated kinase (IRAK1) leading to the inhibition of NF-κB activity and subsequent lessening of pro-inflammatory response in sepsis-associated AKI [87]. Another miRNA from MSC-EVs, miR-125b-5p, after being taken up by kidney cells markedly decreased the expression of p53, leading to the upregulated CDK1 and Cyclin B1, and modulation of Bcl-2 and Bax [88]. Furthermore, in a rat model of renal IRI, human UC-MSC-EVs alleviated kidney damage and enhanced angiogenesis via the transfer of VEGF directly to renal tubular epithelial cells.
Table 2. Proposed mechanisms of MSC-EVs in kidney disease models.
Table 2. Proposed mechanisms of MSC-EVs in kidney disease models.
MSCs OriginMSCs
Manipulation
MSCs Medium before EVs IsolationEVs
Isolation Method
Disease ModelEffectsTarget CellsMechanisms (Molecule(s) or Signaling Pathways Identified)Confirmation MethodRef. No.
hUC/serum-free DMEMUCrat UUO model of RFAmelioration of RF/EVs transfer CK1δ and E3 ubiquitin ligase β-TRCP promoting YAP ubiquitination and degradation knockdown of CK1δ and β-TRCP in MSCs[89]
hATtransfection of GDNF in MSCsFCS-free RPMI supplemented with 0.5% BSAUCmouse UUO model of RFAmelioration of RF and peritubular capillary lossHUVEC cellsSirtuin 1 (SIRT1) pathwaySIRT1 knockdown[72]
hUC/ low glucose DMEMUCrat cisplatin-induced AKIImproved renal function, morphology, and renal cell proliferation; decreased tubular apoptosis and oxidative stressNRK-52E p-p38 and caspase 3; PCNA and p-ERKInhibition of MEK1 and MEK2 with U0126[78]
mBM/medium containing EVs-free serumUCmouse kidney IRIReduction of renal injury/ CCR2 on MSC-exo binds to CCL2 lowering the recruitment of macrophagesCCR2 knockdown [55]
BM/FCS-deprived RPMI with 0.5% BSAUCglycerol-
induced AKI in severe combined IDM
Morphologic and functional recovery in AKI/microRNAsDepletion of Drosha to alter miRNA
expression
[60]
hBMoverexpression of miRNA-let7cAlpha-MEM with EVs-free FBS“ExoQuick”mouse UUO model of RFAttenuation of kidney injuryNRK-52E miR-let7c attenuates TGF-β1-driven TGF-βR1 gene expression Labeling pre-miRNA;
inhibition of EVs released by GW4869
[61]
mBMErythropoietin treatmentRPMI with 10% FBSUC or “PureExo exosome isolation kit”mouse UUO model of RFDecrease of tBM disruption NRK-49FmiR-144 targets the tPA 3′-untranslated region and suppresses tPA and MMP9 level and activityDepletion of miR-144[63]
hBMmiR-199a-3p overexpressionDMEM/F12 with 10% EVs-free FBSUC hypoxia/reoxygenation (H/R) injury of HK-2 Alleviation of IRI, functional recovery, and histologic protection; antiapoptotic effectHK-2 miR-199a-3p decrease Sema3A, activate protein kinase B (AKT) and extracellular-signal-regulated kinase (ERK) pathwaysmiR-199a-3p knockdown; Sema3A knockdown[56]
hUC/FBS-depleted DMEM with 0.5% BSAUCrat IRI Amelioration of acute kidney IRI;
reduction of cell apoptosis
rat RTECsmiR-30b/c/d inhibits mitochondrial fission through Dynamin-related protein 1 (DRP1)miR-30
antagomirs
[81]
hUC/DMEM with 10% EVs-depleted FBSSucrose
/D2O cushion UC
sepsis—
associated AKI
Alleviation of AKI and kidney tubular cells apoptosis; increased survivalHK-2 miR-146b inhibits NF-κB activity and translocation of NFκB p-P65;
decrease of IRAK1
miR-146b
mimics and inhibitors
[87]
hUC/FBS-free mix of MSCM and DMEM/F12 UCmurine IRI Alleviation of ischemic AKI and decrease of renal tubular injury; attenuation of cell cycle arrest and apoptosisHK-2 Homing of EVs by VLA-4 and LFA-1; miR-125b-5p suppresses p53 in TECs leading to rescue G2/M arrest through up-regulation of CDK1 and Cyclin B1; apoptosis inhibition via Bcl-2 and BaxmiR-125b-5p Ox and inhibition;
Ox of p53;
transfection with siICAM-1 and siVCAM-1
[88]
hBM/FCS-free RPMI supplemented with 0.5% of BSA UCrat IRI modelReduction of renal function impairment/RNA cargoRNase
treatment
[54]
hUC/FBS-deprived DMEM, with 0.5% BSAUCrat model of unilateral AKIReversal of abnormal kidney structure and function;
tubular cell dedifferentiation and growth; inhibition of apoptosis
tubular cellsExpression of HGF in tubar cells via its mRNA transfer; tubular cells media promote tubular cell dedifferentiation and migration via HGF/c-Met and Erk1/2 signaling
pathways
RNase
treatment;
c-Met inhibitor
or MEK inhibitor
[80]
hUC/FBS-free mediumUCrat IRI model of AKI; hypoxia injury of RTECsReduction of RF and cell apoptosis; increase of proliferation, renal function, and capillary vessel density/VEGF delivery to rat RTECs; RNAAnti-human VEGF antibody;
RNase
treatment
[84]
hUC—human umbilical cord, hAT—human adipose tissue, mBM—mouse bone marrow, hBM—human bone marrow, GDNF—Glial cell line-derived neurotrophic factor, UC—ultracentrifugation, UUO—unilateral ureteral obstruction, AKI—acute kidney disease, IRI—ischaemia-reperfusion injury; RF—renal fibrosis; NRK-52E—rat kidney tubular epithelial cells; IDM—immunodeficient mice; tBM—tubular basement membrane; NRK-49F—rat renal fibroblast cell line; HK-2—human kidney proximal tubular epithelial cell line; RTECs—kidney tubular epithelial cells; MSCM—mesenchymal stem cell medium; Ox—overexpression.
Described beneficial effects of EVs were abrogated by RNase treatment [54,80,84]. Thus, UC-MSC-EVs promoted regeneration after AKI by accelerating tubular cell dedifferentiation and growth through HGF induction and Erk1/2 signaling, while the RNase treatment abrogated all MSC-EVs effects [80]. Additionally, the effect of MSC-EVs in alleviating AKI in the IRI model, based on inhibiting apoptosis and stimulating tubular epithelial cell proliferation, was shown to be ineffective after RNase treatment [54]. However, some reports caution against miR transfer as the major road to the biological activity of EVs [41,112]. By employing sophisticated purification and stringent identification criteria, Ago proteins (that bind miR in active RISC complexes) can clearly be separated from EVs. Additionally, the probability of physiological effects of miRs may depend on the abundance of one or more mRNA targets in the recipient cell, considering that as every miR may regulate multiple target mRNAs and downregulations of one mRNA may not recapitulate total miR effects [113].
Thus, details on the mechanism of MSC-EVs’ interaction with target cells, signal delivery, intracellular events, and the cellular response on RNA and the protein level need more clarification. MSC-EVs undoubtedly produce beneficial effects both in in vitro and in vivo models. However, for their wide clinical use in different kidney disorders, as well as in other diseases, it is necessary to elucidate the details of their MoAs.

5.3.2. Mechanism of MSC-EVs Actions Related to Immunomodulation

It has been reported that MSC-EVs exert their immunomodulatory properties on cells of both innate and adaptive immune response [114] (Supplementary Table S1). They were shown to generally induce polarization of macrophages towards an anti-inflammatory M2 phenotype and inhibit the development of pro-inflammatory M1 macrophages. Indeed, human UC-MSC-EVs alleviated inflammation of the fallopian tubes in mice by inducing macrophage polarization from M1 to M2 type via the NF-κB signaling pathway [115]. Mouse BM-MSC-EVs attenuated myocardial injury favoring the M2 anti-inflammatory state by inhibition of the TLR4/NF-κB signaling and the activation of the PI3K/Akt pathway [116]. In sepsis-induced acute respiratory distress syndrome, mBM-MSC-EVs induced a switch towards M2 macrophages and downregulated several essential proteins of glycolysis via inhibition of HIF-1α [117]. Furthermore, MSC-EV’s capacity to shift the THP-1 cell’s inflammatory phenotype towards an anti-inflammatory profile was MyD88-dependent and involved the TLR/NF-κB signaling pathway [118].
Various miRNAs have been shown to play a role in MSC-EV’s MoAs related to immunomodulation. Namely, Lu et al. showed that miR-223-3p from mBM-MSC-EVs attenuated the release of LPS-induced proinflammatory cytokines from macrophages and their STAT3 expression [119], while in the mouse model of lung fibrosis, hUC-MSC-EVs transferred the same miRNA to macrophages, thus increasing the circPWWP2A and NLRP3 pro-inflammatory signaling [120]. In a mouse model of sepsis, miR-27b from mBM-MSC-EVs targeted JMJD3 and inhibited the recruitment of NF-κB to the promoter region of proinflammatory genes [121]. In a spinal cord injury model, BM-MSC-EVs exerted neuroprotective effects by targeting interferon regulatory factor 5 (IRF5) expression in macrophages via miR-125a [122]. In a mouse model of atherosclerosis, miR-21a-5p from MSC-derived exosomes promoted macrophage polarization and reduced their infiltration within the atheroma by targeting KLF6 and ERK1/2 signaling pathways [123], while miR-let7 from MSC-EVs were shown to promote M2 macrophage polarization through miR-let7/HMGA2/NF-κB pathway, and suppress macrophage infiltration via miR-let7/IGF2BP1/PTEN pathway [124].
Diverse proteins have also been shown to play important roles in exerting MSC-EVs effects on macrophages. Thus, STAT3 from adipose-derived MSC-EVs activated the M2 phenotype through the transactivation of arginase-1, while methallothionein-2 from hBM-MSC-EVs elicited its anti-inflammatory effects in a mouse model of inflammatory bowel disease by suppressing NF-κB activation and inhibiting the phosphorylation of IκBα [125].
MSC-EVs have also been shown to suppress the maturation and activation of dendritic cells (DC) [126,127,128]. However, only a few studies attempt to elucidate the mechanisms of this action. Wu et al. found that BM-MSC-EVs enhanced the miR-146a and inhibited Fas expression in immature and mature DCs while reducing IL-12 expression/production of mature DCs [129]. However, the authors did not confirm whether miR-146a from MSC-EVs was responsible for described effects. hMB-MSCs-EVs halt DC maturation, decreased expression of maturation/activation markers, and secretion of pro-inflammatory cytokines, diminishing CCR7 expression after LPS stimulation. miR-21-5p was identified as a possible candidate for the modulation of CCR7 expression and DCs migration [130]. MSC-EVs can modulate NK cells by decreasing their number in the spleen or on the site of an injury [131], as well as impairing their function, which was shown to be at least in part dependent on TGFβ signaling [132]. Studies on the influence of MSC-EVs on granulocytes demonstrate their effects on neutrophils [133,134]. MSC-EVs attenuated the complement-mediated NET formation and neutrophil IL-17 production through the CD59-dependent mechanism [135]. Furthermore, treatment of neutrophiles with hUC-MSC-EVs inhibited their activity by attenuating the respiratory burst and oxidative stress [136].
Several studies demonstrated the influence of MSC-EVs on mast cells and indicated possible mechanisms. Thus, MSC-EVs suppressed mast cell activation through the mechanism involving an increase of prostaglandin E2 (PGE2) production and E-prostanoid 4 (EP4) receptor in mast cells [137]. Attenuation of mast cell activation, i.e., ROS generation, degranulation, and cytokine production were accompanied by the reduction of NF-κB, p-p38, p-JNK, and p-STAT5 levels [138]. MSC-EVs were also shown to suppress the proliferation of inflammatory cells while increasing regulatory T cells through TGF-β signaling [139,140] or adenosine signaling through the A2A receptor [140,141]. The effects of MSC-EVs on B cells have also been described; however, precise mechanisms are lacking [142].

5.3.3. Mechanism of MSC-EVs Actions Related to the Regeneration

Apart from having immunomodulatory effects, including ones involved in the control of inflammation post-injury, MSC-EVs also have effects on the repair and remodeling phases of tissue regeneration (Supplementary Table S2). They are involved in the attenuation of apoptosis, angiogenesis, and repopulation (proliferation). Moreover, they also have a damage control role by diminishing oxidative stress. Mechanisms by which MSC-EVs exert these effects are versatile.
MSC-EVs exert their anti-oxidative effect by transferring antioxidant glutathione peroxidase 1 (GPX1) to recipient cells [143]. They can also suppress the expression of NADPH oxidase 2 (NOX2) and thus the production of reactive oxygen species [82]. These effects can also be indirect, i.e., by influencing the anti-oxidative pathway at the level of Nrf2/antioxidant response element (ARE) activity which in turn activates the expression of antioxidant genes [83]. However, mentioned effects were not observed in all models [144], i.e., in the drug-induced, liver injury mouse model, human MSC-EVs did not mitigate hepatocyte injury via modulation of oxidative stress.
Anti-oxidative effects of MSC-EVs are closely related to anti-apoptotic effects since oxidative stress can lead to apoptosis [145]. Thus, in the above-described example of GPX1 transfer, downregulation of apoptosis was also observed [143]. MSC-EVs can also prevent apoptosis through the activation of the Wnt/β-catenin signaling pathway [146] or by downregulation of pro-inflammatory signaling activated by IL-1β, such are Erk1/2, PI3K/Akt, p38, TAK1, and NF-κB [147]. Furthermore, MSC-EVs can cause up-regulation of anti-apoptotic genes, such as Bcl-xL [144], or modulate anti-apoptotic (Akt and GSK-3β) or proapoptotic (c-JNK) factors [148]. Several mechanisms by which MSC-EVs exert anti-apoptotic effect were connected to EVs’ transfer of RNA, i.e., miRNA which can modulate mRNAs associated with apoptosis, such as caspase-3 and -7 [149] or other types of RNA such as Y-RNA-1 [150]. Anti-apoptotic effects of MSC-EVs, exerted through downregulation of methyl CpG binding protein 2 (Mecp2) [151] and by reducing the expression of PUMA (p53 upregulated modulator of apoptosis) [152], were also associated with EVs’ transfer of miRNA (miR-22 and miR-221, respectively). Similar was observed for miR-125b-5p whose transfer via MSC-EVs was related to the suppression of pro-apoptotic genes p53 and BCL2-antagonist/killer 1 (BAK1) [153].
MSC-EV’s ability to promote angiogenesis often relies on shuttling pro-angiogenic transcription factors, such as VEGF, IGF-1, and bFGF [75,84]. Subsequently, the activation of the VEGF pathway can also lead to the activation of the Hippo signaling pathway [154].
MSC-EVs were often found to have significant positive or negative effects on the proliferation rates of target cells, depending on the model. Thus, in liver fibrosis, MSC-EVs decreased the proliferation of hepatic stellate cells (HSCs) by transferring miR-122 [155], while in kidney tubular cells they restored proliferation after damage caused by ATP depletion [149]. The mechanisms governing the MSC-EVs’ effects of proliferation are versatile, as they can activate Wnt/β-catenin [156]), ERK1/2 [78,157], or Akt signaling pathways [157]. They can also up-regulate proliferation proteins such are PCNA and Cyclin D1 [144,158]. In some studies, the effects of MSC-EVs on proliferation were associated with EVs’ transfer of mRNA or miRNA [76]. However, it should be noted that RNase treatment of those EVs diminished their effects, pointing out issues mentioned in Section 5.3.1. MSC-EVs can also influence the proliferation rate indirectly, by transferring mRNA for IGF-1R and thus increasing the sensitivity of recipient cells to locally produced IGF-1 [159].
The above examples of a vast variety of mechanisms of MSC-EVs’ actions point both toward the heterogeneity of EVs from different sources and conditions, as well as to the heterogenous milieus that are present in different targets (i.e., the different expression of the target molecules and/or influences of other signals on the same pathways). Obviously, MSC-EVs are able to influence different processes using different mechanisms even for the same process (i.e., inflammation or regeneration). Currently, there is a lack of studies that comprehensively elucidate the mechanisms of MSC-EVs’ actions. Most studies identified only fragments of such pathways. Given the high complexity of EVs, it is conceivable that the mechanism of MSC-EVs’ actions comprises a combination of targeting molecules and active molecules, and possibly more than one of each. Furthermore, in consideration of this plethora of information about the mechanism of MSC-EVs’ actions, different origins of MSCs (adult or neonatal, different tissues) as well as different conditions of their isolation, cultivation (influence of medium) or manipulation (knockdown, overexpression, EVs release inhibition, etc.), as reviewed in [48], should be considered as they can affect cells’ physiology and thus composition/action of EVs. Also, importantly, conditions and methods of isolation of EVs can also influence their composition and actions. Finally, the methods for confirmation of molecules identified as mechanism participants should also be considered. All this urges further efforts towards the elucidation of MSC-EVs mechanisms so we can fully exploit EVs’ versatile therapeutic potentials for kidney diseases (as reviewed in Chapter 4), as well as for other pathologies.

5.4. Potency Testing

Potency is an essential parameter used to assess the therapeutic activity of a drug product. Establishing a suitable potency assay is crucial for the production of MSC and MSC-EV products. However, setting up such assays, especially for MSC-EV products, is challenging [160,161]. Ideally, potency assays should be reliable, quick, replicable, and measurable, to accurately evaluate the therapeutic product’s desired function. Furthermore, such assays should satisfy specific regulatory guidelines (https://database.ich.org/sites/default/files/Q6B_Guideline.pdf, accessed on 25 February 2023) and ideally relate to the product’s MoA (www.fda.gov/media/79856/download, accessed on 25 February 2023).
Taking into consideration that immunomodulatory activities of MSC and MSC-EVs products are considered to be an essential part of their MoA in many disease models, they are often examined in various in vitro assays, primarily focusing on T cells or macrophages. Potency assays coupled with clinical MSC vary based on their intended clinical use but are closely related to MoA. For MSCs used in the treatment of autoimmune disease or organ transplantation, potency assays include assessment of T cell proliferation or cytokine secretion, with varying formats [162,163,164]. For MSCs used in tissue repair, tests assessing cell behavior (adhesion, proliferation, differentiation, and angiogenesis) are more appropriate [165,166,167]. However, not all of these assays measure the same immunomodulatory activity, and not all activities measured are relevant to the therapeutic effects of the cells for a specific disease [160]. Previous attempts to predict the clinical effectiveness of administered MSCs using in vitro assays have been unreliable and inconsistent. Therefore, it is important that potency assays used to predict the therapeutic function of EV-based therapeutics measure activities that are directly relevant to the EV products’ MoA for a specific disease, which may be a compendium of different individual activities or MoA attributes, respectively [168]. Thus, appropriate potency testing may require an assay matrix to be established [168].
There are several aspects that have to be taken into account when designing the right potency assay for kidney diseases. The design of the potency assays for MSC-EV therapies in kidney disease will greatly depend on the pathophysiological stage of the disease, whether it is acute or chronic, and the type of underlying condition, either inflammatory or fibrotic. Thus, when designing the potency assay for the MSC-EV product that will be used for the AKI, the accent should be placed on immunomodulatory, anti-inflammatory, and renoprotective effects. Conversely, potency assays measuring the extent of ECM in kidney fibrosis may be more advantageous for MSC-EV products that will be used in the later stages of kidney disease. In addition, whenever we encounter difficulties for routine tests (due to high variability or time consumption), it is preferable to utilize surrogate measurements (indirect measures) that have been previously shown to correlate with functional assays, such as specific gene expression or secretion factors. Thus, the effect of MSC-EVs on inflammatory response in AKI can be assessed in vitro by measuring the secretion of pro- and anti-inflammatory factors, such as IL-6, TNF-α, and IL-10, while the effect of MSC-EV product on renoprotection and tubular renal senescence could be measured by assessing the levels of renal αKlotho, known antiaging and renoprotective protein.
Even though the current pre- and clinical data imply a huge potential for MSC-EV products, there are some caveats inherited from the MSC field [160]. It is obvious that MSCs are a very heterogenous cell entity [161,169,170,171] and that not all MSC products succeeded in clinical trials [172,173,174]. Namely, a comparison of therapeutic activities of independent MSC-EV preparations all manufactured with the same standardized procedure, demonstrated differences in the potency of different MSC-EV batches in modulating disease symptoms in various animal models, specifically in mouse models of ischemic stroke, Niemann-Pick type C disease and aGvHD [175,176,177]. Thus, it is an important task in the field, now, to set up assays that appropriately evaluate the therapeutic activities of MSC-EV products, allowing discrimination between potent and non-potent products before applying them to larger patient cohorts in clinical trials [160,168,178].

6. Perspectives of MSC-EV Use in the Treatment of Kidney Disease

While incidences of kidney disease are rising worldwide, we still lack effective treatments to repair damaged kidney tissue and/or prevent disease progression. The use of MSC technology, already proven to be promising in the treatment of a variety of diseases, emerged as such a strategy. This especially refers to their EVs, which offer important advantages to cellular therapy, modulating vital processes important for kidney physiology such as immune response, tissue repair, angiogenesis, oxidative stress, and inhibition of cell death.
The beneficial and renoprotective effects of MSC-EVs in kidney pathologies have been well documented in a large number of different preclinical models. However, their use in clinical trials is clearly very limited. One possible explanation is that there is still much that is not yet known about the mechanisms of action of MSC-EV products, their safety, efficacy, and optimal dosing regimens, as well as the lack of suitable potency assays.
In order to make a significant step toward the clinical application of MSC-EVs, we must ensure that crucial obstacles are overcome. This involves standardization of methods for MSC-EVs isolation and characterization. Different aspects of quality control such as quantity, purity, sterility, and endotoxin should not be disregarded. In parallel, we need to clarify the sophisticated mechanisms of MSC-EVs’ action and design appropriate potency assays that will precisely assess the therapeutic activity of MSC-EV products in a specific kidney disease. In this regard, depending on the pathophysiological stage of kidney disease, assessment of the effect of MSC-EV products on kidney tubular or glomerular inflammation, apoptosis, or senescence could be considered as one of the potential in vitro potency assays. Furthermore, designing the potency assay that will work on a kidney-on-a-chip in vitro model would surpass the usual limitations of a conventional 2D-cell culture model and allow the study of the MSC-EVs´ dynamic within the nephron structure, which is very important taking into consideration the complex kidney structure.
Due to the considerable complexity of the kidney structure and the multifaceted pathogenesis of kidney diseases, kidney-targeted drug delivery is a difficult task as there are currently very limited strategies available to target the right kidney cell and tackle the precise signaling pathway. Hence, we need to work on enhancing the efficacy of MSC-EV-based renal therapy. Increasing the therapeutic efficacy of MSC-EVs, by presenting different targeting moieties such as antibodies, peptides, and small molecules on their surface would lead to better homing of MSC-EVs towards different kidney cells and a decrease in target effects.
Despite challenges in MSC-EV production and characterization, the potential of MSC-EV-based therapeutics as a novel type of nanomedicine for kidney disease is promising. As our comprehension of kidney disease continues to thrive, the development of MSC-EV products will also continue to evolve, paving the way for an effective treatment option for patients with kidney disease.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/pharmaceutics15071911/s1, Table S1: Proposed mechanisms of MSC-EVs related to immunological effects, Table S2: Proposed mechanisms of MSC-EVs related to regenerative effects.

Author Contributions

Conceptualization, M.K., B.M., M.B.; literature review, M.K., B.M., M.B.; writing—original draft preparation, M.K., B.M., T.J.K., Y.M., M.B.; writing—review and editing, M.K., B.M., M.B.; supervision, M.B.; funding acquisition, M.B. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by Instituto de Salud Carlos III (ISCIII) through projects CP19/00027 and PI21/00204 and was co-funded by the European Union. M.B. was supported by the Miguel Servet contract from the Instituto de Salud Carlos III (ISCIII) (CP19/00027), ESF “Investing in your future’’, and MV22/00017 grant. M.K. was supported by a research grant from the Ministry of Science, Technological Development and Innovation of the Republic of Serbia (Contract No. 451-03-47/2023-01/200019).

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Bernd Giebel from the Institute for Transfusion Medicine, University Hospital Essen, Germany for the critical review of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Levey, A.S.; Levin, A.; Kellum, J.A. Definition and Classification of Kidney Diseases. Am. J. Kidney Dis. 2013, 61, 686–688. [Google Scholar] [CrossRef]
  2. Fais, S.; O’Driscoll, L.; Borras, F.E.; Buzas, E.; Camussi, G.; Cappello, F.; Carvalho, J.; Cordeiro-Da-Silva, A.; Del Portillo, H.; El Andaloussi, S.; et al. Evidence-Based Clinical Use of Nanoscale Extracellular Vesicles in Nanomedicine. ACS Nano 2016, 10, 3886–3899. [Google Scholar] [CrossRef]
  3. Kosanović, M.; Milutinovic, B.; Glamočlija, S.; Morlans, I.M.; Ortiz, A.; Bozic, M. Extracellular Vesicles and Acute Kidney Injury: Potential Therapeutic Avenue for Renal Repair and Regeneration. Int. J. Mol. Sci. 2022, 23, 3792. [Google Scholar] [CrossRef]
  4. Eckardt, K.-U.; Coresh, J.; Devuyst, O.; Johnson, R.J.; Köttgen, A.; Levey, A.S.; Levin, A. Evolving importance of kidney disease: From subspecialty to global health burden. Lancet 2013, 382, 158–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Bonventre, J.V.; Yang, L. Cellular pathophysiology of ischemic acute kidney injury. J. Clin. Investig. 2011, 121, 4210–4221. [Google Scholar] [CrossRef]
  6. Susantitaphong, P.; Cruz, D.N.; Cerda, J.; Abulfaraj, M.; Alqahtani, F.; Koulouridis, I.; Jaber, B.L.; Acute Kidney Injury Advisory Group of the American Society of Nephrology. World Incidence of AKI: A meta-analysis. Clin. J. Am. Soc. Nephrol. 2013, 8, 1482–1493. [Google Scholar] [CrossRef] [Green Version]
  7. Guo, L.; Zhang, T.; Wang, F.; Chen, X.; Xu, H.; Zhou, C.; Chen, M.; Yu, F.; Wang, S.; Yang, D.; et al. Targeted inhibition of Rev-erb-α/β limits ferroptosis to ameliorate folic acid-induced acute kidney injury. Br. J. Pharmacol. 2020, 178, 328–345. [Google Scholar] [CrossRef] [PubMed]
  8. Venkatachalam, M.A.; Griffin, K.A.; Lan, R.; Geng, H.; Saikumar, P.; Bidani, A.K.; Polichnowski, A.J.; Fu, Y.; Tang, C.; Cai, J.; et al. Acute kidney injury: A springboard for progression in chronic kidney disease. Am. J. Physiol. Physiol. 2010, 298, F1078–F1094. [Google Scholar] [CrossRef] [Green Version]
  9. Ruiz-Ortega, M.; Rayego-Mateos, S.; Lamas, S.; Ortiz, A.; Rodrigues-Diez, R.R. Targeting the progression of chronic kidney disease. Nat. Rev. Nephrol. 2020, 16, 269–288. [Google Scholar] [CrossRef] [PubMed]
  10. Tan, H.L.; Yap, J.Q.; Qian, Q. Acute Kidney Injury: Tubular Markers and Risk for Chronic Kidney Disease and End-Stage Kidney Failure. Blood Purif. 2016, 41, 144–150. [Google Scholar] [CrossRef]
  11. Musso, G.; Cassader, M.; Cohney, S.; De Michieli, F.; Pinach, S.; Saba, F.; Gambino, R. Fatty Liver and Chronic Kidney Disease: Novel Mechanistic Insights and Therapeutic Opportunities. Diabetes Care 2016, 39, 1830–1845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Burns, W.; Kantharidis, P.; Thomas, M. The Role of Tubular Epithelial-Mesenchymal Transition in Progressive Kidney Disease. Cells Tissues Organs 2007, 185, 222–231. [Google Scholar] [CrossRef]
  13. Levey, A.S.; Coresh, J.; Bolton, K.; Culleton, B.; Harvey, K.S.; Ikizler, T.A.; Johnson, C.A.; Kausz, A.; Kimmel, P.L.; Kusek, J.; et al. K/DOQI clinical practice guidelines for chronic kidney disease: Evaluation, classification, and stratification. Am. J. Kidney Dis. 2002, 39, S1–S266. [Google Scholar]
  14. Couser, W.G.; Remuzzi, G.; Mendis, S.; Tonelli, M. The contribution of chronic kidney disease to the global burden of major noncommunicable diseases. Kidney Int. 2011, 80, 1258–1270. [Google Scholar] [CrossRef] [Green Version]
  15. Bozic, M.; Caus, M.; Rodrigues-Diez, R.R.; Pedraza, N.; Ruiz-Ortega, M.; Garí, E.; Gallel, P.; Panadés, M.J.; Martinez, A.; Fernández, E.; et al. Protective role of renal proximal tubular alpha-synuclein in the pathogenesis of kidney fibrosis. Nat. Commun. 2020, 11, 1943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Kosanović, M.; Llorente, A.; Glamočlija, S.; Valdivielso, J.M.; Bozic, M. Extracellular Vesicles and Renal Fibrosis: An Odyssey toward a New Therapeutic Approach. Int. J. Mol. Sci. 2021, 22, 3887. [Google Scholar] [CrossRef]
  17. Bozic, M.; Valdivielso, J.M. Calcium Signaling in Renal Tubular Cells. Adv. Exp. Med. Biol. 2012, 740, 933–944. [Google Scholar] [CrossRef]
  18. Borràs, M.; Cambray, S.; Crespo-Masip, M.; Pérez-Fontán, M.; Bozic, M.; Bermudez-López, M.; Fernández, E.; Betriu, À.; Valdivielso, J.M. Peritoneal Dialysis Is an Independent Factor Associated to Lower Intima Media Thickness in Dialysis Patients Free From Previous Cardiovascular Disease. Front. Physiol. 2018, 9, 1743. [Google Scholar] [CrossRef] [Green Version]
  19. Yun, C.W.; Lee, S.H. Potential and Therapeutic Efficacy of Cell-based Therapy Using Mesenchymal Stem Cells for Acute/chronic Kidney Disease. Int. J. Mol. Sci. 2019, 20, 1619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Poomani, M.S.; Mariappan, I.; Perumal, R.; Regurajan, R.; Muthan, K.; Subramanian, V. Mesenchymal Stem Cell (MSCs) Therapy for Ischemic Heart Disease: A Promising Frontier. Glob. Heart 2022, 17, 19. [Google Scholar] [CrossRef]
  21. Staff, N.P.; Jones, D.T.; Singer, W. Mesenchymal Stromal Cell Therapies for Neurodegenerative Diseases. Mayo Clin. Proc. 2019, 94, 892–905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Eom, Y.W.; Yoon, Y.; Baik, S.K. Mesenchymal stem cell therapy for liver disease: Current status and future perspectives. Curr. Opin. Gastroenterol. 2021, 37, 216–223. [Google Scholar] [CrossRef] [PubMed]
  23. Chen, Y.; Yu, Q.; Hu, Y.; Shi, Y. Current Research and Use of Mesenchymal Stem Cells in the Therapy of Autoimmune Diseases. Curr. Stem Cell Res. Ther. 2019, 14, 579–582. [Google Scholar] [CrossRef]
  24. Morello, W.; Budelli, S.; Bernstein, D.A.; Montemurro, T.; Montelatici, E.; Lavazza, C.; Ghio, L.; Edefonti, A.; Peruzzi, L.; Molino, D.; et al. First clinical application of cord blood mesenchymal stromal cells in children with multi-drug resistant nephrotic syndrome. Stem Cell Res. Ther. 2022, 13, 420. [Google Scholar] [CrossRef]
  25. Lener, T.; Gimona, M.; Aigner, L.; Börger, V.; Buzas, E.; Camussi, G.; Chaput, N.; Chatterjee, D.; Court, F.A.; del Portillo, H.A.; et al. Applying extracellular vesicles based therapeutics in clinical trials—An ISEV position paper. J. Extracell. Vesicles 2015, 4, 30087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Lai, R.C.; Arslan, F.; Lee, M.M.; Sze, N.S.K.; Choo, A.; Chen, T.S.; Salto-Tellez, M.; Timmers, L.; Lee, C.N.; El Oakley, R.M.; et al. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010, 4, 214–222. [Google Scholar] [CrossRef] [Green Version]
  27. Bruno, S.; Grange, C.; Deregibus, M.C.; Calogero, R.A.; Saviozzi, S.; Collino, F.; Morando, L.; Busca, A.; Falda, M.; Bussolati, B.; et al. Mesenchymal Stem Cell-Derived Microvesicles Protect Against Acute Tubular Injury. J. Am. Soc. Nephrol. 2009, 20, 1053–1067. [Google Scholar] [CrossRef] [Green Version]
  28. Doeppner, T.R.; Herz, J.; Görgens, A.; Schlechter, J.; Ludwig, A.-K.; Radtke, S.; de Miroschedji, K.; Horn, P.A.; Giebel, B.; Hermann, D.M. Extracellular Vesicles Improve Post-Stroke Neuroregeneration and Prevent Postischemic Immunosuppression. Stem Cells Transl. Med. 2015, 4, 1131–1143. [Google Scholar] [CrossRef] [Green Version]
  29. Buzas, E.I. The roles of extracellular vesicles in the immune system. Nat. Rev. Immunol. 2022, 23, 236–250. [Google Scholar] [CrossRef]
  30. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Kowal, J.; Arras, G.; Colombo, M.; Jouve, M.; Morath, J.P.; Primdal-Bengtson, B.; Dingli, F.; Loew, D.; Tkach, M.; Théry, C. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc. Natl. Acad. Sci. USA 2016, 113, E968–E977. [Google Scholar] [CrossRef]
  32. Van Der Pol, E.; Böing, A.N.; Gool, E.L.; Nieuwland, R. Recent developments in the nomenclature, presence, isolation, detection and clinical impact of extracellular vesicles. J. Thromb. Haemost. 2016, 14, 48–56. [Google Scholar] [CrossRef] [Green Version]
  33. Witwer, K.W.; Théry, C. Extracellular vesicles or exosomes? On primacy, precision, and popularity influencing a choice of nomenclature. J. Extracell. Vesicles 2019, 8, 1648167. [Google Scholar] [CrossRef]
  34. Lötvall, J.; Hill, A.F.; Hochberg, F.; Buzás, E.I.; Di Vizio, D.; Gardiner, C.; Gho, Y.S.; Kurochkin, I.V.; Mathivanan, S.; Quesenberry, P.; et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: A position statement from the International Society for Extracellular Vesicles. J. Extracell. Vesicles 2014, 3, 26913. [Google Scholar] [CrossRef] [PubMed]
  35. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [Green Version]
  36. Ratajczak, J.; Miękus, K.; Kucia, M.; Zhang, J.; Reca, R.; Dvorak, P.; Ratajczak, M.Z. Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: Evidence for horizontal transfer of mRNA and protein delivery. Leukemia 2006, 20, 847–856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Ridder, K.; Keller, S.; Dams, M.; Rupp, A.-K.; Schlaudraff, J.; Del Turco, D.; Starmann, J.; Macas, J.; Karpova, D.; Devraj, K.; et al. Extracellular Vesicle-Mediated Transfer of Genetic Information between the Hematopoietic System and the Brain in Response to Inflammation. PLoS Biol. 2014, 12, e1001874. [Google Scholar] [CrossRef]
  38. Ridder, K.; Sevko, A.; Heide, J.; Dams, M.; Rupp, A.-K.; Macas, J.; Starmann, J.; Tjwa, M.; Plate, K.H.; Sültmann, H.; et al. Extracellular vesicle-mediated transfer of functional RNA in the tumor microenvironment. Oncoimmunology 2015, 4, e1008371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Pegtel, D.M.; Cosmopoulos, K.; Thorley-Lawson, D.A.; van Eijndhoven, M.A.J.; Hopmans, E.S.; Lindenberg, J.L.; de Gruijl, T.D.; Würdinger, T.; Middeldorp, J.M. Functional delivery of viral miRNAs via exosomes. Proc. Natl. Acad. Sci. USA 2010, 107, 6328–6333. [Google Scholar] [CrossRef]
  40. Lai, R.C.; Tan, S.S.; Yeo, R.W.Y.; Choo, A.B.H.; Reiner, A.T.; Su, Y.; Shen, Y.; Fu, Z.; Alexander, L.; Sze, S.K.; et al. MSC secretes at least 3 EV types each with a unique permutation of membrane lipid, protein and RNA. J. Extracell. Vesicles 2016, 5, 29828. [Google Scholar] [CrossRef] [PubMed]
  41. Albanese, M.; Chen, Y.-F.A.; Hüls, C.; Gärtner, K.; Tagawa, T.; Mejias-Perez, E.; Keppler, O.T.; Göbel, C.; Zeidler, R.; Shein, M.; et al. MicroRNAs are minor constituents of extracellular vesicles that are rarely delivered to target cells. PLoS Genet. 2021, 17, e1009951. [Google Scholar] [CrossRef]
  42. Mosbach, M.-L.; Pfafenrot, C.; von Strandmann, E.P.; Bindereif, A.; Preußer, C. Molecular Determinants for RNA Release into Extracellular Vesicles. Cells 2021, 10, 2674. [Google Scholar] [CrossRef]
  43. Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Zavec, A.B.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 2015, 4, 27066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Daly, R.; O’driscoll, L. Extracellular vesicles in blood: Are they viable as diagnostic and predictive tools in breast cancer? Drug Discov. Today 2020, 26, 778–785. [Google Scholar] [CrossRef]
  45. Balaphas, A.; Meyer, J.; Sadoul, R.; Morel, P.; Gonelle-Gispert, C.; Buhler, L. Extracellular vesicles: Future diagnostic and therapeutic tools for liver disease and regeneration. Liver Int. 2019, 39, 1801–1817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Tertel, T.; Tomić, S.; Đokić, J.; Radojević, D.; Stevanović, D.; Ilić, N.; Giebel, B.; Kosanović, M. Serum-derived extracellular vesicles: Novel biomarkers reflecting the disease severity of COVID-19 patients. J. Extracell. Vesicles 2022, 11, e12257. [Google Scholar] [CrossRef]
  47. Kordelas, L.; Rebmann, V.; Ludwig, A.-K.; Radtke, S.; Ruesing, J.; Doeppner, T.R.; Epple, M.; Horn, P.A.; Beelen, D.W.; Giebel, B. MSC-derived exosomes: A novel tool to treat therapy-refractory graft-versus-host disease. Leukemia 2014, 28, 970–973. [Google Scholar] [CrossRef]
  48. Börger, V.; Bremer, M.; Ferrer-Tur, R.; Gockeln, L.; Stambouli, O.; Becic, A.; Giebel, B. Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles and Their Potential as Novel Immunomodulatory Therapeutic Agents. Int. J. Mol. Sci. 2017, 18, 1450. [Google Scholar] [CrossRef] [Green Version]
  49. Wiese, D.M.; Ruttan, C.C.; Wood, C.A.; Ford, B.N.; Braid, L.R. Accumulating Transcriptome Drift Precedes Cell Aging in Human Umbilical Cord-Derived Mesenchymal Stromal Cells Serially Cultured to Replicative Senescence. Stem Cells Transl. Med. 2019, 8, 945–958. [Google Scholar] [CrossRef] [Green Version]
  50. Cui, L.-L.; Kerkelä, E.; Bakreen, A.; Nitzsche, F.; Andrzejewska, A.; Nowakowski, A.; Janowski, M.; Walczak, P.; Boltze, J.; Lukomska, B.; et al. The cerebral embolism evoked by intra-arterial delivery of allogeneic bone marrow mesenchymal stem cells in rats is related to cell dose and infusion velocity. Stem Cell Res. Ther. 2015, 6, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Nassar, W.; El-Ansary, M.; Sabry, D.; Mostafa, M.A.; Fayad, T.; Kotb, E.; Temraz, M.; Saad, A.-N.; Essa, W.; Adel, H. Umbilical cord mesenchymal stem cells derived extracellular vesicles can safely ameliorate the progression of chronic kidney diseases. Biomater. Res. 2016, 20, 21. [Google Scholar] [CrossRef] [Green Version]
  52. Warnecke, A.; Prenzler, N.; Harre, J.; Köhl, U.; Gärtner, L.; Lenarz, T.; Laner-Plamberger, S.; Wietzorrek, G.; Staecker, H.; Lassacher, T.; et al. First-in-human intracochlear application of human stromal cell-derived extracellular vesicles. J. Extracell. Vesicles 2021, 10, e12094. [Google Scholar] [CrossRef]
  53. Sengupta, V.; Sengupta, S.; Lazo, A.; Woods, P.; Nolan, A.; Bremer, N. Exosomes Derived from Bone Marrow Mesenchymal Stem Cells as Treatment for Severe COVID-19. Stem Cells Dev. 2020, 29, 747–754. [Google Scholar] [CrossRef] [PubMed]
  54. Gatti, S.; Bruno, S.; Deregibus, M.C.; Sordi, A.; Cantaluppi, V.; Tetta, C.; Camussi, G. Microvesicles derived from human adult mesenchymal stem cells protect against ischaemia-reperfusion-induced acute and chronic kidney injury. Nephrol. Dial. Transplant. 2011, 26, 1474–1483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Shen, B.; Liu, J.; Zhang, F.; Wang, Y.; Qin, Y.; Zhou, Z.; Qiu, J.; Fan, Y. CCR2 Positive Exosome Released by Mesenchymal Stem Cells Suppresses Macrophage Functions and Alleviates Ischemia/Reperfusion-Induced Renal Injury. Stem Cells Int. 2016, 2016, 1240301. [Google Scholar] [CrossRef] [Green Version]
  56. Zhu, G.; Pei, L.; Lin, F.; Yin, H.; Li, X.; He, W.; Liu, N.; Gou, X. Exosomes from human-bone-marrow-derived mesenchymal stem cells protect against renal ischemia/reperfusion injury via transferring miR-199a-3p. J. Cell. Physiol. 2019, 234, 23736–23749. [Google Scholar] [CrossRef]
  57. Zhou, Y.; Liu, S.; Zhao, M.; Wang, C.; Li, L.; Yuan, Y.; Li, L.; Liao, G.; Bresette, W.; Zhang, J.; et al. Injectable extracellular vesicle-released self-assembling peptide nanofiber hydrogel as an enhanced cell-free therapy for tissue regeneration. J. Control. Release 2019, 316, 93–104. [Google Scholar] [CrossRef]
  58. Bruno, S.; Grange, C.; Collino, F.; Deregibus, M.C.; Cantaluppi, V.; Biancone, L.; Tetta, C.; Camussi, G. Microvesicles Derived from Mesenchymal Stem Cells Enhance Survival in a Lethal Model of Acute Kidney Injury. PLoS ONE 2012, 7, e33115. [Google Scholar] [CrossRef]
  59. Reis, L.A.; Borges, F.T.; Simões, M.J.; Borges, A.A.; Sinigaglia-Coimbra, R.; Schor, N. Bone Marrow-Derived Mesenchymal Stem Cells Repaired but Did Not Prevent Gentamicin-Induced Acute Kidney Injury through Paracrine Effects in Rats. PLoS ONE 2012, 7, e44092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Collino, F.; Bruno, S.; Incarnato, D.; Dettori, D.; Neri, F.; Provero, P.; Pomatto, M.; Oliviero, S.; Tetta, C.; Quesenberry, P.J.; et al. AKI Recovery Induced by Mesenchymal Stromal Cell-Derived Extracellular Vesicles Carrying MicroRNAs. J. Am. Soc. Nephrol. 2015, 26, 2349–2360. [Google Scholar] [CrossRef] [Green Version]
  61. Wang, B.; Yao, K.; Huuskes, B.M.; Shen, H.-H.; Zhuang, J.; Godson, C.; Brennan, E.P.; Wilkinson-Berka, J.L.; Wise, A.F.; Ricardo, S.D. Mesenchymal Stem Cells Deliver Exogenous MicroRNA-let7c via Exosomes to Attenuate Renal Fibrosis. Mol. Ther. 2016, 24, 1290–1301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. He, J.; Wang, Y.; Lu, X.; Zhu, B.; Pei, X.; Wu, J.; Zhao, W. Micro-vesicles derived from bone marrow stem cells protect the kidney both in vivo and in vitro by microRNA-dependent repairing. Nephrology 2015, 20, 591–600. [Google Scholar] [CrossRef] [PubMed]
  63. Zhou, Y.; Fang, L.; Yu, Y.; Niu, J.; Jiang, L.; Cao, H.; Sun, Q.; Zen, K.; Dai, C.; Yang, J. Erythropoietin protects the tubular basement membrane by promoting the bone marrow to release extracellular vesicles containing tPA-targeting miR-144. Am. J. Physiol. Physiol. 2016, 310, F27–F40. [Google Scholar] [CrossRef] [Green Version]
  64. Wang, Y.; Lu, X.; He, J.; Zhao, W. Influence of erythropoietin on microvesicles derived from mesenchymal stem cells protecting renal function of chronic kidney disease. Stem Cell Res. Ther. 2015, 6, 100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Kholia, S.; Sanchez, M.B.H.; Cedrino, M.; Papadimitriou, E.; Tapparo, M.; Deregibus, M.C.; Bruno, S.; Antico, F.; Brizzi, M.F.; Quesenberry, P.J.; et al. Mesenchymal Stem Cell Derived Extracellular Vesicles Ameliorate Kidney Injury in Aristolochic Acid Nephropathy. Front. Cell Dev. Biol. 2020, 8, 188. [Google Scholar] [CrossRef] [PubMed]
  66. Grange, C.; Tritta, S.; Tapparo, M.; Cedrino, M.; Tetta, C.; Camussi, G.; Brizzi, M.F. Stem cell-derived extracellular vesicles inhibit and revert fibrosis progression in a mouse model of diabetic nephropathy. Sci. Rep. 2019, 9, 4468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Lin, K.-C.; Yip, H.-K.; Shao, P.-L.; Wu, S.-C.; Chen, K.-H.; Chen, Y.-T.; Yang, C.-C.; Sun, C.-K.; Kao, G.-S.; Chen, S.-Y.; et al. Combination of adipose-derived mesenchymal stem cells (ADMSC) and ADMSC-derived exosomes for protecting kidney from acute ischemia–reperfusion injury. Int. J. Cardiol. 2016, 216, 173–185. [Google Scholar] [CrossRef]
  68. Gao, F.; Zuo, B.; Wang, Y.; Li, S.; Yang, J.; Sun, D. Protective function of exosomes from adipose tissue-derived mesenchymal stem cells in acute kidney injury through SIRT1 pathway. Life Sci. 2020, 255, 117719. [Google Scholar] [CrossRef]
  69. 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]
  70. Eirin, A.; Zhu, X.-Y.; Jonnada, S.; Lerman, A.; van Wijnen, A.J.; Lerman, L.O. Mesenchymal Stem Cell-Derived Extracellular Vesicles Improve the Renal Microvasculature in Metabolic Renovascular Disease in Swine. Cell Transplant. 2018, 27, 1080–1095. [Google Scholar] [CrossRef] [Green Version]
  71. Zhu, F.; Shin, O.L.S.C.L.; Pei, G.; Hu, Z.; Yang, J.; Zhu, H.; Wang, M.; Mou, J.; Sun, J.; Wang, Y.; et al. Adipose-derived mesenchymal stem cells employed exosomes to attenuate AKI-CKD transition through tubular epithelial cell dependent Sox9 activation. Oncotarget 2017, 8, 70707–70726. [Google Scholar] [CrossRef] [Green Version]
  72. Chen, L.; Wang, Y.; Li, S.; Zuo, B.; Zhang, X.; Wang, F.; Sun, D. Exosomes derived from GDNF-modified human adipose mesenchymal stem cells ameliorate peritubular capillary loss in tubulointerstitial fibrosis by activating the SIRT1/eNOS signaling pathway. Theranostics 2020, 10, 9425–9442. [Google Scholar] [CrossRef] [PubMed]
  73. Sanchez, M.B.H.; Bruno, S.; Grange, C.; Tapparo, M.; Cantaluppi, V.; Tetta, C.; Camussi, G. Human liver stem cells and derived extracellular vesicles improve recovery in a murine model of acute kidney injury. Stem Cell Res. Ther. 2014, 5, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Kholia, S.; Sanchez, M.B.H.; Cedrino, M.; Papadimitriou, E.; Tapparo, M.; Deregibus, M.C.; Brizzi, M.F.; Tetta, C.; Camussi, G. Human Liver Stem Cell-Derived Extracellular Vesicles Prevent Aristolochic Acid-Induced Kidney Fibrosis. Front. Immunol. 2018, 9, 1639. [Google Scholar] [CrossRef] [Green Version]
  75. Choi, H.Y.; Moon, S.J.; Ratliff, B.B.; Ahn, S.H.; Jung, A.; Lee, M.; Lee, S.; Lim, B.J.; Kim, B.S.; Plotkin, M.D.; et al. Microparticles from Kidney-Derived Mesenchymal Stem Cells Act as Carriers of Proangiogenic Signals and Contribute to Recovery from Acute Kidney Injury. PLoS ONE 2014, 9, e87853. [Google Scholar] [CrossRef] [Green Version]
  76. 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]
  77. Choi, H.Y.; Lee, H.G.; Kim, B.S.; Ahn, S.H.; Jung, A.; Lee, M.; Lee, J.E.; Kim, H.J.; Ha, S.K.; Park, H.C. Mesenchymal stem cell-derived microparticles ameliorate peritubular capillary rarefaction via inhibition of endothelial-mesenchymal transition and decrease tubulointerstitial fibrosis in unilateral ureteral obstruction. Stem Cell Res. Ther. 2015, 6, 18. [Google Scholar] [CrossRef] [Green Version]
  78. 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]
  79. Zou, X.; Zhang, G.; Cheng, Z.; Yin, D.; Du, T.; Ju, G.; Miao, S.; Liu, G.; Lu, M.; Zhu, Y. Microvesicles derived from human Wharton’s Jelly mesenchymal stromal cells ameliorate renal ischemia-reperfusion injury in rats by suppressing CX3CL1. Stem Cell Res. Ther. 2014, 5, 40. [Google Scholar] [CrossRef] [Green Version]
  80. Ju, G.-Q.; Cheng, J.; Zhong, L.; Wu, S.; Zou, X.-Y.; Zhang, G.-Y.; Gu, D.; Miao, S.; Zhu, Y.-J.; Sun, J.; et al. Microvesicles Derived from Human Umbilical Cord Mesenchymal Stem Cells Facilitate Tubular Epithelial Cell Dedifferentiation and Growth via Hepatocyte Growth Factor Induction. PLoS ONE 2015, 10, e0121534. [Google Scholar] [CrossRef] [Green Version]
  81. Gu, D.; Zou, X.; Ju, G.; Zhang, G.; Bao, E.; Zhu, Y. Mesenchymal Stromal Cells Derived Extracellular Vesicles Ameliorate Acute Renal Ischemia Reperfusion Injury by Inhibition of Mitochondrial Fission through miR-30. Stem Cells Int. 2016, 2016, 2093940. [Google Scholar] [CrossRef] [Green Version]
  82. Zhang, G.; Zou, X.; Miao, S.; Chen, J.; Du, T.; Zhong, L.; Ju, G.; Liu, G.; Zhu, Y. The Anti-Oxidative Role of Micro-Vesicles Derived from Human Wharton-Jelly Mesenchymal Stromal Cells through NOX2/gp91(phox) Suppression in Alleviating Renal Ischemia-Reperfusion Injury in Rats. PLoS ONE 2014, 9, e92129. [Google Scholar] [CrossRef]
  83. Zhang, G.; Zou, X.; Huang, Y.; Wang, F.; Miao, S.; Liu, G.; Chen, M.; Zhu, Y. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Protect Against Acute Kidney Injury Through Anti-Oxidation by Enhancing Nrf2/ARE Activation in Rats. Kidney Blood Press. Res. 2016, 41, 119–128. [Google Scholar] [CrossRef]
  84. Zou, X.; Gu, D.; Xing, X.; Cheng, Z.; Gong, D.; Zhang, G.; Zhu, Y. Human mesenchymal stromal cell-derived extracellular vesicles alleviate renal ischemic reperfusion injury and enhance angiogenesis in rats. Am. J. Transl. Res. 2016, 8, 4289–4299. [Google Scholar] [PubMed]
  85. Wang, B.; Jia, H.; Zhang, B.; Wang, J.; Ji, C.; Zhu, X.; Yan, Y.; Yin, L.; Yu, J.; Qian, H.; et al. Pre-incubation with hucMSC-exosomes prevents cisplatin-induced nephrotoxicity by activating autophagy. Stem Cell Res. Ther. 2017, 8, 75. [Google Scholar] [CrossRef] [Green Version]
  86. Zhang, Z.-Y.; Hou, Y.-P.; Zou, X.-Y.; Xing, X.-Y.; Ju, G.-Q.; Zhong, L.; Sun, J. Oct-4 Enhanced the Therapeutic Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles in Acute Kidney Injury. Kidney Blood Press. Res. 2020, 45, 95–108. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, R.; Zhu, Y.; Li, Y.; Liu, W.; Yin, L.; Yin, S.; Ji, C.; Hu, Y.; Wang, Q.; Zhou, X.; et al. Human umbilical cord mesenchymal stem cell exosomes alleviate sepsis-associated acute kidney injury via regulating microRNA-146b expression. Biotechnol. Lett. 2020, 42, 669–679. [Google Scholar] [CrossRef]
  88. Cao, J.-Y.; Wang, B.; Tang, T.-T.; Wen, Y.; Li, Z.-L.; Feng, S.-T.; Wu, M.; Liu, D.; Yin, D.; Ma, K.-L.; et al. Exosomal miR-125b-5p deriving from mesenchymal stem cells promotes tubular repair by suppression of p53 in ischemic acute kidney injury. Theranostics 2021, 11, 5248–5266. [Google Scholar] [CrossRef]
  89. Ji, C.; Zhang, J.; Zhu, Y.; Shi, H.; Yin, S.; Sun, F.; Wang, Q.; Zhang, L.; Yan, Y.; Zhang, X.; et al. Exosomes derived from hucMSC attenuate renal fibrosis through CK1δ/β-TRCP-mediated YAP degradation. Cell Death Dis. 2020, 11, 327. [Google Scholar] [CrossRef] [PubMed]
  90. Liu, Y.; Cui, J.; Wang, H.; Hezam, K.; Zhao, X.; Huang, H.; Chen, S.; Han, Z.; Han, Z.-C.; Guo, Z.; et al. Enhanced therapeutic effects of MSC-derived extracellular vesicles with an injectable collagen matrix for experimental acute kidney injury treatment. Stem Cell Res. Ther. 2020, 11, 161. [Google Scholar] [CrossRef] [Green Version]
  91. Zhang, C.; Shang, Y.; Chen, X.; Midgley, A.C.; Wang, Z.; Zhu, D.; Wu, J.; Chen, P.; Wu, L.; Wang, X.; et al. Supramolecular Nanofibers Containing Arginine-Glycine-Aspartate (RGD) Peptides Boost Therapeutic Efficacy of Extracellular Vesicles in Kidney Repair. ACS Nano 2020, 14, 12133–12147. [Google Scholar] [CrossRef]
  92. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.C.; Krause, D.S.; Deans, R.J.; Keating, A.; Prockop, D.J.; Horwitz, E.M. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  93. Witwer, K.W.; Van Balkom, B.W.M.; Bruno, S.; Choo, A.; Dominici, M.; Gimona, M.; Hill, A.F.; De Kleijn, D.; Koh, M.; Lai, R.C.; et al. Defining mesenchymal stromal cell (MSC)-derived small extracellular vesicles for therapeutic applications. J. Extracell. Vesicles 2019, 8, 1609206. [Google Scholar] [CrossRef] [Green Version]
  94. Abumoawad, A.; Saad, A.; Ferguson, C.M.; Eirin, A.; Herrmann, S.M.; Hickson, L.J.; Goksu, B.B.; Bendel, E.; Misra, S.; Glockner, J.; et al. In a Phase 1a escalating clinical trial, autologous mesenchymal stem cell infusion for renovascular disease increases blood flow and the glomerular filtration rate while reducing inflammatory biomarkers and blood pressure. Kidney Int. 2020, 97, 793–804. [Google Scholar] [CrossRef] [Green Version]
  95. Ban, T.H.; Lee, S.; Kim, H.D.; Ko, E.J.; Kim, B.-M.; Kim, K.-W.; Chung, B.H.; Yang, C.W. Clinical Trial of Allogeneic Mesenchymal Stem Cell Therapy for Chronic Active Antibody-Mediated Rejection in Kidney Transplant Recipients Unresponsive to Rituximab and Intravenous Immunoglobulin. Stem Cells Int. 2021, 2021, 6672644. [Google Scholar] [CrossRef] [PubMed]
  96. Večerić-Haler, Ž.; Sever, M.; Kojc, N.; Halloran, P.F.; Boštjančič, E.; Mlinšek, G.; Oblak, M.; Poženel, P.; Švajger, U.; Hartman, K.; et al. Autologous Mesenchymal Stem Cells for Treatment of Chronic Active Antibody-Mediated Kidney Graft Rejection: Report of the Phase I/II Clinical Trial Case Series. Transpl. Int. 2022, 35, 10772. [Google Scholar] [CrossRef] [PubMed]
  97. Yang, Y.; Gao, J.; Wang, S.; Wang, W.; Zhu, F.-L.; Wang, X.; Liang, S.; Feng, Z.; Lin, S.; Zhang, L.; et al. Efficacy of umbilical cord mesenchymal stem cell transfusion for the treatment of severe AKI: A protocol for a randomised controlled trial. BMJ Open 2022, 12, e047622. [Google Scholar] [CrossRef] [PubMed]
  98. Welsh, J.A.; van der Pol, E.; Arkesteijn, G.J.A.; Bremer, M.; Brisson, A.; Coumans, F.; Dignat-George, F.; Duggan, E.; Ghiran, I.; Giebel, B.; et al. MIFlowCyt-EV: A framework for standardized reporting of extracellular vesicle flow cytometry experiments. J. Extracell. Vesicles 2020, 9, 1713526. [Google Scholar] [CrossRef] [PubMed]
  99. Silva, A.K.; Morille, M.; Piffoux, M.; Arumugam, S.; Mauduit, P.; Larghero, J.; Bianchi, A.; Aubertin, K.; Blanc-Brude, O.; Noël, D.; et al. Development of extracellular vesicle-based medicinal products: A position paper of the group “Extracellular Vesicle translatiOn to clinicaL perspectiVEs—EVOLVE France”. Adv. Drug Deliv. Rev. 2021, 179, 114001. [Google Scholar] [CrossRef] [PubMed]
  100. Swaminathan, M.; Stafford-Smith, M.; Chertow, G.M.; Warnock, D.G.; Paragamian, V.; Brenner, R.M.; Lellouche, F.; Fox-Robichaud, A.; Atta, M.G.; Melby, S.; et al. Allogeneic Mesenchymal Stem Cells for Treatment of AKI after Cardiac Surgery. J. Am. Soc. Nephrol. 2018, 29, 260–267. [Google Scholar] [CrossRef] [Green Version]
  101. Tögel, F.E.; Westenfelder, C. Kidney Protection and Regeneration Following Acute Injury: Progress Through Stem Cell Therapy. Am. J. Kidney Dis. 2012, 60, 1012–1022. [Google Scholar] [CrossRef] [PubMed]
  102. Westenfelder, C.; Togel, F.E. Protective actions of administered mesenchymal stem cells in acute kidney injury: Relevance to clinical trials. Kidney Int. Suppl. 2011, 1, 103–106. [Google Scholar] [CrossRef] [Green Version]
  103. Lee, M.S.; Lee, F.-Y.; Chen, Y.-L.; Sung, P.-H.; Chiang, H.-J.; Chen, K.-H.; Huang, T.-H.; Chen, Y.-L.; Chiang, J.Y.; Yin, T.-C.; et al. Investigated the safety of intra-renal arterial transfusion of autologous CD34+ cells and time courses of creatinine levels, endothelial dysfunction biomarkers and micro-RNAs in chronic kidney disease patients-phase I clinical trial. Oncotarget 2017, 8, 17750–17762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Yang, C.-C.; Sung, P.-H.; Cheng, B.-C.; Li, Y.-C.; Chen, Y.-L.; Lee, M.S.; Yip, H.-K. Safety and efficacy of intrarenal arterial autologous CD34+ cell transfusion in patients with chronic kidney disease: A randomized, open-label, controlled phase II clinical trial. Stem Cells Transl. Med. 2020, 9, 827–838. [Google Scholar] [CrossRef] [Green Version]
  105. Villanueva, S.; González, F.; Lorca, E.; Tapia, A.; López, V.G.; Strodthoff, R.; Fajre, F.; Carreño, J.E.; Valjalo, R.; Vergara, C.; et al. Adipose tissue-derived mesenchymal stromal cells for treating chronic kidney disease: A pilot study assessing safety and clinical feasibility. Kidney Res. Clin. Pract. 2019, 38, 176–185. [Google Scholar] [CrossRef] [Green Version]
  106. Makhlough, A.; Shekarchian, S.; Moghadasali, R.; Einollahi, B.; Hosseini, S.E.; Jaroughi, N.; Bolurieh, T.; Baharvand, H.; Aghdami, N. Safety and tolerability of autologous bone marrow mesenchymal stromal cells in ADPKD patients. Stem Cell Res. Ther. 2017, 8, 116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Gu, F.; Wang, D.; Zhang, H.; Feng, X.; Gilkeson, G.S.; Shi, S.; Sun, L. Allogeneic mesenchymal stem cell transplantation for lupus nephritis patients refractory to conventional therapy. Clin. Rheumatol. 2014, 33, 1611–1619. [Google Scholar] [CrossRef]
  108. Wang, D.; Li, J.; Zhang, Y.; Zhang, M.; Chen, J.; Li, X.; Hu, X.; Jiang, S.; Shi, S.; Sun, L. Umbilical cord mesenchymal stem cell transplantation in active and refractory systemic lupus erythematosus: A multicenter clinical study. Arthritis Res. Ther. 2014, 16, R79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Sun, L.; Wang, D.; Liang, J.; Zhang, H.; Feng, X.; Wang, H.; Hua, B.; Liu, B.; Ye, S.; Hu, X.; et al. Umbilical cord mesenchymal stem cell transplantation in severe and refractory systemic lupus erythematosus. Arthritis Rheum. 2010, 62, 2467–2475. [Google Scholar] [CrossRef]
  110. Barbado, J.; Tabera, S.; Sánchez, A.; García-Sancho, J. Therapeutic potential of allogeneic mesenchymal stromal cells transplantation for lupus nephritis. Lupus 2018, 27, 2161–2165. [Google Scholar] [CrossRef]
  111. Deng, D.; Zhang, P.; Guo, Y.; Lim, T.O. A randomised double-blind, placebo-controlled trial of allogeneic umbilical cord-derived mesenchymal stem cell for lupus nephritis. Ann. Rheum. Dis. 2017, 76, 1436–1439. [Google Scholar] [CrossRef]
  112. Jeppesen, D.K.; Fenix, A.M.; Franklin, J.L.; Higginbotham, J.N.; Zhang, Q.; Zimmerman, L.J.; Liebler, D.C.; Ping, J.; Liu, Q.; Evans, R.; et al. Reassessment of Exosome Composition. Cell 2019, 177, 428–445.e418. [Google Scholar] [CrossRef] [Green Version]
  113. Subramanyam, D.; Lamouille, S.; Judson, R.L.; Liu, J.Y.; Bucay, N.; Derynck, R.; Blelloch, R. Multiple targets of miR-302 and miR-372 promote reprogramming of human fibroblasts to induced pluripotent stem cells. Nat. Biotechnol. 2011, 29, 443–448. [Google Scholar] [CrossRef] [PubMed]
  114. Liu, X.; Wei, Q.; Lu, L.; Cui, S.; Ma, K.; Zhang, W.; Ma, F.; Li, H.; Fu, X.; Zhang, C. Immunomodulatory potential of mesenchymal stem cell-derived extracellular vesicles: Targeting immune cells. Front. Immunol. 2023, 14, 1094685. [Google Scholar] [CrossRef]
  115. Zhang, C.; Liao, W.; Li, W.; Li, M.; Xu, X.; Sun, H.; Xue, Y.; Liu, L.; Qiu, J.; Zhang, C.; et al. Human umbilical cord mesenchymal stem cells derived extracellular vesicles alleviate salpingitis by promoting M1-to-M2 transformation. Front. Physiol. 2023, 14, 1131701. [Google Scholar] [CrossRef] [PubMed]
  116. Zhao, J.; Li, X.; Hu, J.; Chen, F.; Qiao, S.; Sun, X.; Gao, L.; Xie, J.; Xu, B. Mesenchymal stromal cell-derived exosomes attenuate myocardial ischaemia-reperfusion injury through miR-182-regulated macrophage polarization. Cardiovasc. Res. 2019, 115, 1205–1216. [Google Scholar] [CrossRef] [Green Version]
  117. Deng, H.; Wu, L.; Liu, M.; Zhu, L.; Chen, Y.; Zhou, H.; Shi, X.; Wei, J.; Zheng, L.; Hu, X.; et al. Bone Marrow Mesenchymal Stem Cell-Derived Exosomes Attenuate LPS-Induced ARDS by Modulating Macrophage Polarization Through Inhibiting Glycolysis in Macrophages. Shock 2020, 54, 828–843. [Google Scholar] [CrossRef] [PubMed]
  118. Zhang, B.; Yin, Y.; Lai, R.C.; Tan, S.S.; Choo, A.B.H.; Lim, S.K. Mesenchymal Stem Cells Secrete Immunologically Active Exosomes. Stem Cells Dev. 2014, 23, 1233–1244. [Google Scholar] [CrossRef]
  119. Lu, F.-B.; Chen, D.-Z.; Chen, L.; Hu, E.-D.; Wu, J.-L.; Li, H.; Gong, Y.-W.; Lin, Z.; Wang, X.-D.; Li, J.; et al. Attenuation of Experimental Autoimmune Hepatitis in Mice with Bone Mesenchymal Stem Cell-Derived Exosomes Carrying MicroRNA-223-3p. Mol. Cells 2019, 42, 906–918. [Google Scholar] [CrossRef]
  120. Hou, L.; Zhu, Z.; Jiang, F.; Zhao, J.; Jia, Q.; Wang, H.; Xue, W.; Wang, Y.; Tian, L. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles alleviated silica induced lung inflammation and fibrosis in mice via circPWWP2A/miR-223–3p/NLRP3 axis. Ecotoxicol. Environ. Saf. 2023, 251, 114537. [Google Scholar] [CrossRef]
  121. Sun, J.; Sun, X.; Chen, J.; Liao, X.; He, Y.; Wang, J.; Chen, R.; Hu, S.; Qiu, C. microRNA-27b shuttled by mesenchymal stem cell-derived exosomes prevents sepsis by targeting JMJD3 and downregulating NF-κB signaling pathway. Stem Cell Res. Ther. 2021, 12, 14. [Google Scholar] [CrossRef]
  122. Chang, Q.; Hao, Y.; Wang, Y.; Zhou, Y.; Zhuo, H.; Zhao, G. Bone marrow mesenchymal stem cell-derived exosomal microRNA-125a promotes M2 macrophage polarization in spinal cord injury by downregulating IRF5. Brain Res. Bull. 2021, 170, 199–210. [Google Scholar] [CrossRef]
  123. Ma, J.; Chen, L.; Zhu, X.; Li, Q.; Hu, L.; Li, H. Mesenchymal stem cell-derived exosomal miR-21a-5p promotes M2 macrophage polarization and reduces macrophage infiltration to attenuate atherosclerosis. Acta Biochim. Biophys. Sin. 2021, 53, 1227–1236. [Google Scholar] [CrossRef] [PubMed]
  124. Li, J.; Xue, H.; Li, T.; Chu, X.; Xin, D.; Xiong, Y.; Qiu, W.; Gao, X.; Qian, M.; Xu, J.; et al. Exosomes derived from mesenchymal stem cells attenuate the progression of atherosclerosis in ApoE−/− mice via miR-let7 mediated infiltration and polarization of M2 macrophage. Biochem. Biophys. Res. Commun. 2019, 510, 565–572. [Google Scholar] [CrossRef] [PubMed]
  125. Liu, H.; Liang, Z.; Wang, F.; Zhou, C.; Zheng, X.; Hu, T.; He, X.; Wu, X.; Lan, P. Exosomes from mesenchymal stromal cells reduce murine colonic inflammation via a macrophage-dependent mechanism. J. Clin. Investig. 2019, 4, e131273. [Google Scholar] [CrossRef] [Green Version]
  126. Favaro, E.; Carpanetto, A.; Caorsi, C.; Giovarelli, M.; Angelini, C.; Cavallo-Perin, P.; Tetta, C.; Camussi, G.; Zanone, M.M. Human mesenchymal stem cells and derived extracellular vesicles induce regulatory dendritic cells in type 1 diabetic patients. Diabetologia 2016, 59, 325–333. [Google Scholar] [CrossRef] [Green Version]
  127. Guo, R.; Liang, Q.; He, Y.; Wang, C.; Jiang, J.; Chen, T.; Zhang, D.; Hu, K. Mesenchymal Stromal Cells-Derived Extracellular Vesicles Regulate Dendritic Cell Functions in Dry Eye Disease. Cells 2022, 12, 33. [Google Scholar] [CrossRef]
  128. Shigemoto-Kuroda, T.; Oh, J.Y.; Kim, D.-K.; Jeong, H.J.; Park, S.Y.; Lee, H.J.; Park, J.W.; Kim, T.W.; An, S.Y.; Prockop, D.J.; et al. MSC-derived Extracellular Vesicles Attenuate Immune Responses in Two Autoimmune Murine Models: Type 1 Diabetes and Uveoretinitis. Stem Cell Rep. 2017, 8, 1214–1225. [Google Scholar] [CrossRef] [Green Version]
  129. Wu, X.-Q.; Yan, T.-Z.; Wang, Z.-W.; Wu, X.; Cao, G.-H.; Zhang, C. BM-MSCs-derived microvesicles promote allogeneic kidney graft survival through enhancing micro-146a expression of dendritic cells. Immunol. Lett. 2017, 191, 55–62. [Google Scholar] [CrossRef] [PubMed]
  130. Reis, M.; Mavin, E.; Nicholson, L.; Green, K.; Dickinson, A.M.; Wang, X.-N. Mesenchymal Stromal Cell-Derived Extracellular Vesicles Attenuate Dendritic Cell Maturation and Function. Front. Immunol. 2018, 9, 2538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Zou, X.; Gu, D.; Zhang, G.; Zhong, L.; Cheng, Z.; Liu, G.; Zhu, Y. NK Cell Regulatory Property is Involved in the Protective Role of MSC-Derived Extracellular Vesicles in Renal Ischemic Reperfusion Injury. Hum. Gene Ther. 2016, 27, 926–935. [Google Scholar] [CrossRef]
  132. Fan, Y.; Herr, F.; Vernochet, A.; Mennesson, B.; Oberlin, E.; Durrbach, A.; Patterson, A.L.; George, J.; Chatterjee, A.; Carpenter, T.; et al. Human Fetal Liver Mesenchymal Stem Cell-Derived Exosomes Impair Natural Killer Cell Function. Stem Cells Dev. 2019, 28, 44–55. [Google Scholar] [CrossRef]
  133. Mahmoudi, M.; Taghavi-Farahabadi, M.; Namaki, S.; Baghaei, K.; Rayzan, E.; Rezaei, N.; Hashemi, S.M. Exosomes derived from mesenchymal stem cells improved function and survival of neutrophils from severe congenital neutropenia patients in vitro. Hum. Immunol. 2019, 80, 990–998. [Google Scholar] [CrossRef] [PubMed]
  134. Taghavi-Farahabadi, M.; Mahmoudi, M.; Rezaei, N.; Hashemi, S.M. Wharton’s Jelly Mesenchymal Stem Cells Exosomes and Conditioned Media Increased Neutrophil Lifespan and Phagocytosis Capacity. Immunol. Investig. 2020, 50, 1042–1057. [Google Scholar] [CrossRef]
  135. Loh, J.T.; Zhang, B.; Teo, J.K.H.; Lai, R.C.; Choo, A.B.H.; Lam, K.-P.; Lim, S.K. Mechanism for the attenuation of neutrophil and complement hyperactivity by MSC exosomes. Cytotherapy 2022, 24, 711–719. [Google Scholar] [CrossRef]
  136. Yao, J.; Zheng, J.; Cai, J.; Zeng, K.; Zhou, C.; Zhang, J.; Li, S.; Li, H.; Chen, L.; He, L.; et al. Extracellular vesicles derived from human umbilical cord mesenchymal stem cells alleviate rat hepatic ischemia-reperfusion injury by suppressing oxidative stress and neutrophil inflammatory response. FASEB J. 2019, 33, 1695–1710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Liu, J.; Kuwabara, A.; Kamio, Y.; Hu, S.; Park, J.; Hashimoto, T.; Lee, J.-W. Human Mesenchymal Stem Cell-Derived Microvesicles Prevent the Rupture of Intracranial Aneurysm in Part by Suppression of Mast Cell Activation via a PGE2-Dependent Mechanism. Stem Cells 2016, 34, 2943–2955. [Google Scholar] [CrossRef] [Green Version]
  138. Lin, T.-Y.; Chang, T.-M.; Huang, H.-C. Extracellular Vesicles Derived from Human Umbilical Cord Mesenchymal Stem Cells Attenuate Mast Cell Activation. Antioxidants 2022, 11, 2279. [Google Scholar] [CrossRef] [PubMed]
  139. Álvarez, V.; Sánchez-Margallo, F.M.; Macías-García, B.; Gómez-Serrano, M.; Jorge, I.; Vázquez, J.; Blázquez, R.; Casado, J.G. The immunomodulatory activity of extracellular vesicles derived from endometrial mesenchymal stem cells on CD4+ T cells is partially mediated by TGFbeta. J. Tissue Eng. Regen. Med. 2018, 12, 2088–2098. [Google Scholar] [CrossRef]
  140. Crain, S.K.; Robinson, S.R.; Thane, K.E.; Davis, A.M.; Meola, D.M.; Barton, B.A.; Yang, V.K.; Hoffman, A.M. Extracellular Vesicles from Wharton’s Jelly Mesenchymal Stem Cells Suppress CD4 Expressing T Cells Through Transforming Growth Factor Beta and Adenosine Signaling in a Canine Model. Stem Cells Dev. 2019, 28, 212–226. [Google Scholar] [CrossRef] [PubMed]
  141. Kerkelä, E.; Laitinen, A.; Räbinä, J.; Valkonen, S.; Takatalo, M.; Larjo, A.; Veijola, J.; Lampinen, M.; Siljander, P.; Lehenkari, P.; et al. Adenosinergic Immunosuppression by Human Mesenchymal Stromal Cells Requires Co-Operation with T cells. Stem Cells 2016, 34, 781–790. [Google Scholar] [CrossRef] [Green Version]
  142. Khare, D.; Or, R.; Resnick, I.; Barkatz, C.; Almogi-Hazan, O.; Avni, B. Mesenchymal Stromal Cell-Derived Exosomes Affect mRNA Expression and Function of B-Lymphocytes. Front. Immunol. 2018, 9, 3053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Yan, Y.; Jiang, W.; Tan, Y.; Zou, S.; Zhang, H.; Mao, F.; Gong, A.; Qian, H.; Xu, W. hucMSC Exosome-Derived GPX1 Is Required for the Recovery of Hepatic Oxidant Injury. Mol. Ther. 2017, 25, 465–479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Tan, C.Y.; Lai, R.C.; Wong, W.; Dan, Y.Y.; Lim, S.-K.; Ho, H.K. Mesenchymal stem cell-derived exosomes promote hepatic regeneration in drug-induced liver injury models. Stem Cell Res. Ther. 2014, 5, 76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Wang, K. Molecular mechanisms of hepatic apoptosis. Cell Death Dis. 2014, 5, e996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Li, C.; Jiao, G.; Wu, W.; Wang, H.; Ren, S.; Zhang, L.; Zhou, H.; Liu, H.; Chen, Y. Exosomes from Bone Marrow Mesenchymal Stem Cells Inhibit Neuronal Apoptosis and Promote Motor Function Recovery via the Wnt/β-catenin Signaling Pathway. Cell Transplant. 2019, 28, 1373–1383. [Google Scholar] [CrossRef] [PubMed]
  147. Li, S.; Stöckl, S.; Lukas, C.; Götz, J.; Herrmann, M.; Federlin, M.; Grässel, S. hBMSC-Derived Extracellular Vesicles Attenuate IL-1β-Induced Catabolic Effects on OA-Chondrocytes by Regulating Pro-inflammatory Signaling Pathways. Front. Bioeng. Biotechnol. 2020, 8, 603598. [Google Scholar] [CrossRef]
  148. Arslan, F.; Lai, R.C.; Smeets, M.B.; Akeroyd, L.; Choo, A.; Aguor, E.N.E.; Timmers, L.; Van Rijen, H.V.; Doevendans, P.A.; Pasterkamp, G.; et al. Mesenchymal stem cell-derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res. 2013, 10, 301–312. [Google Scholar] [CrossRef] [Green Version]
  149. Lindoso, R.S.; Collino, F.; Bruno, S.; Araujo, D.S.; Sant’Anna, J.F.; Tetta, C.; Provero, P.; Quesenberry, P.J.; Vieyra, A.; Einicker-Lamas, M.; et al. Extracellular Vesicles Released from Mesenchymal Stromal Cells Modulate miRNA in Renal Tubular Cells and Inhibit ATP Depletion Injury. Stem Cells Dev. 2014, 23, 1809–1819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Haga, H.; Yan, I.K.; Takahashi, K.; Matsuda, A.; Patel, T. Extracellular Vesicles from Bone Marrow-Derived Mesenchymal Stem Cells Improve Survival from Lethal Hepatic Failure in Mice. Stem Cells Transl. Med. 2017, 6, 1262–1272. [Google Scholar] [CrossRef]
  151. Feng, Y.; Huang, W.; Wani, M.; Yu, X.; Ashraf, M. Ischemic Preconditioning Potentiates the Protective Effect of Stem Cells through Secretion of Exosomes by Targeting Mecp2 via miR-22. PLoS ONE 2014, 9, e88685. [Google Scholar] [CrossRef]
  152. Yu, B.; Gong, M.; Wang, Y.; Millard, R.W.; Pasha, Z.; Yang, Y.; Ashraf, M.; Xu, M. Cardiomyocyte Protection by GATA-4 Gene Engineered Mesenchymal Stem Cells Is Partially Mediated by Translocation of miR-221 in Microvesicles. PLoS ONE 2013, 8, e73304. [Google Scholar] [CrossRef] [Green Version]
  153. Zhu, L.-P.; Tian, T.; Wang, J.-Y.; He, J.-N.; Chen, T.; Pan, M.; Xu, L.; Zhang, H.-X.; Qiu, X.-T.; Li, C.-C.; et al. Hypoxia-elicited mesenchymal stem cell-derived exosomes facilitates cardiac repair through miR-125b-mediated prevention of cell death in myocardial infarction. Theranostics 2018, 8, 6163–6177. [Google Scholar] [CrossRef] [PubMed]
  154. Huang, Y.; He, B.; Wang, L.; Yuan, B.; Shu, H.; Zhang, F.; Sun, L. Bone marrow mesenchymal stem cell-derived exosomes promote rotator cuff tendon-bone healing by promoting angiogenesis and regulating M1 macrophages in rats. Stem Cell Res. Ther. 2020, 11, 496. [Google Scholar] [CrossRef]
  155. Lou, G.; Yang, Y.; Liu, F.; Ye, B.; Chen, Z.; Zheng, M.; Liu, Y. MiR-122 modification enhances the therapeutic efficacy of adipose tissue-derived mesenchymal stem cells against liver fibrosis. J. Cell. Mol. Med. 2017, 21, 2963–2973. [Google Scholar] [CrossRef]
  156. Zhang, B.; Wang, M.; Gong, A.; Zhang, X.; Wu, X.; Zhu, Y.; Shi, H.; Wu, L.; Zhu, W.; Qian, H.; et al. HucMSC-Exosome Mediated-Wnt4 Signaling Is Required for Cutaneous Wound Healing. Stem Cells 2015, 33, 2158–2168. [Google Scholar] [CrossRef]
  157. Chew, J.R.J.; Chuah, S.J.; Teo, K.Y.W.; Zhang, S.; Lai, R.C.; Fu, J.H.; Lim, L.P.; Lim, S.K.; Toh, W.S. Mesenchymal stem cell exosomes enhance periodontal ligament cell functions and promote periodontal regeneration. Acta Biomater. 2019, 89, 252–264. [Google Scholar] [CrossRef]
  158. Mao, Q.; Nguyen, P.D.; Shanti, R.M.; Shi, S.; Shakoori, P.; Zhang, Q.; Le, A.D. Gingiva-Derived Mesenchymal Stem Cell-Extracellular Vesicles Activate Schwann Cell Repair Phenotype and Promote Nerve Regeneration. Tissue Eng. Part A 2019, 25, 887–900. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Tomasoni, S.; Longaretti, L.; Rota, C.; Morigi, M.; Conti, S.; Gotti, E.; Capelli, C.; Introna, M.; Remuzzi, G.; Benigni, A.; et al. Transfer of Growth Factor Receptor mRNA Via Exosomes Unravels the Regenerative Effect of Mesenchymal Stem Cells. Stem Cells Dev. 2013, 22, 772–780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Papait, A.; Silini, A.R.; Gazouli, M.; Malvicini, R.; Muraca, M.; O’driscoll, L.; Pacienza, N.; Toh, W.S.; Yannarelli, G.; Ponsaerts, P.; et al. Perinatal derivatives: How to best validate their immunomodulatory functions. Front. Bioeng. Biotechnol. 2022, 10, 981061. [Google Scholar] [CrossRef]
  161. Vogel, W.; Grünebach, F.; Messam, C.A.; Kanz, L.; Brugger, W.; Bühring, H.-J. Heterogeneity among human bone marrow-derived mesenchymal stem cells and neural progenitor cells. Haematologica 2003, 88, 126–133. [Google Scholar] [PubMed]
  162. Chinnadurai, R.; Rajan, D.; Qayed, M.; Arafat, D.; Garcia, M.; Liu, Y.; Kugathasan, S.; Anderson, L.J.; Gibson, G.; Galipeau, J. Potency Analysis of Mesenchymal Stromal Cells Using a Combinatorial Assay Matrix Approach. Cell Rep. 2018, 22, 2504–2517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Lechanteur, C.; Briquet, A.; Bettonville, V.; Baudoux, E.; Beguin, Y. MSC Manufacturing for Academic Clinical Trials: From a Clinical-Grade to a Full GMP-Compliant Process. Cells 2021, 10, 1320. [Google Scholar] [CrossRef]
  164. Herzig, M.C.; Delavan, C.P.; Jensen, K.J.; Cantu, C.; Montgomery, R.K.; Christy, B.A.; Cap, A.P.; Bynum, J.A. A streamlined proliferation assay using mixed lymphocytes for evaluation of human mesenchymal stem cell immunomodulation activity. J. Immunol. Methods 2021, 488, 112915. [Google Scholar] [CrossRef] [PubMed]
  165. Labedz-Maslowska, A.; Szkaradek, A.; Mierzwinski, T.; Madeja, Z.; Zuba-Surma, E. Processing and Ex Vivo Expansion of Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells for the Development of an Advanced Therapy Medicinal Product for use in Humans. Cells 2021, 10, 1908. [Google Scholar] [CrossRef]
  166. Aghayan, H.R.; Salimian, F.; Abedini, A.; Ghazi, S.F.; Yunesian, M.; Alavi-Moghadam, S.; Makarem, J.; Majidzadeh-A, K.; Hatamkhani, A.; Moghri, M.; et al. Human placenta-derived mesenchymal stem cells transplantation in patients with acute respiratory distress syndrome (ARDS) caused by COVID-19 (phase I clinical trial): Safety profile assessment. Stem Cell Res. Ther. 2022, 13, 365. [Google Scholar] [CrossRef]
  167. Mebarki, M.; Iglicki, N.; Marigny, C.; Abadie, C.; Nicolet, C.; Churlaud, G.; Maheux, C.; Boucher, H.; Monsel, A.; Menasché, P.; et al. Development of a human umbilical cord-derived mesenchymal stromal cell-based advanced therapy medicinal product to treat immune and/or inflammatory diseases. Stem Cell Res. Ther. 2021, 12, 571. [Google Scholar] [CrossRef]
  168. Gimona, M.; Brizzi, M.F.; Choo, A.B.H.; Dominici, M.; Davidson, S.M.; Grillari, J.; Hermann, D.M.; Hill, A.F.; de Kleijn, D.; Lai, R.C.; et al. Critical considerations for the development of potency tests for therapeutic applications of mesenchymal stromal cell-derived small extracellular vesicles. Cytotherapy 2021, 23, 373–380. [Google Scholar] [CrossRef]
  169. Phinney, D.G. Functional heterogeneity of mesenchymal stem cells: Implications for cell therapy. J. Cell. Biochem. 2012, 113, 2806–2812. [Google Scholar] [CrossRef]
  170. Phinney, D.G.; Kopen, G.; Righter, W.; Webster, S.; Tremain, N.; Prockop, D.J. Donor variation in the growth properties and osteogenic potential of human marrow stromal cells. J. Cell. Biochem. 1999, 75, 424–436. [Google Scholar] [CrossRef]
  171. Radtke, S.; Görgens, A.; Liu, B.; Horn, P.A.; Giebel, B. Human mesenchymal and murine stromal cells support human lympho-myeloid progenitor expansion but not maintenance of multipotent haematopoietic stem and progenitor cells. Cell Cycle 2016, 15, 540–545. [Google Scholar] [CrossRef] [Green Version]
  172. Galipeau, J. The mesenchymal stromal cells dilemma—Does a negative phase III trial of random donor mesenchymal stromal cells in steroid-resistant graft-versus-host disease represent a death knell or a bump in the road? Cytotherapy 2013, 15, 2–8. [Google Scholar] [CrossRef]
  173. Kebriaei, P.; Hayes, J.; Daly, A.; Uberti, J.; Marks, D.I.; Soiffer, R.; Waller, E.K.; Burke, E.; Skerrett, D.; Shpall, E.; et al. A Phase 3 Randomized Study of Remestemcel-L versus Placebo Added to Second-Line Therapy in Patients with Steroid-Refractory Acute Graft-versus-Host Disease. Biol. Blood Marrow Transplant. 2020, 26, 835–844. [Google Scholar] [CrossRef]
  174. Baron, F.; Storb, R. Mesenchymal Stromal Cells: A New Tool against Graft-versus-Host Disease? Biol. Blood Marrow Transplant. 2012, 18, 822–840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Wang, C.; Börger, V.; Sardari, M.; Murke, F.; Skuljec, J.; Pul, R.; Hagemann, N.; Dzyubenko, E.; Dittrich, R.; Gregorius, J.; et al. Mesenchymal Stromal Cell–Derived Small Extracellular Vesicles Induce Ischemic Neuroprotection by Modulating Leukocytes and Specifically Neutrophils. Stroke 2020, 51, 1825–1834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Van Hoecke, L.; Van Cauwenberghe, C.; Börger, V.; Bruggeman, A.; Castelein, J.; Van Imschoot, G.; Van Wonterghem, E.; Dittrich, R.; Claeys, W.; Xie, J.; et al. Anti-Inflammatory Mesenchymal Stromal Cell-Derived Extracellular Vesicles Improve Pathology in Niemann–Pick Type C Disease. Biomedicines 2021, 9, 1864. [Google Scholar] [CrossRef] [PubMed]
  177. Madel, R.J.; Börger, V.; Dittrich, R.; Bremer, M.; Tertel, T.; Phuong, N.N.T.; Baba, H.A.; Kordelas, L.; Staubach, S.; Stein, F.; et al. Independent human mesenchymal stromal cell–derived extracellular vesicle preparations differentially attenuate symptoms in an advanced murine graft-versus-host disease model. Cytotherapy 2023, 25, 821–836. [Google Scholar] [CrossRef] [PubMed]
  178. Bauer, F.N.; Tertel, T.; Stambouli, O.; Wang, C.; Dittrich, R.; Staubach, S.; Börger, V.; Hermann, D.M.; Brandau, S.; Giebel, B. CD73 activity of mesenchymal stromal cell-derived extracellular vesicle preparations is detergent-resistant and does not correlate with immunomodulatory capabilities. Cytotherapy 2023, 25, 138–147. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kosanović, M.; Milutinović, B.; Kutzner, T.J.; Mouloud, Y.; Bozic, M. Clinical Prospect of Mesenchymal Stromal/Stem Cell-Derived Extracellular Vesicles in Kidney Disease: Challenges and the Way Forward. Pharmaceutics 2023, 15, 1911. https://doi.org/10.3390/pharmaceutics15071911

AMA Style

Kosanović M, Milutinović B, Kutzner TJ, Mouloud Y, Bozic M. Clinical Prospect of Mesenchymal Stromal/Stem Cell-Derived Extracellular Vesicles in Kidney Disease: Challenges and the Way Forward. Pharmaceutics. 2023; 15(7):1911. https://doi.org/10.3390/pharmaceutics15071911

Chicago/Turabian Style

Kosanović, Maja, Bojana Milutinović, Tanja J. Kutzner, Yanis Mouloud, and Milica Bozic. 2023. "Clinical Prospect of Mesenchymal Stromal/Stem Cell-Derived Extracellular Vesicles in Kidney Disease: Challenges and the Way Forward" Pharmaceutics 15, no. 7: 1911. https://doi.org/10.3390/pharmaceutics15071911

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