Next Article in Journal
Drought Resistance and Ginsenosides Biosynthesis in Response to Abscisic Acid in Panax ginseng C. A. Meyer
Previous Article in Journal
Identification of Proteins Involved in Cell Membrane Permeabilization by Nanosecond Electric Pulses (nsEP)
Previous Article in Special Issue
SOCS3 Protein Mediates the Therapeutic Efficacy of Mesenchymal Stem Cells against Acute Lung Injury
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

miRNA-Guided Regulation of Mesenchymal Stem Cells Derived from the Umbilical Cord: Paving the Way for Stem-Cell Based Regeneration and Therapy

by
Arsinoe C. Thomaidou
1,
Maria Goulielmaki
2,
Antonis Tsintarakis
3,
Panagiotis Zoumpourlis
3,
Marialena Toya
3,
Ioannis Christodoulou
3 and
Vassilis Zoumpourlis
3,*
1
Laboratory of Clinical Virology, Medical School, University of Crete, 71500 Heraklion, Greece
2
Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
3
Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(11), 9189; https://doi.org/10.3390/ijms24119189
Submission received: 3 May 2023 / Revised: 19 May 2023 / Accepted: 21 May 2023 / Published: 24 May 2023

Abstract

:
The human body is an abundant source of multipotent cells primed with unique properties that can be exploited in a multitude of applications and interventions. Mesenchymal stem cells (MSCs) represent a heterogenous population of undifferentiated cells programmed to self-renew and, depending on their origin, differentiate into distinct lineages. Alongside their proven ability to transmigrate toward inflammation sites, the secretion of various factors that participate in tissue regeneration and their immunoregulatory function render MSCs attractive candidates for use in the cytotherapy of a wide spectrum of diseases and conditions, as well as in different aspects of regenerative medicine. In particular, MSCs that can be found in fetal, perinatal, or neonatal tissues possess additional capabilities, including predominant proliferation potential, increased responsiveness to environmental stimuli, and hypoimmunogenicity. Since microRNA (miRNA)-guided gene regulation governs multiple cellular functions, miRNAs are increasingly being studied in the context of driving the differentiation process of MSCs. In the present review, we explore the mechanisms of miRNA-directed differentiation of MSCs, with a special focus on umbilical cord-derived mesenchymal stem cells (UCMSCs), and we identify the most relevant miRNAs and miRNA sets and signatures. Overall, we discuss the potent exploitations of miRNA-driven multi-lineage differentiation and regulation of UCMSCs in regenerative and therapeutic protocols against a range of diseases and/or injuries that will achieve a meaningful clinical impact through maximizing treatment success rates, while lacking severe adverse events.

1. Introduction

Mesenchymal stem cells (MSCs) are multipotent stromal cells of adult or fetal origin, characterized by the unique abilities of self-renewal, differentiation, and tissue regeneration [1,2], while they have also been shown to induce anti-inflammatory and immunosuppressive responses [3]. Although MSCs can be detected in almost all post-natal tissues, bone marrow (ΒΜ) and adipose tissue (AΤ) are rich and popular sources for MSC isolation [3]. ΒΜ- and AT-originated MSCs share a number of common characteristics, including specific cell surface marker expression, plastic adherence, and the capacity to differentiate into cells of mesenchymal lineage [4]. Apart from their phenotypic resemblance to MSCs of adult origin, fetal MSCs that can be found in fetal, perinatal, or neonatal tissues, such as cord blood (CB), umbilical cord (UC), and placenta [5,6], bear strong advantages [7]. Among these is the higher proliferation capacity and karyotypic stability in cell culture, greater ease of ex vivo genetic manipulation, increased survival during cryopreservation, as well as increased response to environmental stimuli and hypoimmunogenicity [8,9]. Both endogenous and exogenous MSCs have the ability to home to injured sites within the body, where they interact with the local microenvironment to repair the damaged tissue [10,11]. The unique properties of MSCs, especially those derived from the peri- and neonatal tissues, render them prime candidates for regenerative and therapeutic applications. To date, MSCs have been successfully applied to the treatment of the injured myocardium, skin, pancreas, and bone, as well as against several cancer types and other disorders [12,13].
MicroRNAs (miRNAs) are limited-sized RNA sequences with key regulatory properties that differ from protein coding [14]. Primary miRNAs are biosynthesized inside the nucleus via canonical or non-canonical pathways and then transported to the cytoplasm to be processed into precursor and finally mature elements [15]. These active miRNAs can either suppress or infrequently promote gene expression via binding on distinct sites of their target genes, either locally or at distant sites through microvesicle-mediated translocation [16,17]. In detail, the principal role of an activated miRNA is to detect its target; this is accomplished via the recognition of highly conserved complementary sequences in specific sites of the target mRNA. Most often, miRNAs engage to the 3′ untranslated region (3′ UTR) of their target mRNA, thereby inducing mRNA degradation and translational pause. In less common cases, miRNAs may interact with other loci, including the 3′ and 5′ UTRs, gene promoters, and other untranslated sequences to promote their functions [15,18,19]. More specifically, target gene silencing is performed with the complicity of the minimal miRNA-induced silencing complex (miRISC). The miRISC binds to the target mRNA to block its translation, possibly through eIF4F complex interference [20]. Interestingly, it has been proposed that miRNAs can also regulate transcriptional and post-transcriptional mRNA modification and nuclear degradation via a low molecular weight miRISC-guided mechanism that is yet to be elucidated [21]. Since miRNA signaling is an integral part of normal organismic development and maintenance, aberrant expression of specific miRNAs has been linked to certain abnormalities and diseases [22]. Recently, it was shown that miRNAs regulate multiple functions and properties of both embryonic and adult stem cells, including stemness preservation, self-renewal, and differentiation potential [23,24], while MSC-derived exosomes are being exploited as targeted microRNA delivery systems in different diseases [25].
The present review aims at unraveling the mechanisms that govern miRNA-directed differentiation of MSCs, with a special focus on umbilical cord-derived mesenchymal stem cells (UCMSCs). Specifically, UCMSCs’ differentiation potential toward bone, liver, cartilage, neurons, epithelia and other tissues and organs, in the context of miRNA upregulation or suppression, is discussed. Additionally, putative applications of human UCMSCs in conjunction with self-extracellular vesicles carrying specific miRNAs involved in critical cellular functions are described as appealing candidates in the context of stem-cell-based therapies.

2. miRNA-Guided Differentiation Potential of UCMSCs

Multipotent stem cells from multiple sources possess a multi-lineage differentiation potential that can be further exploited in several clinical settings, including tissue regeneration and organ rehabilitation for therapeutic and/or aesthetic applications, therapeutic interventions of distinct syndromes and disorders, as well as cancer treatment. When it comes to UCMSCs, this differentiation potential and accordingly the range of applications are significantly enhanced. A summarized report of the miRNAs that are known for driving distinct differentiation fates of UCMSCs is depicted in Figure 1, while the up-to-date recorded research on the role of different miRNAs in the differentiation process of perinatal MSCs is displayed in detail in Table 1.

2.1. Osteogenic/Osteoblastic Differentiation

miR-21 appears to play an important role in the osteogenic differentiation of UCMSCs, since human UCMSCs transfected with miR-21 have demonstrated increased expression levels of several known osteogenic genes, such as alkaline phosphatase (ALP), runt-related transcription factor-2 (RUNX-2), and osteocalcin (OCN), an effect that is possibly achieved through the targeting of the PI3K/β-catenin (PI3K-AKT-GSK3β) pathway by miR-21 [31]. Accordingly, genetic or chemical depletion of miR-21 in human UCMSCs (hUCMSCs) resulted in the suppression of their osteogenic properties, via targeting the Wnt/β-catenin pathway [30].
miR-342-3p levels were also found to be significantly upregulated during osteogenesis [28]. Their overexpression in vitro was shown to promote the osteogenic differentiation of hUCMSCs through the elevated expression of various markers, such as OCN and ALP [28], as well as the possible regulation of the transforming growth factor beta (TGF-β) [50] and the sonic hedgehog (Shh) signaling pathways [28], which are generally found to be overactivated during the osteogenic process. However, the findings regarding the role of the Shh signaling in the osteogenic differentiation of mesenchymal stem cells (MSCs) appear to be rather controversial in the literature. A different study demonstrated that the Shh signaling pathway can also act as a suppressor of the osteogenic differentiation of human umbilical cord blood-derived mesenchymal stem cells (hUCBMSCs), possibly via negative regulation of the miR-148 family members, especially miR-148b, which is one of its downstream targets [51]. This could very well explain the pathway’s anti-osteogenic properties in hUCBMSCs, since the expression levels of miR-148b, along with several other microRNAs, including let-7i, miR-23b, miR-141, miR-148a, and miR-152, were found to be significantly upregulated during osteogenic differentiation [51].
miR-196a-5p has been reported to favor the in vitro osteogenic differentiation capacity of Wharton’s jelly-derived mesenchymal stem cells (WJMSCs) by enhancing the activity of several osteogenic markers such as ALP, OCN, Dentin Matrix Protein 1 (DMP1), Bone Sialoprotein (BSP) and Dentin Sialophosphoprotein (DSPP), while also suppressing their proliferative potential. Furthermore, miR-196a-5p overexpression in a calvarial bone defect (segmental defect, SD) rat model was shown to promote defect closure and bone regeneration within 12 weeks after sheet transplantation [29]. These findings appear to be also in agreement with another study that reported that overexpression of miR-196b-5p, a micro-RNA belonging in the same family as miR-196a, results in the inhibition of cell cycle progression and proliferation of WJMSCs [52], potentially favoring a differentiating fate over the preservation of stemness.
It was reported that miR-210 overexpression could potentially lead hUCBMSCs to differentiate into osteoblasts via the upregulation of ALP, OCN, and RUNX-2 during various steps of the differentiation process [53]. miR-216a was found capable of reversing the inhibiting effects of dexamethasone on the osteogenic process, while its expression levels were shown to positively correlate with osteoblastic differentiation and bone formation in adipose tissue-derived mesenchymal stem cells (ATMSCs) and UCMSCs [35]. Furthermore, it was demonstrated that an ex vivo engineered vector combining miR-424 and the bone morphogenetic protein-2 (BMP2) encoding gene has the capacity to promote osteogenesis in WJMSCs, with miR-424 potentially leading to the overexpression of OCN and BMP2 protein levels, the latter of which is considered an important regulator of the osteogenic/osteoblastic differentiation, and ultimately bone development [26].
On the other hand, other microRNAs appear to promote self-renewal and an undifferentiated state over a specific fate determination path. In contrast to miR-424, miR-140-5p has shown the exact opposite effect on BMP2 by suppressing its expression. miR-140-5p and BMP2 endogenous levels are inversely related at distinct time points during the osteogenic process of undifferentiated MSCs derived from multiple sources including the adipose tissue, the bone marrow, and the umbilical cord, while miR-140-5p inhibition seems to allow a significant increase in the expression levels of the osteogenic markers RUNX-2, ALP, OCN, and OPN [27]. Similarly, miR-132 was demonstrated to function as a negative regulator of the osteogenic differentiation of hUCMSCs, via inhibition of the Wnt/β-catenin pathway and suppression of the expression of Osterix, a transcriptional factor required for the osteoblastic differentiation and development [34]. miR-25-3p and miR-33b-5p were also found to inhibit the expression of SMAD5 and Wnt10b, respectively, both of which are important factors for the signaling and promotion of the osteogenic differentiation of mesenchymal stem cells. However, suppression of those two miRNAs by a novel lncRNA, linc02349, was found capable of effectively reversing their effects and allowing the expression of SMAD5 and Wnt10b, thus facilitating the osteogenic differentiation of hUCMSCs [33].
A number of hUCMSC exosome-secreted microRNAs, including miR-429, miR-34b-3p, miR-370-5p, miR-1270, miR-4454, miR-619-5p, miR-150-5p, miR-365b-3p, miR-365a-3p, miR-10a-5p, miR-615-3p, miR-328-3p, let-7d-3p, miR-675-5p, miR-10b-5p, miR-574-3p, miR-204-5p, miR-433-3p, miR-2110, miR-382-5p, miR-25-3p, miR-345-5p, miR-146a-5p, miR-629-5p, miR-590-3p, miR-21-5p, miR-377-5p, miR-1246, miR-188-5p, miR-329-3p, miR-3074-5p, miR-136-3p, miR-27a-3p, miR-598-3p, miR-30d-5p, miR-299-3p, miR-337-5p, miR-549a-5p, miR-655-3p, miR-410-5p, and miR-4423-5p, were also identified as critical modulators of the osteoblastic proliferation and differentiation processes, with miR-2110 and miR-328-3p highlighted as the most probable of regulating bone differentiation, based on their specific target genes [32]. These 41 miRNAs were also reported capable of reversing ovariectomy-induced osteoporosis to a certain degree in an in vivo mouse model, suggesting that exosome-derived microRNAs might also present great potential for therapeutic application in the clinical setting.

2.2. Hepatic Differentiation

The second most-studied field in the umbilical cord mesenchymal stem cell fate determination involving the mediation of microRNAs appears to be the path toward hepatic differentiation. A next-generation sequencing analysis identified a total of 63 miRNAs, both novel and known, that presented significant changes in their expression patterns during different time points of hepatic differentiation of WJMSCs, while a bioinformatics analysis of their target genes shed some light on the molecular pathways and liver-specific transcription factors involved in the hepatic differentiation process [54]. Another miRNA-level analysis of human umbilical cord Wharton’s jelly revealed various significant microRNAs during hepatic differentiation, including the miR-23b cluster (miR-27b-3p, miR-24-1-5p, and miR-23b-3p), miR-26a-5p, miR-30a-5p, miR-122-5p, miR-148a-3p, and miR-192-5p), all of which are suggested to promote stem cell induction into hepatocytes through the negative regulation of targets related to the inhibition of hepatic differentiation, with miR-122-5p specifically suppressing the mesenchymal markers Sex Determining Region-Box 11 (SOX11) and Vimentin (VIM) [36]. A number of 61 unique microRNA profiles were reported as differentially expressed consistently during the hepatic differentiation process, thus promoting or suppressing the differentiation of umbilical cord lining-derived mesenchymal stem cells, accordingly [55]. Seven of those unique miRNAs, including miR-1246, miR-1290, miR-148a, miR-30a, miR-424, miR-542-5p, and miR-122, many of which are normally found upregulated during hepatic differentiation, have been reported capable of stimulating the conversion of UCMSCs into fully functional hepatocytes when concomitantly overexpressed in vitro. Both hepatocytes and MSCs expressing those specific seven microRNAs possessed the ability to repair liver injury and to improve liver function in a CCL4-injured mouse model after two weeks of transplantation [56]. In addition, a more recent study concluded that an optimized combination of only five of those microRNAs, specifically, miR-122, miR-148a, miR-424, miR-542-5p, and miR-1246, could produce the same results in both an in vitro and an in vivo environment, without the need for the presence of miR-30a and miR-1290 [57]. Similarly, the concurrent overexpression of another three-miRNA set, including miR-106a, miR-574-3p, and miR-451, was reported capable of promoting the differentiation of hUCMSCs into mature and functional hepatocytes in vitro [37], suggesting that specific combinations of miRNAs could provide an alternative procedure through which mature hepatocyte-like cells are generated for therapeutic purposes in the future.

2.3. Neural Differentiation

A number of miRNAs are also suggested to play an integral part in the neural differentiation of UCMSCs. Several studies demonstrated that there exist statistically important differences in the expression levels of certain microRNAs between WJMSCs trans-differentiated into neuronal cells and undifferentiated WJMSCs. More specifically, as part of a microarray analysis, a total of 161 differentially expressed miRNAs were detected, 28 of which demonstrated a high fold change of more than 5, with miR-4521, miR-222-5p, miR-92a-1-5p, miR-543, and miR-548a-3p presenting significantly downregulated levels and miR-575, miR-4440, miR-297, miR-4793-3p, miR-371b-5p, miR-3617-5p, miR-125b-2-3p, miR-26b, miR-124a, miR-7152-3p, miR-1290, miR-5093, miR-663b, miR-6861-5p, miR-212-5p, miR-194, miR-129-5p, miR-132-5p, miR-1202, miR-3687, miR-195-3p, miR-192-5p, and miR-5572 showing significant upregulation in WJMSC-derived neurons compared to the undifferentiated controls [58]. Similarly, miR-345, miR-106a, miR-17-5p, miR-20a, and miR-20b were characterized as upregulated in undifferentiated WJMSCs, whereas miR-206, miR-34a, miR-374, miR-424, miR-100, miR-101, miR-323, miR-368, miR-137, miR-138, and miR-377 were found to be significantly upregulated in trans-differentiated WJMSCs. Moreover, miR-20a, miR-20b, miR-17-5p, and miR-106a, which all belong to the miR-17 family, were found to be downregulated during the neurogenesis of WJMSCs. Among these, miR-34a overexpression was shown to significantly limit stem cell motility, indicating that the suppression of endogenous miR-34a could possibly be utilized as a method of enhancing transplanted stem cell motility toward injury sites in stem cell-based therapies [59]. Similarly, miR-203 was found to suppress neural retina differentiation through targeting three retina development-related genes, namely, DKK1, CRX, and NRL. However, anti-miR-203 transfection of hUCBMSCs was shown to successfully reverse this inhibiting effect and allow the expression of those target genes, thus converting cells into photoreceptor cell types [39]. The combination of two more microRNAs, miR-20b and miR-106a, might also have an important role to play in the negative regulation of neural differentiation of hUCMSCs, possibly via the suppression of Neurogenin-2 (Ngn2), which is known for controlling the cell cycle progression and promoting neural differentiation [38]. These findings further indicate that stem cells treated with either specific miRNAs or corresponding miRNA inhibitors might provide a reliable source of desired cell types or even subtypes for stem cell-based therapies of non-regenerative diseases.
Another large-scale analysis determined that the expression levels of various miRNAs related to motor neuronal cell differentiation and proliferation are altered in relation to different stages of hUCBMSC differentiation. miR-9-5p and miR-324-5p (related to differentiation) demonstrated significant upregulation during early time points of neuronal differentiation, while miR-137 and miR-let-7b (related to proliferation) showed significant downregulation during the latter stages of differentiation. Different expression profiles were also detected between hUCBMSCs treated with neurogenic induction agents (retinoic acid and Shh in combination), with miR-449c-5p, miR-1249-3p, miR-9-5p, and miR-324 showing elevated expression levels and miR-335-3p and miR-335-5p demonstrating decreased expression in treated cells compared to the untreated controls. Furthermore, this comparison of miRNA profiles between undifferentiated and treated/induced hUCBMSCs revealed a number of new differentially expressed miRNAs, including novel-miR-17, novel-miR-18, and novel-miR-20, which appeared to be significantly upregulated, and novel-miR-1 and novel-miR-2, which showed significant downregulation [60]. It should be noted here that a number of those previously unknown miRNAs were found to target genes in six critical pathways involved in the neuron differentiation process, including the cholinergic synapse, axon guidance, hedgehog, MAPK, TGF-β, and JAK-STAT signaling pathways, although their exact mechanistic functions remain a subject of future investigation [60].

2.4. Chondrogenic Differentiation

In regard to chondrogenic differentiation, miR-29b-3p expression was shown to decrease during the chondrogenic process, through targeting of SRY-related high-mobility-group box 9 (SOX9), a crucial transcription factor for chondrocyte differentiation, while, similar to other long non-coding RNAs, lncRNA H19 was observed to function as a sponge of miR-29b-3p, essentially inhibiting its effect and allowing for the expression of SOX9 and the subsequent platelet lysate (PL)-induced chondrogenic differentiation of hUCMSCs [40]. Furthermore, miR-340-5p, miR-130a-3p, miR-381-3p, miR576-5p, and miR-337-3p were found to be highly upregulated in small extracellular vesicles (sEVs) obtained from hUCMSCs treated with Kartogenin (KGN) compared to untreated control cells, whereas miR-200b-3p, miR-200c-3p, miR-375, miR-122-5p, and miR-182-5p appeared to be downregulated in treated compared with untreated stem cells. Among these, miR-381-3p overexpression was shown to be capable of promoting chondrogenic differentiation of MSCs and upregulation of chondrogenesis-related genes, such as SOX9, aggrecan, and collagen II, while directly binding and inhibiting the expression of TAOK1, an upstream regulator of the Hippo signaling pathway [41]. It should be noted that transplanted KGN-sEVs, along with hUCMSCs, were also observed to restore tissue injury in a knee articular cartilage defect rabbit model four weeks after the operation [41], suggesting that sEVs might also have a role to play in the in vivo induction of chondrogenesis as a future therapeutic approach.

2.5. Epithelial Differentiation

In terms of MSC epithelial differentiation, miR-145 was shown to promote the conversion of UCMSCs into type II alveolar epithelial cells under hypoxic conditions, possibly via targeting the TGF-β receptor II (TGFβRII), which in turn results in the suppression of differentiation into fibroblasts by the TGF-β signaling pathway [43]. Since the TGF-β pathway is highly upregulated during fibrosis after acute lung injury (ALI), hypoxia-induced miR-145 could present a new method of stem cell-based therapies for lung injury [43]. A number of microRNAs, including miR-100, miR-127-3p, miR-136, miR-146a, miR-199a-5p, miR-214, miR-224, miR-299-5p, miR-337-5p, miR-34a, miR-376a, miR-376c, miR-377, miR-379, miR-381, miR-409-3p, miR-410, miR-424, miR-654-3p, miR-758, and miR-762, which are normally depleted in the retinal pigment epithelium (RPE) and in the ARPE-19 cell line, were found to be overexpressed in undifferentiated hUCBMSCs [42]. Among these, miR-410 was demonstrated to target more than one RPE-related gene, specifically, OTX2 and RPE65, while its inhibition could induce RPE differentiation in human amniotic epithelial stem cells [61]. In agreement with these results, it was later demonstrated that multiple transfections with anti-miR-410 in UCBMSCS could indeed induce upregulation of RPE-specific factors, such as Melanocyte Inducing Transcription Factor (MITF), LRAT, RPE65, Bestrophin, and EMMPRIN, and therefore result in direct RPE differentiation [42].

2.6. Differentiation toward Other Lineages

In regard to differentiation into insulin-producing cells (IPCs), miR-200b-3p demonstrated the capability of directing UCMSCs toward this fate via the inhibition of the ZEB2 transcription factor, while its suppression was shown to induce hypoglycemia and to inhibit insulinogenesis in successfully differentiated IPCs from UCMSCs in an in vivo diabetic mouse model [44]. In addition, miR-375 and miR-26a were shown to be highly enriched in undifferentiated chicken nestin-positive UCMSCs (N-UCMSCs) compared with nestin-positive pancreatic mesenchymal stem cells (N-PMSCs). Combined miR-375 and miR-26a transfection was found capable of suppressing several target genes, including mtpn, sox6, bhlhe22, and ccnd, thus inducing IPC differentiation 12 days after treatment. Furthermore, the two microRNAs’ function was further validated in vivo by transplanting successfully differentiated N-UCMSCs in a hyperglycemic mouse model and confirming that IPCs from N-UCMSCs do indeed secrete chicken insulin into the host animals’ blood within two weeks after they are injected with glucose, suggesting that cell-based transplantation therapies for diabetes might become a promising strategy in the future [45].
In regard to myoblastic differentiation, a group of four microRNAs, specifically, miR-21, miR-23a, miR-125b, and miR-145, were shown to be highly upregulated in exosomes derived from UCMSCs. High-throughput RNA analysis revealed that these specific miRNAs have an essential role in the inhibition of myofibroblast and scar formation in both an in vivo skin wound mouse model and in vitro, by suppressing genes in the TGF-β/SMAD2 pathway, such as TGF-β2, TGF-βR2, and SMAD2, as well as α-smooth muscle actin (α-SMA) and collagen I expression, suggesting that UCMSC-derived exosomes could pose an alternative to cell-based therapies to prevent tissue fibrosis and wound scarring in the clinical setting [48]. Additionally, miR-503-5p and miR-222-5p were both identified as contrasting regulators of smooth muscle cell differentiation from hUCMSCs, with miR-503 promoting the differentiation process by directly targeting SMAD7 and miR-222-5p inhibiting the same process through dual targeting of ROCK2 and αSMA [49].
In regard to hematopoietic differentiation, miR-218, miR-150, and miR-451 were all found to be upregulated during the hematopoietic induction of UCMSCs via combined Aza/GF treatment. Among these microRNAs, miR-218 was revealed to negatively regulate the transcription of MITF, while its overexpression might possibly result in the promotion of hematopoietic differentiation via the upregulation of NF-Ya and HoxB4, both of which are important transcriptional inducers of hematopoiesis [46].
In regard to adipogenic differentiation, it was demonstrated that although UCMSCs do possess the capacity to differentiate into adipocytes, their potential does not seem to be as strong compared to that of MSCs derived from different tissue origin, such as adipose tissue or bone marrow, which is an important parameter to consider when it comes to selecting specific stem cell sources for therapeutic purposes [47]. Nevertheless, the miR-301b-miR-130b cluster, which is theorized to inhibit PPARγ, a critical adipogenetic transcription factor, was proposed as a negative regulator of adipogenic induction in MSCs derived from all three sources, with the two miRNAs’ endogenous expression levels directly correlating with each MSC type’s adipogenic potential [47].
Although in most cases the specific molecular mechanisms through which different miRNAs control and regulate the determination of cell fate during the differentiation of hUCMSCs still remain poorly understood, taken together, the above findings do indicate that the overexpression or inhibition of unique or multiple miRNAs could eventually serve as a promising method of efficiently generating functional cells of desired lineages in the clinical setting, especially when it comes to matters of regenerative medicine, as will be discussed further in the following section of this study.

3. Applications of UCMSCs in Regenerative and Therapeutic Medicine and the Role of miRNAs

In recent years, especially in the last decades, the potential therapeutic applications of hUCMSCs have been intensively explored for the treatment of various pathologies, as can be deduced from the literature. Human UCMSCs, as well as their secreted exosomes and other extracellular vesicles, which often carry specific miRNAs involved in key cellular functions, appear to possess significant regenerative, anti-inflammatory, protective, and in some cases, even tumor-suppressing capabilities [62]. Their ameliorative properties, along with their strong self-renewal rate, low immunogenic potential, and non-invasive isolation does indeed make UC-derived MSCs one of the most appealing candidate sources for novel stem-cell-based therapies as well as other applications, including general and tissue-specific toxicity screening [63]. Promising applications of UCMSCs in the context of miRNA-directed regenerative/healing and therapeutic potential are presented in Table 2 and Table 3 and in the corresponding Figure 2 and Figure 3, respectively.

3.1. Ischemic/Reperfusion (I/R) Injuries

Ischemic-reperfusion injury (IRI) is a critical condition that can affect various organ systems and for which there is currently no effective therapeutic strategy, but mostly supportive treatment. IRI is currently a major cause for transplantation complications and graft loss; however, MSC-based therapies appear to present a promising new method, since a number of different microRNAs, specifically delivered through UCMSC-exosomes, have been reported to play important functional and regulatory roles in the complex biological processes associated with IRI. UCMSC microvesicle-transferred miR-21, one of the most universally studied microRNAs and known for its involvement in the processes of angiogenesis, apoptosis, and inflammation, was found to be capable of ameliorating renal IRI both in vitro and in an in vivo rat model, possibly by regulating programmed cell death protein 4 (PDCD4) and inhibiting tubular epithelial cell apoptosis under hypoxic conditions [67]. Similarly, miR-1246, delivered through UCBMSC-exosomes, appears to pose a novel therapeutic option for hepatic IRI, since its presence was shown to shield hepatocytes against the apoptotic effects of hypoxia/reoxygenation (H/R) damage via decreasing the expression of the pro-inflammatory factors TNF-α, IL-6, IL-1β, and IL-17 [126,127] and regulating the GSK3β/Wnt/β-catenin signaling pathway [126]. miR-1246 was also shown to be capable of restoring the Th17/Treg cell imbalance caused by IRI in the liver, through the IL-6-gp130-STAT3 signaling pathway [127], while I/R rat treatment with miR-20a-containing exosomes appears to mediate the autophagic and apoptotic activity of hepatocytes by inhibiting the expression of Beclin-I, fatty acid synthase (FAS), and active Caspase-3 [66].
miR-24 is another microRNA found to be involved in the protective role that UCMSC-extracellular vesicles (EVs) appear to play in cerebral IRI. miR-24, carried by UCMSC-EVs, was reported to protect the brain tissue from I/R injury and ameliorate the cerebral damage both in cultured cells and in an in vivo rat model via inhibiting the expression of AQP4 and activating the P38 MAPK/ERK1/2/PI3K/AKT pathway [79]. miR-26b-5p was shown to play an important role in the prevention of nerve damage caused by cerebral I/R, since its delivery via UCMSC-exosomes represses microglia M1 polarization and neuroinflammation by negatively regulating inflammatory factor CH25H and inhibiting the activity of the toll-like receptor (TLR) pathway [81]. Additionally, exosome-derived miR-26b-5p was reported to have a protective effect over PC12 neurons in the brain tissue by targeting the neuro-inflammatory factor MAT2A and inhibiting the MAPK/STAT3 pathway, thus allowing for the possible recovery of early brain injury damage [82]. miR-410, delivered via UCMSC-EVs to the neurons of hypoxia-ischemia brain damage (HIBD) neonatal mice, improves the viability of neuronal cells while reducing the apoptotic level of damaged cells by downregulating HDAC1, thus enabling the expression of neuroprotective factors EGR2 and Bcl2 [117]. Furthermore, modified UCMSC-exosomes that underwent knockdown of miR-206, a microRNA that directly targets the neuroprotective BDNF gene, presented significant ameliorating effects in the improvement of impaired brain function and edema, as well as the prevention of neuronal apoptosis [112]. In the case of H/R-induced cardiac injury, UCMSC-exosomes carrying lncRNA UCA1 were suggested to shield cardiac microvascular endothelial cells from I/R damage, with lncRNA UCA1 functioning as a negative regulator of miR-143, which naturally aggravates the effects of oxidative stress in cardiomyocytes [103]. In addition, miR-21, was shown to enhance the production of new blood vessels in critical limb ischemia (CLI) by targeting the carboxyl terminus of Hsc70-interacting protein (CHIP) and increasing the angiogenetic activity of HIF-1a [68].
Transplantation of UCMSC-exosomes, specifically enhanced with silk fibroin hydrogel and carrying miR-675, was also proposed as a novel strategy for the treatment of vascular disease, with miR-675 significantly inhibiting the aging-related vascular dysfunction and promoting blood perfusion in an ischemic hindlimb mouse model [124]. Similar results were reported in the case of skeletal muscle ischemic injury, since combined treatment with WJMSCs and miR-29a has shown a particularly efficient therapeutic potential by greatly enhancing the angiogenetic potential of vein endothelial cells and reversing the impaired blood perfusion in the hindlimb of BPVC-injured mice [85]. Treatment of co-cultured UCMSCs and endothelial colony-forming cells (ECFCs) with hyaluronic acid was shown to cause significant downregulation of miR-139-5p through CD44 activation in order to facilitate the blood flow in the tissues of ischemic hindlimb rats as well as to enhance their angiogenetic properties [100]. The therapeutic properties of UCMSC-exosomes in injured hindlimb mice were also attributed to miR-24, which targets the pro-apoptotic factor Bim, thus enhancing muscle motility and blood flow [80], while recently it was suggested that the exosomal circular RNA circHIPK3 could act as a sponge for miR-421, essentially allowing for the expression of its target, FOXO3a, and the inhibition of pyroptosis and inflammation observed in ischemic mice [118].

3.2. Acute Organ Injuries

Acute kidney injury (AKI) is yet another severe clinical syndrome for which there appear to be a rising number of proposed cell-based studies involving UCMSCs. UCMSCs were found to enhance the autophagic potential of HK-2 cells in vitro through the secretion of miR-145, which in turn negatively regulates the PI3K/AKT/mTOR pathway [104]. Treatment with UCMSCs demonstrated a significant increase in the survivability of mice with sepsis-associated AKI through the upregulation of miR-146b, which subsequently blocks NF-κB pro-inflammatory activity via the suppression of its activator protein, IRAK1 [107]. Another recent study demonstrated that not only do UCMSC-exosomes specifically prefer to home to the proximal tubules of injured kidneys, but they also effectively mediate the typical effects of I/R in an AKI mouse model. Via miR-125b-5p delivery, injected exosomes appear to suppress the activation of p53 and protect damaged tubular epithelial cells from G2/M cycle arrest and apoptosis, while further promoting their proliferation and repair [96].
In addition to AKI, UCMSCs possess similar beneficial and regenerative effects over a number of various models of ischemic injury, including acute liver injury (ALI), acute myocardial infarction/ischemia (AMI), and acute spinal cord injury (SCI). UCMSC-exosomes, enriched with miR-455-3p, inhibited the activation of monocytes/macrophages in the liver by targeting PIK3r1 and downregulated the expression of key inflammatory cytokines, such as IL-6, in an in vivo mouse model, thus improving liver response and repair [120]. Exosome-derived miR-451 was reported to alleviate inflammation in burn-induced ALI rats, by significantly downregulating the expression levels of various known inflammatory cytokines, including TNF-α, IL-1β, and IL-6, via the suppression of the TLR4/NF-κB pathway [130], as well as the regulation of the MIF/PI3K/Akt signaling pathway to induce M1 to M2 macrophage polarization [119]. UCBMSC-derived exosomal miR-22-3p also demonstrated therapeutic properties in the prevention of lipopolysaccharide (LPS)-induced ALI. Its delivery reduces inflammatory and oxidative stress responses both in vitro in lung cells and in vivo in rat lung tissues via silencing of inflammatory factors TNF-α, IL-1β, IL-6, and the frizzled class receptor 6 (FZD6) gene [75]. Exosomal miR-377-3p was demonstrated to play a role in the mediation of inflammation in LPS-induced ALI mice by inducing cell autophagy through downregulation of its target gene RPTOR [120], while miR-100 delivery through WJMSC-microvesicles (MVs) appears to function similarly by negatively regulating mTOR expression and increasing autophagy levels in BLM-induced ALI rats [91]. Exosomal miR-100-5p presents significant protective properties over cardiomyocytes that have undergone H/R damage by suppressing the transcription factors FOXO3 and NLRP and consequently shielding cells from the effects of inflammasome activation, cytokine release, and pyroptosis [92].
In like manner, miR-19a in UCMSC-exosomes mitigates the effects of hypoxic damage via the suppression of SOX6 and the regulation of the mitochondrial apoptotic pathway AKT/JNK3/caspase-3 [65]. EV-delivered miR-223 provides yet another potential therapeutic tool for AMI as it both represses the inflammatory response in cardiomyocytes and induces the angiogenic properties of vein endothelial cells, protecting rats from myocardial fibrosis while concurrently promoting myocardial healing via the modulation of the P53/S100A9 axis [113]. A somewhat different approach was provided in the case of miR-125b-5p, as it was suggested that its upregulation might contribute to the effects of myocardium infraction after hypoxic injury. However, UCMSC-exosomes were shown to negate its harmful properties by upregulating Smad7 to suppress its expression levels and facilitate cardiomyocyte repair [95]. Interestingly, migration inhibitory factor (MIF) engineered UCMSC-exosomes demonstrated even more efficient cardioprotective properties in rats in comparison to non-modified UCMSC-exosomes, possibly through the significant upregulation of miR-133a-3p, which in turn activates the AKT signaling pathway and increases VEGF expression to promote ischemic damage repair [98]. UCBMSC-exosomes loaded with miR-23a-3p were shown to significantly decrease H/R-induced myocardial cell ferroptosis in mice via the targeting of the DMT1 gene [77], while the delivery of exosomal miR-136 could potentially revive aged and senescent BMMSCs to improve their cardiac repair functions by inhibiting Apaf1 expression in MI mice [99].
With regards to SCI, miR-29b-3p delivery through UCMSC-EVs was shown to improve the motility and nerve function repair of model rats by downregulating the expression of PTEN and activating the Akt/mTOR pathway [87], while exosomal miR-29b-3p also prevents the apoptotic effects of LPS-induced SCI in damaged neurons via inhibition of PTEN and induction of the PI3K/AKT pathway [88]. Exosomal miR-199a-3p and miR-145-5p function synergistically to promote neuronal cell differentiation and improve the impaired hindlimb motility of SCI rats while decreasing inflammation severity at the spinal cord damage site by suppressing the expression of the Cblb and Cbl genes and subsequently regulating the NGF/TrkA pathway [131]. Similar results were reported following the treatment of SCI rats with miR-126-loaded UCMSC-exosomes, as this particular microRNA appears to reduce apoptosis and enhance both angiogenesis and neurogenesis in the injured spinal cord by potentially inhibiting the expression of its two target genes, SPRED1 and PIK3R2 [97]. Lastly, it was reported that treatment with miR-146a-5p-transfected exosomes leads to the reduction of astrocyte neurotoxicity in the spinal cord through targeting the Traf 6/Irak1/NFκB pathway [106].
Table 3. miRNA-guided therapeutic applications of perinatal and neonatal MSCs.
Table 3. miRNA-guided therapeutic applications of perinatal and neonatal MSCs.
miRNATissue OriginVehicle TypeTarget (Gene/Pathway)FunctionClinical ApplicationReference
miR-17-3pUCMSCsExosomesSTAT1Inflammation/Apoptosis Suppression, Oxidative Injury ReductionDiabetic Retinopathy[132]
miR-17-5pUCMSCsExosomesSIRT7ROS Reduction, Proliferation PromotionPremature Ovarian Insufficiency[133]
miR-18bUCMSCsEVsMAP3K1/NF-κB/p65Apoptosis/Inflammation InhibitionDiabetic Retinopathy[134]
UCMSCsEVsNotch2/TIM3/mTORC1Proliferation/Migration Promotion, Blood Pressure ReductionPre-Eclampsia[135]
miR-21UCMSCsEVsTGF-β2Myoblast Differentiation InhibitionLung Fibrosis[136]
UCMSCsExosomesLATS1Estrogen Secretion PromotionPremature Ovarian Insufficiency[137]
UCMSCsExosomesp38 MAPKApoptosis/ER Stress SuppressionDiabetes[138]
miR-23UCMSCsEVsTGF-βR2Myoblast Differentiation InhibitionLung Fibrosis[136]
miR-24-3pUCMSCsExosomesKeap-1Lipid Accumulation/ROS Generation/Inflammation InhibitionNon-Alcoholic Fatty Liver Disease[139]
miR-26a-5pUCMSCsExosomesMETTL14/NLRP3Cell Survival Promotion, Pyroptosis InhibitionIntervertebral Disc Degeneration[140]
miR-27bUCMSCsExosomesHOXC6EMT SuppressionSubretinal Fibrosis[141]
miR-29aUCMSCsEVsHBP1/Wnt/β-cateninProliferation Promotion, Apoptosis Inhibition, Ovarian Function RestorationPremature Ovarian Insufficiency[142]
Placenta-derived MSCsExosomes-Differentiation Promotion, Utrophin Increase, Fibrosis/Inflammation InhibitionDuchenne Muscular Dystrophy[143]
miR-30c-5pUCMSCsEVsPLCG1/PKC/NF-κBInflammation SuppressionDiabetic Retinopathy[144]
miR-100UCMSCsEVsHS3ST2Proliferation/Invasion/Migration/EMT PromotionEndometriosis[145]
miR-100-5pUCMSCs-NOX4/NLRP3, GSDMDInflammation/Oxidative Stress/Apoptosis InhibitionPremature Ovarian Insufficiency[146]
UCMSCsEVs-M2 Polarization Promotion, Treg GenerationSS Dry Eye[147]
UCMSCsEVsNOX4ROS/Oxidative Stress/Apoptosis InhibitionHeart Failure[148]
UCMSCsExosomesFZD5/Wnt/β-cateninMigration Inhibition, Apoptosis Promotion, Inflammation SuppressionAtherosclerosis[149]
miR-101UCMSCsEVsBRD4/NF-κB/CXCL11Proliferation/Migration PromotionPre-Eclampsia[150]
miR-125bUCMSCsEVsIL-6R, IFV genesViral Activities/Infection InhibitionRespiratory Virus-associated Diseases[151]
miR-126UCMSCsExosomesHMGB1Inflammation SuppressionRetinal Inflammation[152]
miR-126-3pUCMSCsExosomesPIK3R2, PI3K/AKT/mTORProliferation/Angiogenesis Promotion, Apoptosis SuppressionPremature Ovarian Insufficiency[153]
miR-133UCMSCsExosomes-Proliferation/Survival Promotion, Bregs ProductionImmune Thrombocytopenia[154]
miR-133bUCMSCsExosomesSGK1Proliferation/Cell Cycle Progression/Migration/Invasion Promotion, Apoptosis InhibitionPre-Eclampsia[155]
miR-140-5pUCMSCsExosomesFSTL3Cell Growth/Angiogenesis Promotion, Inflammation SuppressionPre-Eclampsia[156]
miR-146aUCBMSCs--Inflammation SuppressionInflammatory Diseases[157]
UCMSCsExosomesSUMO1/β-cateninColitis Deterioration/CAC Progression InhibitionColitis[158]
UCMSCsExosomesTRAF6, IRAK1, NF-κBFibroblast Activation/Inflammation/Fibrosis InhibitionUrethral Stricture Diseases[159]
WJMSCsExosomes-M2 Macrophage Polarization Promotion, Inflammation InhibitionInflammatory Disorders/Sepsis[160]
miR-146a-5pUCMSCsExosomesTRAF6Neuroinflammation/Pyroptosis Suppression, Autophagy PromotionInflammatory Pain[161]
UCMSCsExosomesNOTCH1Bleeding/Inflammation/M1 Polarization Suppression, M2 Polarization PromotionSLE-associated DAH[162]
UCMSCs-TRAF6/STAT1M2 Polarization Promotion, Renal Function Improvement, Inflammation SuppressionDiabetic Nephropathy[163]
UCMSCsEVs(TGF-β1/Smad2/3)Allergic Inflammation/Fibrosis/Airway Remodeling SuppressionAsthma[164]
miR-147WJMSCsEVs-Inflammation Suppression, Macrophage ActivationAbdominal Aortic Aneurysm[165]
miR-148a-5pUCMSCs-Notch2Proliferation Promotion, Apoptosis/Fibrosis InhibitionLiver Fibrosis[166]
miR-153-3pUCMSCs-Snai1EMT SuppressionPeritoneal Fibrosis[167]
UCMSCs-PELI1Proliferation/Migration Inhibition, Tfh/Treg Imbalance PromotionSLE[168]
miR-181aUCMSCs--T Lymphocyte RegulationSLE[169]
miR-195UCMSCsExosomesTFPI2Hypoxic Damage ReductionPre-Eclampsia[170]
miR-199UCMSCs-KGF(Fibrosis Promotion)Cirrhosis[171]
miR-199a-5pUCMSCs-Sirt1/p53CD4+ T-cell Senescence PromotionSLE[172]
miR-203a-3p.2UCMSCsExosomescasp11/4Macrophage Pyroptosis/Inflammation InhibitionIBD[173]
miR-204WJMSCs, BMMSCsExosomesSTAT3Proliferation InhibitionPulmonary Hypertension[174]
anti-miR-206UCMSCs-BDNF, (Egr-1, PSD-95)Neuroprotection, Neuronal Function PromotionAge-related Cognitive Decline[175]
anti-miR-210UCMSCsEVs-Immunosuppressive PropertiesPsoriasis[176]
miR-223UCMSCs, (BMMSCs)ExosomesICAM-1T cell Adhesion/Migration/Infiltration Inhibition, Inflammatory Factors SuppressionAcute Graft-versus-Host Disease[177]
anti-miR-301a-3pUCMSCs-IGF-1, PI3K/Akt/FOXO3aBurn-induced Apoptosis/Organ Vascular Permeability InhibitionVascular Endothelial Barrier Dysfunction[178]
miR-302d-3pUCMSCsExosomesFLT4, VEGFR3/AKTMigration/Tube Formation/Lymphangiogenesis InhibitionIBD[179]
miR-326UCMSCsExosomesNEDD8, NF-κBNeddylation/Inflammation InhibitionIBD[180]
miR-335-5pUCMSCsExosomesADAM19Inflammation/EMT InhibitionRenal Fibrosis[181]
miR-342-3pUCMSCsExosomesEDNRAThrombus Formation Inhibition, Angiogenesis PromotionDeep Vein Thrombosis[182]
miR-378UCMSCsEVsPSMD14/TGF-β1/Smad2/3Mesangial Hyperplasia/Fibrosis/Proliferation SuppressionMesangial Proliferative Glomerulonephritis (MsPGN)[183]
miR-378a-5pUCMSCsExosomesNLRP3Macrophage Pyroptosis/Inflammation Inhibition, Cell Survival PromotionIBD/Colitis[184]
miR-455-3pUCMSCs-PAK2Profibrogenic Markers SuppressionLiver Fibrosis[185]
miR-499WJMSCs-TGFβR 1/3Creatine Kinase Decrease, Muscle Regeneration, Apoptosis/Fibrosis InhibitionDuchenne Muscular Dystrophy[186]
miR-627-5pUCMSCsExosomesFTOCell Survival Promotion, Apoptosis Inhibition, Glucose/Lipid Metabolism ImprovementNon-Alcoholic Fatty Liver Disease[187]
miR-1246UCMSCsExosomesPRSS23/Snail/α-SMAAngiogenesis Promotion, Apoptosis/Hypoxic Damage ReductionChronic Heart Failure[188]
miR-1348a-3pUCMSCsExosomesSerpine1Vascular Smooth Muscle Cell Phenotypic Switching/Migration InhibitionNeointimal Hyperplasia[189]
UCMSCs: Umbilical Cord-derived Mesenchymal Stem Cells, UCBMSCs: Umbilical Cord Blood-derived Mesenchymal Stem Cells, WJMSCs: Wharton’s Jelly-derived Mesenchymal Stem Cells, EVs: Extracellular vesicles, SLE: Systemic Lupus Erythematosus, DAH: Diffuse Alveolar Hemorrhage, IBD: Inflammatory Bowel Disease.

3.3. Regenerative Medicine (Wound Healing/Bone Regeneration)

UCMSCs have additional noteworthy parts to play in the general area of regenerative medicine, especially in bone and skin regeneration, as their unique proliferative and angiogenic biological properties appear to be directly involved in the repair and reconstruction of bone fractures and epithelial injuries. miR-21 is yet again one of the most commonly mentioned microRNAs involved in both cases. Its delivery through UCMSC-EVs was reported to promote corneal wound healing both in vitro and in vivo, via the modulation of the PTEN/PI3K/Akt pathway [70], while miR-21-5p along with miR-125b-5p, transferred through UCBMSC-exosomes, could potentially target and inhibit TGF-β receptors I and II to prevent scar formation and increase wound closure rate [190]. Exosomal miR-150-5p was found to also target the PTEN/PI3K/Akt pathway to facilitate skin wound restoration [108], while miR-125b was further implicated in the promotion of wound healing under hypoxic conditions through the inhibition of TP53INP1-induced endothelial cell apoptosis [191]. EV-transferred miR-27b was shown to target Itchy E3 ubiquitin protein ligase (ITCH) in order to enhance cutaneous wound healing in model mice [83], while miR-17-5p was observed to act similarly in the case of diabetic wounds by regulating the PTEN/AKT/HIF-1α/VEGF axis and ameliorating the impaired angiogenic abilities of endothelial cells in a high glucose environment [64].
In regard to bone repair, exosomal miR-21 injection was proposed as a novel therapeutic intervention for osteonecrosis of the femoral head (ONFH), with miR-21 suppressing the expression of SOX5 and EZH2 [74] as well as regulating the PTEN/Akt pathway [71] to enhance angiogenesis, osteogenesis, and osteocyte survival in injured rats. miR-21-containing exosomes, artificially reinforced with hyaluronic acid hydrogel and nanohydroxyapatite/poly-ε-caprolactone, were also observed to enhance the healing of large bone defects through the NOTCH1/DLL4 pathway and the induction of the angiogenetic process [72]. Additionally, recent studies have reported that several other UCMSC-derived miRNAs have a mediating role to play when it comes to the clinical management of osteoarthritis (OA) and cartilage-related injuries. miR-122-5p, miR-148a-3p, miR-486-5p, miR-let-7a-5p, and miR-100-5p were identified as highly expressed in hUCMSC-EVs, and their presence might explain the chondroprotective properties of hUCMSC-EV treatment, since all five microRNAs were reported to be capable of preventing cartilage degradation by promoting M2 instead of M1 macrophage polarization and easing inflammatory and immune system reactions in OA rats [192]. Treatment with exosomes carrying miR-140-3p was discovered to improve joint injury as well as reduce chondrocyte cell apoptosis in rats with rheumatoid arthritis (RA) through the targeting of SGK1 [101], while intra-articular injection with UCMSCs previously transfected with miR-140-5p could further induce the chondrogenetic and self-healing capacity of injured cartilage tissue in OA rats [102]. miR-181c-5p, delivered by hUCMSC-EVs, was shown to contribute to the repair of cartilage injuries by negatively regulating SMAD7 and promoting BMP2 expression, which in turn amplifies the proliferation, migration, and osteoblastic differentiation capacity of bone marrow stem cells [111].
WJMSC-derived small extracellular vesicles appear to function as natural nanomaterials and through the delivery of multiple microRNAs, including let-7e-5p, miR-423-5p, miR-199a-3p, miR-125b-5p, miR-142-3p, and miR-92a-3p, and the regulation of their various target genes, they were reported to contribute to the maintenance of cell homeostasis in chondrocytes, enhancing proliferation, migration, and infiltration of M2 macrophages while suppressing injury degeneration in in vivo osteoarthritis (OA) model rats [193]. Furthermore, exosomal miR-100-5p and EV-miR-1208 were both demonstrated to inhibit OA development and progression by regulating their target genes NOX4 [93] and METTL3 [125], respectively.
Interestingly, attempts to produce specifically engineered UCMSCs containing gelatin methacrylate and nanoclay hydrogel demonstrated particularly promising results for cartilage defect regeneration through the stable release of EVs that carry miR-23a-3p, which successfully induces PTEN/AKT pathway activity [78]. It should also be noted that UCMSC priming with different cytokines was recently discovered to alter the composition of the miRNA UCMSC-EV cargo, ultimately determining the EVs’ therapeutic effectiveness [109]. As such, UCMSC-EVs previously treated with anti-inflammatory cytokines such as TGF-β and IFN-α presented reduced packaging of miR-181b-3p, which negatively modulates chondrocyte proliferation and regeneration and might naturally contribute to several chondrocyte-associated diseases [109].

3.4. Anti-Cancer Treatment

An increasing number of studies suggest that UCMSC-exosomes could also play a mediating role when it comes to the treatment of various cancer types, since many of their cargo miRNAs are reportedly associated with several hallmark oncogenic processes (Table 4). UCMSC-derived exosomes transfected with miR-21-5p were shown to possess tumor-suppressing properties in the case of breast cancer cells by directly targeting ZNF367 and suppressing its migration and invasion properties [194]. Similarly, UCMSC-exosomes modified to overexpress miR-148b-3p were found to inhibit tumor formation and the process of EMT in mice by blocking the oncogenic properties of breast cancer cells and promoting their apoptosis [195], while two more novel anti-oncogenic microRNAs, miR-3182 and miR-3143, were identified as rather promising candidates for the exosome-based therapy of triple negative cancer, with their targets revealed as key genes in cancer pathogenesis [196,197]. On the other hand, miR-224-5p was found highly upregulated in both breast cancer cells and tissues, promoting the autophagic and the oncogenic activity of breast cancer cells by targeting HOXA5 [198]. Interestingly, UCMSC-derived exosomes transfected with the miR-224-5p inhibitor were shown to decrease the proliferation and viability levels of breast cancer cells as well as reduce the volume of tumor tissue in model mice [198].
Delivery of exosomes and EVs loaded with anti-tumorigenic microRNAs also proved efficient in the potential treatment of endometrial and ovarian cancers. miR-503-3p [199] and miR-302a [200] were both found to be capable of suppressing endometrial cancer progression via the inhibition of MEST and cyclin D1, respectively, while miR-200c, combined with UCMSC-secreted IL-21, presented the same attenuating effects in the case of ovarian adenocarcinoma cells through inhibition of the Wnt/β-catenin pathway [201]. Additionally, exosomal microRNA-146a appears to sensitize ovarian cancer cells to the chemotherapeutic agents docetaxel and taxane via the LAMC2/PI3K/Akt axis [202], a property that could prove quite beneficial when it comes to tumor chemoresistance in the clinical setting. High-grade gliomas are another example of highly malignant tumors for which there presently exist no curative treatment protocols and thus for which the need for novel therapeutic methods is imperative. Synthetic miR-124 and miR-145 delivered through MSCs of various origins, including the umbilical cord, were shown to successfully suppress SCP-1 and Sox2, respectively, in order to decrease the migration of cancer cells and the self-renewal abilities of glioma stem cells [203], while WJMSC-delivered miR-124 was suggested to increase the sensitivity of glioblastoma cells to temozolomide treatment [204]. Exosomes carrying packaged lncRNA PTENP1 were also reported to be capable of suppressing the oncogenic activity of miR-10a-5p in glioma cells and enhancing PTEN expression, ultimately decreasing cancer cell viability [205].
Several encouraging studies on the effects of UCMSC-exosome delivery exist in the case of cholangiocarcinoma, as exosomal miR-15a-5p and miR-127-3p were demonstrated to regulate a number of key oncogenic cancer cell properties, including growth, invasion, metastasis, apoptosis, and EMT, through the downregulation of their respective target genes, CHEK1 [206] and ITGA6 [207]. miR-320a was revealed to function in a similar tumor-suppressing manner in lung cancer cells via the SOX4/Wnt/β-catenin pathway [208], while in contrast, EV-derived miR-410 was shown to promote the malignant properties of lung adenocarcinoma cells [209], suggesting that its artificial inhibition could perhaps pose a future therapeutic option. The beneficial effects of UCMSC-transferred microRNAs were also explored in nasopharyngeal and esophageal cancer, with exosome-delivered miR-181a significantly delaying nasopharyngeal carcinoma progression via the inhibition of KDM5C [210], while microRNAs miR-655-3p and microRNA-375 downregulated HIF-1α [211] and ENAH [212], respectively, to achieve the same suppressing effect in esophageal carcinoma. Additionally, quite recently the anti-tumor properties of exosome and EV-enclosed microRNAs were proven to be efficient in the cases of colorectal [213], gastric [214], bladder [215], thyroid [216], and prostate [217] cancer, as well as in hepatocellular carcinoma [218], pancreatic ductal adenocarcinoma [219], Wilms tumor [220], and chronic myeloid leukemia [221], further affirming that UCMSC-transferred microRNAs could indeed become valuable tools for prospective clinical applications in the field of cancer therapy.
Table 4. miRNA-guided applications of perinatal and neonatal MSCs in anti-cancer treatment.
Table 4. miRNA-guided applications of perinatal and neonatal MSCs in anti-cancer treatment.
miRNATissue OriginVehicle TypeTarget (Gene/Pathway)FunctionClinical ApplicationReference
miR-10a-5pUCMSCsExosomesPTENCell Growth/Cell Survival PromotionGlioma[205]
miR-15a-5pUCMSCsExosomesCHEK1Proliferation/Invasion/Migration/EMT Inhibition, Apoptosis PromotionCholangiocarcinoma[206]
UCMSCsExosomesSEPT2Proliferation/Migration/Invasion Inhibition, Apoptosis PromotionWilms Tumor[220]
miR-21-5pUCMSCsExosomesZNF367Migration/Invasion InhibitionBreast Cancer[194]
miR-30c-5pUCMSCsEVsPELI1/PI3K/AKTProliferation/Migration InhibitionPapillary Thyroid Carcinoma[216]
miR-100-5pUCMSCsExosomes-Proliferation/Migration PromotionPancreatic Ductal Adenocarcinoma[222]
miR-124WJMSCs-CDK6Proliferation/Migration Suppression, Chemosensitivity PromotionGlioblastoma Multiform[204]
miR-125bWJMSCsEVsHIF1αProliferation/EMT/Angiogenesis InhibitionTriple Negative Breast Cancer[223]
miR-127-3pUCMSCsEVsITGA6/TGF-β1/SmadProliferation/Invasion/Migration/EMT Inhibition, Apoptosis PromotionCholangiocarcinoma[207]
miR-139-5pUCMSCsExosomesPRC1Proliferation/Migration/Invasion SuppressionBladder Cancer[215]
miR-145-5pUCMSCsExosomesTGF-β/Smad3Proliferation/Invasion Inhibition, Apoptosis/Cell Cycle Arrest PromotionPancreatic Ductal Adenocarcinoma[219]
miR-145a-5pUCMSCsExosomesUSP6/GLS1Apoptosis/Chemosensitivity PromotionChronic Myeloid Leukemia[221]
miR-146aUCMSCsExosomesLAMC2/PI3K/AktCell Growth Suppression, Chemosensitivity PromotionOvarian Cancer[202]
miR-148b-3pUCMSCsExosomesTRIM59Proliferation/Invasion/Migration Inhibition, Apoptosis PromotionBreast Cancer[195]
miR-181aUCMSCsExosomesKDM5CCell Growth SuppressionNasopharyngeal Carcinoma[210]
miR-200cUCMSCs-Wnt/β-cateninTumor Growth/Metastasis InhibitionOvarian Cancer[201]
anti-miR-224-5pUCMSCsExosomesHOXA5Proliferation Inhibition, Apoptosis PromotionBreast Cancer[198]
miR-302aUCMSCsEVscyclin D1/AKTProliferation/Migration InhibitionEndometrial Cancer[200]
miR-320aUCMSCsExosomesSOX4/Wnt/β-cateninProliferation/Metastasis InhibitionLung Cancer[208]
miR-375UCMSCsExosomesENAHProliferation/Invasion/Migration/Tumorsphere Formation Inhibition, Apoptosis PromotionEsophageal Squamous Cell Carcinoma[212]
anti-miR-375UCMSCsExosomesPTPN4/STAT3Proliferation/Migration/Invasion/Chemoresistance Suppression, Apoptosis PromotionProstate Cancer[217]
miR-410UCMSCsEVsPTENProliferation Promotion, Apoptosis SuppressionLung Adenocarcinoma[209]
miR-431-5pUCMSCsExosomesPRDX1Cell Growth Suppression, Prognostic MarkerColorectal Cancer[213]
miR-451aUCMSCsExosomesADAM10Paclitaxel Resistance/Cell Cycle Transition/Proliferation/Migration/Invasion Inhibition, Apoptosis PromotionHepatocellular Carcinoma[218]
miR-503-3pUCBMSCsExosomesMESTCell Growth SuppressionEndometrial Cancer[199]
miR-655-3pUCMSCsEVsHIF-1α/LMO4/HDAC2Cell Growth/Metastasis SuppressionEsophageal Cancer[211]
miR-3182UCMSCsExosomesmTOR, S6KB1Proliferation/Migration Inhibition, Apoptosis PromotionTriple Negative Breast Cancer[197]
miR-6785-5pUCMSCsExosomesINHBAAngiogenesis/Metastasis SuppressionGastric Cancer[214]
miR-11401UCMSCsEVsSCOTIN/p53Doxorubicin-induced Apoptosis InhibitionChemotherapy[224]
UCMSCs: Umbilical Cord-derived Mesenchymal Stem Cells, UCBMSCs: Umbilical Cord Blood-derived Mesenchymal Stem Cells, WJMSCs: Wharton’s Jelly-derived Mesenchymal Stem Cells, EVs: Extracellular vesicles, EMT: Epithelial-to-Mesenchymal Transition.
Finally, apart from separate miRNAs, miRNA groups and signatures are being exploited for their ability to guide UCMSC-based regenerative and therapeutic applications (Table 5).

4. Future Directions

As thoroughly described, the exploitation of miRNA regulatory properties along with the unique characteristics of UCMSCs is a quite promising research field with a multitude of applications, expanding from aesthetic interventions to therapeutic regimes. Nevertheless, there are currently certain limitations that need to be overcome for the successful implementation of miRNA-based therapies in the clinic. Upon administration, the first obstacle that naked and unmodified miRNA molecules need to overcome is low cell membrane penetration [235], followed by rapid degradation and clearance from the circulation [236,237]. A solution to these problems is the use of evolved delivery vehicles, including nanocarriers and exosomes, that stabilize and protect miRNAs until they reach their targets. Still, targeting of extra-hepatic tissues remains a barrier, thus limiting the use of miRNA-based therapies. Although lipid and polymer nanoparticles can achieve efficient cellular uptake and stability of delivered miRNAs, they selectively accumulate in the liver instead of the intended target tissues [238]. Of high significance are also the off-target effects of miRNAs, which are mainly attributed to the multiple intracellular targets of each miRNA [239], along with their ability to abnormally activate the host’s immune system [240]; These effects can lead to serious toxicities and adverse events that lower the therapeutic value of miRNA-based therapies. The combined use of MSCs could alleviate these effects, given the immunosuppressive effects of the latter.
Currently, there are a few clinical studies in different stages of processing (https://clinicaltrials.gov). In some of those, MSCs from different sources are evaluated for their safety and efficacy against metabolic and autoimmune diseases based on the expression levels of certain miRNAs among other biomarkers. Accordingly, the intracorporal delivery of MSC-derived exosomes considered to carry several essential elements, including miRNAs, is the main subject of investigation in ongoing phase I/II studies. One completed phase I trial that examined the biodistribution and safety of inhaled MSC-EVs in healthy volunteers revealed no adverse events after seven days of treatment (PMID: 34429860). Interestingly, there are two registered clinical trials assessing the efficacy of MSC exosome inhalation in coronavirus disease 2019 (COVID-19) patients; the completed one determined that inhalation of exosomes twice a day for a total period of 10 days increased SpO2 concentration in the blood of patients with pneumonia in the absence of any adverse events (https://clinicaltrials.gov/NCT04491240 (accessed on 20 May 2023)).
Overall, miRNA-driven multi-lineage differentiation of UCMSCs forms a subcategory of the field of MSC-based therapeutics. Recent advances in the field, combined with the putative cell-free implementation of UCMSC-EVs carrying key miRNAs, pave the way for their successful application in therapeutic and regenerative remedies that will enhance cure rates in the absence of serious side effects. Importantly, the evolution of induced pluripotent stem cell (iPSC) research provides the ability to obtain large numbers of iPSC-derived MSC-like cells with properties similar to their native counterparts, thus offering a double advantage: an abundance of cells that can be exploited in therapeutic protocols with a concomitant absence of ethical issues. At the same time, since altered miRNA levels are detected in cells derived from differentiated MSCs, these miRNAs can be utilized as powerful biomarkers of the differentiation process and the quality of the descendant cells, as well.

Author Contributions

Conceptualization, M.G. and V.Z.; writing—original draft preparation, A.C.T., M.G. and A.T.; review and editing, A.C.T., M.G., P.Z., M.T., I.C. and V.Z.; visualization, A.C.T., A.T., P.Z. and M.T.; supervision, M.G., I.C. and V.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Campagnoli, C.; Roberts, I.A.; Kumar, S.; Bennett, P.R.; Bellantuono, I.; Fisk, N.M. Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver, and bone marrow. Blood 2001, 98, 2396–2402. [Google Scholar] [CrossRef] [PubMed]
  2. Rochefort, G.Y.; Delorme, B.; Lopez, A.; Hérault, O.; Bonnet, P.; Charbord, P.; Eder, V.; Domenech, J. Multipotential mesenchymal stem cells are mobilized into peripheral blood by hypoxia. Stem Cells 2006, 24, 2202–2208. [Google Scholar] [CrossRef] [PubMed]
  3. Aggarwal, S.; Pittenger, M.F. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005, 105, 1815–1822. [Google Scholar] [CrossRef] [PubMed]
  4. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  5. Gökçinar-Yagci, B.; Özyüncü, Ö.; Çelebi-Saltik, B. Isolation, characterisation and comparative analysis of human umbilical cord vein perivascular cells and cord blood mesenchymal stem cells. Cell Tissue Bank. 2016, 17, 345–352. [Google Scholar] [CrossRef]
  6. In ‘t Anker, P.S.; Scherjon, S.A.; Kleijburg-van der Keur, C.; de Groot-Swings, G.M.; Claas, F.H.; Fibbe, W.E.; Kanhai, H.H. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells 2004, 22, 1338–1345. [Google Scholar] [CrossRef]
  7. Christodoulou, I.; Kolisis, F.N.; Papaevangeliou, D.; Zoumpourlis, V. Comparative Evaluation of Human Mesenchymal Stem Cells of Fetal (Wharton’s Jelly) and Adult (Adipose Tissue) Origin during Prolonged In Vitro Expansion: Considerations for Cytotherapy. Stem Cells Int. 2013, 2013, 246134. [Google Scholar] [CrossRef]
  8. Amit, M.; Carpenter, M.K.; Inokuma, M.S.; Chiu, C.P.; Harris, C.P.; Waknitz, M.A.; Itskovitz-Eldor, J.; Thomson, J.A. Clonally derived human embryonic stem cell lines maintain pluripotency and proliferative potential for prolonged periods of culture. Dev. Biol. 2000, 227, 271–278. [Google Scholar] [CrossRef] [PubMed]
  9. Weiss, M.L.; Anderson, C.; Medicetty, S.; Seshareddy, K.B.; Weiss, R.J.; VanderWerff, I.; Troyer, D.; McIntosh, K.R. Immune properties of human umbilical cord Wharton’s jelly-derived cells. Stem Cells 2008, 26, 2865–2874. [Google Scholar] [CrossRef]
  10. Mansilla, E.; Marin, G.H.; Sturla, F.; Drago, H.E.; Gil, M.A.; Salas, E.; Gardiner, M.C.; Piccinelli, G.; Bossi, S.; Salas, E.; et al. Human mesenchymal stem cells are tolerized by mice and improve skin and spinal cord injuries. Transplant. Proc. 2005, 37, 292–294. [Google Scholar] [CrossRef]
  11. Girousse, A.; Mathieu, M.; Sastourné-Arrey, Q.; Monferran, S.; Casteilla, L.; Sengenès, C. Endogenous Mobilization of Mesenchymal Stromal Cells: A Pathway for Interorgan Communication? Front. Cell Dev. Biol. 2020, 8, 598520. [Google Scholar] [CrossRef] [PubMed]
  12. Margiana, R.; Markov, A.; Zekiy, A.O.; Hamza, M.U.; Al-Dabbagh, K.A.; Al-Zubaidi, S.H.; Hameed, N.M.; Ahmad, I.; Sivaraman, R.; Kzar, H.H.; et al. Clinical application of mesenchymal stem cell in regenerative medicine: A narrative review. Stem Cell Res. Ther. 2022, 13, 366. [Google Scholar] [CrossRef] [PubMed]
  13. Hmadcha, A.; Martin-Montalvo, A.; Gauthier, B.R.; Soria, B.; Capilla-Gonzalez, V. Therapeutic Potential of Mesenchymal Stem Cells for Cancer Therapy. Front. Bioeng. Biotechnol. 2020, 8, 43. [Google Scholar] [CrossRef]
  14. Gurtan, A.M.; Sharp, P.A. The role of miRNAs in regulating gene expression networks. J. Mol. Biol. 2013, 425, 3582–3600. [Google Scholar] [CrossRef] [PubMed]
  15. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed]
  16. Broughton, J.P.; Lovci, M.T.; Huang, J.L.; Yeo, G.W.; Pasquinelli, A.E. Pairing beyond the Seed Supports MicroRNA Targeting Specificity. Mol. Cell 2016, 64, 320–333. [Google Scholar] [CrossRef] [PubMed]
  17. Vasudevan, S. Posttranscriptional upregulation by microRNAs. RNA 2012, 3, 311–330. [Google Scholar] [CrossRef]
  18. Vasudevan, S.; Steitz, J.A. AU-rich-element-mediated upregulation of translation by FXR1 and Argonaute 2. Cell 2007, 128, 1105–1118. [Google Scholar] [CrossRef]
  19. Orom, U.A.; Nielsen, F.C.; Lund, A.H. MicroRNA-10a binds the 5’UTR of ribosomal protein mRNAs and enhances their translation. Mol. Cell 2008, 30, 460–471. [Google Scholar] [CrossRef]
  20. Jonas, S.; Izaurralde, E. Towards a molecular understanding of microRNA-mediated gene silencing. Nat. Rev. Genet. 2015, 16, 421–433. [Google Scholar] [CrossRef]
  21. Bottini, S.; Hamouda-Tekaya, N.; Mategot, R.; Zaragosi, L.E.; Audebert, S.; Pisano, S.; Grandjean, V.; Mauduit, C.; Benahmed, M.; Barbry, P.; et al. Post-transcriptional gene silencing mediated by microRNAs is controlled by nucleoplasmic Sfpq. Nat. Commun. 2017, 8, 1189. [Google Scholar] [CrossRef] [PubMed]
  22. Paul, P.; Chakraborty, A.; Sarkar, D.; Langthasa, M.; Rahman, M.; Bari, M.; Singha, R.S.; Malakar, A.K.; Chakraborty, S. Interplay between miRNAs and human diseases. J. Cell. Physiol. 2018, 233, 2007–2018. [Google Scholar] [CrossRef] [PubMed]
  23. Collino, F.; Bruno, S.; Deregibus, M.C.; Tetta, C.; Camussi, G. MicroRNAs and mesenchymal stem cells. Vitam. Horm. 2011, 87, 291–320. [Google Scholar] [CrossRef]
  24. Yang, C.; Luo, M.; Chen, Y.; You, M.; Chen, Q. MicroRNAs as Important Regulators Mediate the Multiple Differentiation of Mesenchymal Stromal Cells. Front. Cell Dev. Biol. 2021, 9, 619842. [Google Scholar] [CrossRef]
  25. Oveili, E.; Vafaei, S.; Bazavar, H.; Eslami, Y.; Mamaghanizadeh, E.; Yasamineh, S.; Gholizadeh, O. The potential use of mesenchymal stem cells-derived exosomes as microRNAs delivery systems in different diseases. Cell Commun. Signal. CCS 2023, 21, 20. [Google Scholar] [CrossRef] [PubMed]
  26. Fallah, A.; Alipour, M. Overexpression Effects of miR-424 and BMP2 on the Osteogenesis of Wharton’s Jelly-Derived Stem Cells. BioMed Res. Int. 2021, 2021, 7031492. [Google Scholar] [CrossRef]
  27. Hwang, S.; Park, S.K.; Lee, H.Y.; Kim, S.W.; Lee, J.S.; Choi, E.K.; You, D.; Kim, C.S.; Suh, N. miR-140-5p suppresses BMP2-mediated osteogenesis in undifferentiated human mesenchymal stem cells. FEBS Lett. 2014, 588, 2957–2963. [Google Scholar] [CrossRef]
  28. Huang, M.; Qing, Y.; Shi, Q.; Cao, Y.; Song, K. miR-342-3p elevates osteogenic differentiation of umbilical cord mesenchymal stem cells via inhibiting Sufu in vitro. Biochem. Biophys. Res. Commun. 2017, 491, 571–577. [Google Scholar] [CrossRef]
  29. Wang, Y.; Zhang, S.; Yang, H.; Cao, Y.; Yu, D.; Zhao, Y.; Cao, Y. MicroRNA-196a-5p overexpression in Wharton’s jelly umbilical cord stem cells promotes their osteogenic differentiation and new bone formation in bone defects in the rat calvarium. Cell Tissue Res. 2022, 390, 245–260. [Google Scholar] [CrossRef]
  30. Zhao, G.; Luo, W.-D.; Yuan, Y.; Lin, F.; Guo, L.-M.; Ma, J.-J.; Chen, H.-B.; Tang, H.; Shu, J. LINC02381, a sponge of miR-21, weakens osteogenic differentiation of hUC-MSCs through KLF12-mediated Wnt4 transcriptional repression. J. Bone Miner. Metab. 2022, 40, 66–80. [Google Scholar] [CrossRef]
  31. Meng, Y.B.; Li, X.; Li, Z.Y.; Zhao, J.; Yuan, X.B.; Ren, Y.; Cui, Z.D.; Liu, Y.D.; Yang, X.J. microRNA-21 promotes osteogenic differentiation of mesenchymal stem cells by the PI3K/β-catenin pathway. J. Orthop. Res. 2015, 33, 957–964. [Google Scholar] [CrossRef] [PubMed]
  32. Yahao, G.; Xinjia, W. The Role and Mechanism of Exosomes from Umbilical Cord Mesenchymal Stem Cells in Inducing Osteogenesis and Preventing Osteoporosis. Cell Transplant. 2021, 30, 9636897211057465. [Google Scholar] [CrossRef] [PubMed]
  33. Cao, L.; Liu, W.; Zhong, Y.; Zhang, Y.; Gao, D.; He, T.; Liu, Y.; Zou, Z.; Mo, Y.; Peng, S. Linc02349 promotes osteogenesis of human umbilical cord-derived stem cells by acting as a competing endogenous RNA for miR-25-3p and miR-33b-5p. Cell Prolif. 2020, 53, e12814. [Google Scholar] [CrossRef] [PubMed]
  34. Xue, Z.L.; Meng, Y.L.; Ge, J.H. Upregulation of miR-132 attenuates osteoblast differentiation of UC-MSCs. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 1580–1587. [Google Scholar] [CrossRef]
  35. Li, H.; Li, T.; Fan, J.; Li, T.; Fan, L.; Wang, S.; Weng, X.; Han, Q.; Zhao, R.C. miR-216a rescues dexamethasone suppression of osteogenesis, promotes osteoblast differentiation and enhances bone formation, by regulating c-Cbl-mediated PI3K/AKT pathway. Cell Death Differ. 2015, 22, 1935–1945. [Google Scholar] [CrossRef]
  36. Raut, A.; Khanna, A. Enhanced expression of hepatocyte-specific microRNAs in valproic acid mediated hepatic trans-differentiation of human umbilical cord derived mesenchymal stem cells. Exp. Cell Res. 2016, 343, 237–247. [Google Scholar] [CrossRef]
  37. Khosravi, M.; Azarpira, N.; Shamdani, S.; Hojjat-Assari, S.; Naserian, S.; Karimi, M.H. Differentiation of umbilical cord derived mesenchymal stem cells to hepatocyte cells by transfection of miR-106a, miR-574-3p, and miR-451. Gene 2018, 667, 1–9. [Google Scholar] [CrossRef]
  38. Wang, H.; Ban, W.; Wang, T.; Li, Z.; Dang, X. miR-20b/106a modulate Ngn2 gene expression during neural differentiation of human umbilical cord mesenchymal stem cells. Neuroreport 2017, 28, 1225–1231. [Google Scholar] [CrossRef]
  39. Choi, S.W.; Shin, J.H.; Kim, J.J.; Shin, T.H.; Seo, Y.; Kim, H.S.; Kang, K.S. Direct cell fate conversion of human somatic stem cells into cone and rod photoreceptor-like cells by inhibition of microRNA-203. Oncotarget 2016, 7, 42139–42149. [Google Scholar] [CrossRef]
  40. Cao, B.; Dai, X. Platelet lysate induces chondrogenic differentiation of umbilical cord-derived mesenchymal stem cells by regulating the lncRNA H19/miR-29b-3p/SOX9 axis. FEBS Open Bio 2020, 10, 2656–2665. [Google Scholar] [CrossRef]
  41. Jing, H.; Zhang, X.; Luo, K.; Luo, Q.; Yin, M.; Wang, W.; Zhu, Z.; Zheng, J.; He, X. miR-381-abundant small extracellular vesicles derived from kartogenin-preconditioned mesenchymal stem cells promote chondrogenesis of MSCs by targeting TAOK1. Biomaterials 2020, 231, 119682. [Google Scholar] [CrossRef] [PubMed]
  42. Choi, S.W.; Kim, J.J.; Seo, M.S.; Park, S.B.; Shin, T.H.; Shin, J.H.; Seo, Y.; Kim, H.S.; Kang, K.S. Inhibition by miR-410 facilitates direct retinal pigment epithelium differentiation of umbilical cord blood-derived mesenchymal stem cells. J. Vet. Sci. 2017, 18, 59–65. [Google Scholar] [CrossRef] [PubMed]
  43. Li, Y.; Shi, X.; Yang, L.; Mou, Y.; Li, Y.; Dang, R.; Li, C. Hypoxia promotes the skewed differentiation of umbilical cord mesenchymal stem cells toward type II alveolar epithelial cells by regulating microRNA-145. Gene 2017, 630, 68–75. [Google Scholar] [CrossRef]
  44. Chen, W.; Jiang, W.; Dong, J.; Wang, J.; Wang, B. miR-200b-3p Induces the Formation of Insulin-Producing Cells from Umbilical Cord Mesenchymal Stem Cells by Targeting ZEB2. Crit. Rev. Eukaryot. Gene Expr. 2022, 32, 33–46. [Google Scholar] [CrossRef]
  45. Bai, C.; Gao, Y.; Li, X.; Wang, K.; Xiong, H.; Shan, Z.; Zhang, P.; Wang, W.; Guan, W.; Ma, Y. MicroRNAs can effectively induce formation of insulin-producing cells from mesenchymal stem cells. J. Tissue Eng. Regen. Med. 2017, 11, 3457–3468. [Google Scholar] [CrossRef]
  46. Hu, K.; Xu, C.; Ni, H.; Xu, Z.; Wang, Y.; Xu, S.; Ji, K.; Xiong, J.; Liu, H. Mir-218 contributes to the transformation of 5-Aza/GF induced umbilical cord mesenchymal stem cells into hematopoietic cells through the MITF pathway. Mol. Biol. Rep. 2014, 41, 4803–4816. [Google Scholar] [CrossRef] [PubMed]
  47. Liu, L.; Liu, H.; Chen, M.; Ren, S.; Cheng, P.; Zhang, H. miR-301b~miR-130b—PPARγ axis underlies the adipogenic capacity of mesenchymal stem cells with different tissue origins. Sci. Rep. 2017, 7, 1160. [Google Scholar] [CrossRef] [PubMed]
  48. Fang, S.; Xu, C.; Zhang, Y.; Xue, C.; Yang, C.; Bi, H.; Qian, X.; Wu, M.; Ji, K.; Zhao, Y.; et al. Umbilical Cord-Derived Mesenchymal Stem Cell-Derived Exosomal MicroRNAs Suppress Myofibroblast Differentiation by Inhibiting the Transforming Growth Factor-β/SMAD2 Pathway During Wound Healing. Stem Cells Transl. Med. 2016, 5, 1425–1439. [Google Scholar] [CrossRef]
  49. Gu, W.; Hong, X.; Le Bras, A.; Nowak, W.N.; Issa Bhaloo, S.; Deng, J.; Xie, Y.; Hu, Y.; Ruan, X.Z.; Xu, Q. Smooth muscle cells differentiated from mesenchymal stem cells are regulated by microRNAs and suitable for vascular tissue grafts. J. Biol. Chem. 2018, 293, 8089–8102. [Google Scholar] [CrossRef]
  50. Qing, Y.; Huang, M.; Cao, Y.; Du, T.; Song, K. Effects of miRNA-342-3p in modulating Hedgehog signaling pathway of human umbilical cord mesenchymal stem cells by down-regulating Sufu. Oral Dis. 2019, 25, 1147–1157. [Google Scholar] [CrossRef]
  51. Hong, I.S.; Lee, H.Y.; Choi, S.W.; Kim, H.S.; Yu, K.R.; Seo, Y.; Jung, J.W.; Kang, K.S. The effects of hedgehog on RNA binding protein Msi1 during the osteogenic differentiation of human cord blood-derived mesenchymal stem cells. Bone 2013, 56, 416–425. [Google Scholar] [CrossRef] [PubMed]
  52. Han, X.; Yang, H.; Liu, H.; Zhang, C.; Cao, Y.; Fan, Z.; Shi, R. miR-196b-5p inhibits proliferation of Wharton’s jelly umbilical cord stem cells. FEBS Open Bio 2021, 11, 278–288. [Google Scholar] [CrossRef]
  53. Asgharzadeh, A.; Alizadeh, S.; Keramati, M.R.; Soleimani, M.; Atashi, A.; Edalati, M.; Kashani Khatib, Z.; Rafiee, M.; Barzegar, M.; Razavi, H. Upregulation of miR-210 promotes differentiation of mesenchymal stem cells (MSCs) into osteoblasts. Bosn. J. Basic Med. Sci. 2018, 18, 328–335. [Google Scholar] [CrossRef] [PubMed]
  54. Raut, A.; Khanna, A. High-throughput sequencing to identify microRNA signatures during hepatic differentiation of human umbilical cord Wharton’s jelly-derived mesenchymal stem cells. Hepatol.Res. Off. J. Jpn. Soc. Hepatol. 2017, 47, 910–927. [Google Scholar] [CrossRef] [PubMed]
  55. Cui, L.; Zhou, X.; Li, J.; Wang, L.; Wang, J.; Li, Q.; Chu, J.; Zheng, L.; Wu, Q.; Han, Z.; et al. Dynamic microRNA profiles of hepatic differentiated human umbilical cord lining-derived mesenchymal stem cells. PLoS ONE 2012, 7, e44737. [Google Scholar] [CrossRef] [PubMed]
  56. Cui, L.; Shi, Y.; Zhou, X.; Wang, X.; Wang, J.; Lan, Y.; Wang, M.; Zheng, L.; Li, H.; Wu, Q.; et al. A set of microRNAs mediate direct conversion of human umbilical cord lining-derived mesenchymal stem cells into hepatocytes. Cell Death Dis. 2013, 4, e918. [Google Scholar] [CrossRef]
  57. Zhou, X.; Cui, L.; Zhou, X.; Yang, Q.; Wang, L.; Guo, G.; Hou, Y.; Cai, W.; Han, Z.; Shi, Y.; et al. Induction of hepatocyte-like cells from human umbilical cord-derived mesenchymal stem cells by defined microRNAs. J. Cell. Mol. Med. 2017, 21, 881–893. [Google Scholar] [CrossRef]
  58. Zhuang, H.; Zhang, R.; Zhang, S.; Shu, Q.; Zhang, D.; Xu, G. Altered expression of microRNAs in the neuronal differentiation of human Wharton’s Jelly mesenchymal stem cells. Neurosci. Lett. 2015, 600, 69–74. [Google Scholar] [CrossRef]
  59. Chang, S.-J.; Weng, S.-L.; Hsieh, J.-Y.; Wang, T.-Y.; Chang, M.D.-T.; Wang, H.-W. MicroRNA-34a modulates genes involved in cellular motility and oxidative phosphorylation in neural precursors derived from human umbilical cord mesenchymal stem cells. BMC Med. Genom. 2011, 4, 65. [Google Scholar] [CrossRef]
  60. Sanooghi, D.; Lotfi, A.; Bagher, Z.; Barati, S.; Karimi, A.; Faghihi, F.; Lotfi, E.; Joghataei, M.T. Large-scale analysis of MicroRNA expression in motor neuron-like cells derived from human umbilical cord blood mesenchymal stem cells. Sci. Rep. 2022, 12, 5894. [Google Scholar] [CrossRef]
  61. Choi, S.W.; Kim, J.J.; Seo, M.S.; Park, S.B.; Kang, T.W.; Lee, J.Y.; Lee, B.C.; Kang, I.; Shin, T.H.; Kim, H.S.; et al. miR-410 Inhibition Induces RPE Differentiation of Amniotic Epithelial Stem Cells via Overexpression of OTX2 and RPE65. Stem Cell Rev. Rep. 2015, 11, 376–386. [Google Scholar] [CrossRef] [PubMed]
  62. Christodoulou, I.; Goulielmaki, M.; Devetzi, M.; Panagiotidis, M.; Koliakos, G.; Zoumpourlis, V. Mesenchymal stem cells in preclinical cancer cytotherapy: A systematic review. Stem Cell Res. Ther. 2018, 9, 336. [Google Scholar] [CrossRef] [PubMed]
  63. Christodoulou, I.; Goulielmaki, M. Suitability of Human Mesenchymal Stem Cells Derived from Fetal Umbilical Cord (Wharton’s Jelly) as an Alternative In Vitro Model for Acute Drug Toxicity Screening. Cells 2022, 11, 1102. [Google Scholar] [CrossRef] [PubMed]
  64. Wei, Q.; Wang, Y.; Ma, K.; Li, Q.; Li, B.; Hu, W.; Fu, X.; Zhang, C. Extracellular Vesicles from Human Umbilical Cord Mesenchymal Stem Cells Facilitate Diabetic Wound Healing Through MiR-17-5p-mediated Enhancement of Angiogenesis. Stem Cell Rev. Rep. 2022, 18, 1025–1040. [Google Scholar] [CrossRef]
  65. Huang, L.; Yang, L.; Ding, Y.; Jiang, X.; Xia, Z.; You, Z. Human umbilical cord mesenchymal stem cells-derived exosomes transfers microRNA-19a to protect cardiomyocytes from acute myocardial infarction by targeting SOX6. Cell Cycle 2020, 19, 339–353. [Google Scholar] [CrossRef]
  66. Zhang, L.; Song, Y.; Chen, L.; Li, D.; Feng, H.; Lu, Z.; Fan, T.; Chen, Z.; Livingston, M.J.; Geng, Q. MiR-20a-containing exosomes from umbilical cord mesenchymal stem cells alleviates liver ischemia/reperfusion injury. J. Cell. Physiol. 2020, 235, 3698–3710. [Google Scholar] [CrossRef]
  67. Du, T.; Zhou, J.; Chen, W.X.; Zhang, X.L.; Ji, T.Y.; Liu, J.; Rong, L.; Wang, L.D.; Zhou, R.J.; Ding, D.G. Microvesicles derived from human umbilical cord mesenchymal stem cells ameliorate renal ischemia-reperfusion injury via delivery of miR-21. Cell Cycle 2020, 19, 1285–1297. [Google Scholar] [CrossRef]
  68. Zhou, Y.; Zhu, Y.; Zhang, L.; Wu, T.; Wu, T.; Zhang, W.; Decker, A.M.; He, J.; Liu, J.; Wu, Y.; et al. Human Stem Cells Overexpressing miR-21 Promote Angiogenesis in Critical Limb Ischemia by Targeting CHIP to Enhance HIF-1α Activity. Stem Cells 2016, 34, 924–934. [Google Scholar] [CrossRef]
  69. Ma, Y.; Zhou, D. Human Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote the Proliferation of Schwann Cells by Regulating the PI3K/AKT Signaling Pathway via Transferring miR-21. Stem Cells Int. 2021, 2021, 1496101. [Google Scholar] [CrossRef]
  70. Liu, X.; Li, X. Umbilical Cord Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Deliver miR-21 to Promote Corneal Epithelial Wound Healing through PTEN/PI3K/Akt Pathway. Stem Cells Int. 2022, 2022, 1252557. [Google Scholar] [CrossRef]
  71. Kuang, M.J.; Huang, Y.; Zhao, X.G.; Zhang, R.; Ma, J.X.; Wang, D.C.; Ma, X.L. Exosomes derived from Wharton’s jelly of human umbilical cord mesenchymal stem cells reduce osteocyte apoptosis in glucocorticoid-induced osteonecrosis of the femoral head in rats via the miR-21-PTEN-AKT signalling pathway. Int. J. Biol. Sci. 2019, 15, 1861–1871. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, Y.; Xie, Y.; Hao, Z.; Zhou, P.; Wang, P.; Fang, S.; Li, L.; Xu, S.; Xia, Y. Correction to “Umbilical Mesenchymal Stem Cell-Derived Exosome-Encapsulated Hydrogels Accelerate Bone Repair by Enhancing Angiogenesis”. ACS Appl. Mater. Interfaces 2022, 14, 14834–14835. [Google Scholar] [CrossRef]
  73. Yao, Z.; Li, J. MicroRNA-21-3p Engineered Umbilical Cord Stem Cell-Derived Exosomes Inhibit Tendon Adhesion. J. Inflamm. Res. 2020, 13, 303–316. [Google Scholar] [CrossRef] [PubMed]
  74. Fang, S.; Liu, Z.; Wu, S.; Chen, X.; You, M.; Li, Y.; Yang, F.; Zhang, S.; Lai, Y.; Liu, P.; et al. Pro-angiognetic and pro-osteogenic effects of human umbilical cord mesenchymal stem cell-derived exosomal miR-21-5p in osteonecrosis of the femoral head. Cell Death Discov. 2022, 8, 226. [Google Scholar] [CrossRef] [PubMed]
  75. Zheng, Y.; Liu, J.; Chen, P.; Lin, L.; Luo, Y.; Ma, X.; Lin, J.; Shen, Y.; Zhang, L. Exosomal miR-22-3p from human umbilical cord blood-derived mesenchymal stem cells protects against lipopolysaccharid-induced acute lung injury. Life Sci. 2021, 269, 119004. [Google Scholar] [CrossRef]
  76. Dong, C.; Chen, M.; Cai, B.; Zhang, C.; Xiao, G.; Luo, W. Mesenchymal Stem Cell-Derived Exosomes Improved Cerebral Infarction via Transferring miR-23a-3p to Activate Microglia. NeuroMol. Med. 2022, 24, 290–298. [Google Scholar] [CrossRef]
  77. Song, Y.; Wang, B.; Zhu, X.; Hu, J.; Sun, J.; Xuan, J.; Ge, Z. Human umbilical cord blood-derived MSCs exosome attenuate myocardial injury by inhibiting ferroptosis in acute myocardial infarction mice. Cell Biol. Toxicol. 2021, 37, 51–64. [Google Scholar] [CrossRef]
  78. Hu, H.; Dong, L.; Bu, Z.; Shen, Y.; Luo, J.; Zhang, H.; Zhao, S.; Lv, F.; Liu, Z. miR-23a-3p-abundant small extracellular vesicles released from Gelma/nanoclay hydrogel for cartilage regeneration. J. Extracell. Vesicles 2020, 9, 1778883. [Google Scholar] [CrossRef]
  79. Wang, W.; Ji, Z.; Yuan, C.; Yang, Y. Mechanism of Human Umbilical Cord Mesenchymal Stem Cells Derived-Extracellular Vesicle in Cerebral Ischemia-Reperfusion Injury. Neurochem. Res. 2021, 46, 455–467. [Google Scholar] [CrossRef]
  80. Zhang, Y.; Wang, Y. Knockout of beta-2 microglobulin reduces stem cell-induced immune rejection and enhances ischaemic hindlimb repair via exosome/miR-24/Bim pathway. J. Cell. Mol. Med. 2020, 24, 695–710. [Google Scholar] [CrossRef]
  81. Li, G.; Xiao, L.; Qin, H.; Zhuang, Q.; Zhang, W.; Liu, L.; Di, C.; Zhang, Y. Exosomes-carried microRNA-26b-5p regulates microglia M1 polarization after cerebral ischemia/reperfusion. Cell Cycle 2020, 19, 1022–1035. [Google Scholar] [CrossRef] [PubMed]
  82. Liu, Z.; Wang, B.; Guo, Q. MiR-26b-5p-modified hUB-MSCs derived exosomes attenuate early brain injury during subarachnoid hemorrhage via MAT2A-mediated the p38 MAPK/STAT3 signaling pathway. Brain Res. Bull. 2021, 175, 107–115. [Google Scholar] [CrossRef] [PubMed]
  83. Cheng, S.; Xi, Z.; Chen, G.; Liu, K.; Ma, R. Extracellular vesicle-carried microRNA-27b derived from mesenchymal stem cells accelerates cutaneous wound healing via E3 ubiquitin ligase ITCH. J. Cell. Mol. Med. 2020, 24, 11254–11271. [Google Scholar] [CrossRef]
  84. Han, Q.; Wang, S.; Chen, D.; Gan, D.; Wang, T. Exosomes derived from human umbilical cord mesenchymal stem cells reduce tendon injuries via the miR-27b-3p/ARHGAP5/RhoA signaling pathway. Acta Biochim. Biophys. Sin. 2022, 54, 232–242. [Google Scholar] [CrossRef]
  85. Su, W.H.; Wang, C.J.; Hung, Y.Y.; Lu, C.W.; Ou, C.Y.; Tseng, S.H.; Tsai, C.C.; Kao, Y.T.; Chuang, P.C. MicroRNA-29a Exhibited Pro-Angiogenic and Anti-Fibrotic Features to Intensify Human Umbilical Cord Mesenchymal Stem Cells-Renovated Perfusion Recovery and Preventing against Fibrosis from Skeletal Muscle Ischemic Injury. Int. J. Mol. Sci. 2019, 20, 5859. [Google Scholar] [CrossRef] [PubMed]
  86. Yao, Z.; Li, J.; Xiong, H.; Cui, H.; Ning, J.; Wang, S.; Ouyang, X.; Qian, Y. MicroRNA engineered umbilical cord stem cell-derived exosomes direct tendon regeneration by mTOR signaling. J. Nanobiotechnology 2021, 19, 169. [Google Scholar] [CrossRef]
  87. Xiao, X.; Li, W.; Rong, D.; Xu, Z.; Zhang, Z.; Ye, H.; Xie, L.; Wu, Y.; Zhang, Y.; Wang, X. Human umbilical cord mesenchymal stem cells-derived extracellular vesicles facilitate the repair of spinal cord injury via the miR-29b-3p/PTEN/Akt/mTOR axis. Cell Death Discov. 2021, 7, 212. [Google Scholar] [CrossRef] [PubMed]
  88. Xiao, X.; Li, W.; Xu, Z.; Sun, Z.; Ye, H.; Wu, Y.; Zhang, Y.; Xie, L.; Jiang, D.; Jia, R.; et al. Extracellular vesicles from human umbilical cord mesenchymal stem cells reduce lipopolysaccharide-induced spinal cord injury neuronal apoptosis by mediating miR-29b-3p/PTEN. Connect. Tissue Res. 2022, 63, 634–649. [Google Scholar] [CrossRef]
  89. Yan, L.; Liu, G.; Wu, X. The umbilical cord mesenchymal stem cell-derived exosomal lncRNA H19 improves osteochondral activity through miR-29b-3p/FoxO3 axis. Clin. Transl. Med. 2021, 11, e255. [Google Scholar] [CrossRef]
  90. Shi, Q.; Sun, B.; Wang, D.; Zhu, Y.; Zhao, X.; Yang, X.; Zhang, Y. Circ6401, a novel circular RNA, is implicated in repair of the damaged endometrium by Wharton’s jelly-derived mesenchymal stem cells through regulation of the miR-29b-1-5p/RAP1B axis. Stem Cell Res. Ther. 2020, 11, 520. [Google Scholar] [CrossRef]
  91. Chen, W.X.; Zhou, J.; Zhou, S.S.; Zhang, Y.D.; Ji, T.Y.; Zhang, X.L.; Wang, S.M.; Du, T.; Ding, D.G. Microvesicles derived from human Wharton’s jelly mesenchymal stem cells enhance autophagy and ameliorate acute lung injury via delivery of miR-100. Stem Cell Res. Ther. 2020, 11, 113. [Google Scholar] [CrossRef]
  92. Liang, C.; Liu, Y.; Xu, H.; Huang, J.; Shen, Y.; Chen, F.; Luo, M. Exosomes of Human Umbilical Cord MSCs Protect Against Hypoxia/Reoxygenation-Induced Pyroptosis of Cardiomyocytes via the miRNA-100-5p/FOXO3/NLRP3 Pathway. Front. Bioeng. Biotechnol. 2020, 8, 615850. [Google Scholar] [CrossRef] [PubMed]
  93. Li, X.; Wang, Y.; Cai, Z.; Zhou, Q.; Li, L.; Fu, P. Exosomes from human umbilical cord mesenchymal stem cells inhibit ROS production and cell apoptosis in human articular chondrocytes via the miR-100-5p/NOX4 axis. Cell Biol. Int. 2021, 45, 2096–2106. [Google Scholar] [CrossRef] [PubMed]
  94. Song, X.J.; Zhang, L.; Li, Q.; Li, Y.; Ding, F.H.; Li, X. hUCB-MSC derived exosomal miR-124 promotes rat liver regeneration after partial hepatectomy via downregulating Foxg1. Life Sci. 2021, 265, 118821. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, X.L.; Zhao, Y.Y.; Sun, L.; Shi, Y.; Li, Z.Q.; Zhao, X.D.; Xu, C.G.; Ji, H.G.; Wang, M.; Xu, W.R.; et al. Exosomes derived from human umbilical cord mesenchymal stem cells improve myocardial repair via upregulation of Smad7. Int. J. Mol. Med. 2018, 41, 3063–3072. [Google Scholar] [CrossRef]
  96. 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] [PubMed]
  97. Huang, J.H.; Xu, Y.; Yin, X.M.; Lin, F.Y. Exosomes Derived from miR-126-modified MSCs Promote Angiogenesis and Neurogenesis and Attenuate Apoptosis after Spinal Cord Injury in Rats. Neuroscience 2020, 424, 133–145. [Google Scholar] [CrossRef] [PubMed]
  98. Zhu, W.; Sun, L.; Zhao, P.; Liu, Y.; Zhang, J.; Zhang, Y.; Hong, Y.; Zhu, Y.; Lu, Y.; Zhao, W.; et al. Macrophage migration inhibitory factor facilitates the therapeutic efficacy of mesenchymal stem cells derived exosomes in acute myocardial infarction through upregulating miR-133a-3p. J. Nanobiotechnology 2021, 19, 61. [Google Scholar] [CrossRef] [PubMed]
  99. Zhang, N.; Zhu, J.; Ma, Q.; Zhao, Y.; Wang, Y.; Hu, X.; Chen, J.; Zhu, W.; Han, Z.; Yu, H. Exosomes derived from human umbilical cord MSCs rejuvenate aged MSCs and enhance their functions for myocardial repair. Stem Cell Res. Ther. 2020, 11, 273. [Google Scholar] [CrossRef]
  100. Luo, Y.; Liang, F.; Wan, X.; Liu, S.; Fu, L.; Mo, J.; Meng, X.; Mo, Z. Hyaluronic Acid Facilitates Angiogenesis of Endothelial Colony Forming Cell Combining With Mesenchymal Stem Cell via CD44/ MicroRNA-139-5p Pathway. Front. Bioeng. Biotechnol. 2022, 10, 794037. [Google Scholar] [CrossRef]
  101. Huang, Y.; Chen, L.; Chen, D.; Fan, P.; Yu, H. Exosomal microRNA-140-3p from human umbilical cord mesenchymal stem cells attenuates joint injury of rats with rheumatoid arthritis by silencing SGK1. Mol. Med. 2022, 28, 36. [Google Scholar] [CrossRef]
  102. Geng, Y.; Chen, J.; Alahdal, M.; Chang, C.; Duan, L.; Zhu, W.; Mou, L.; Xiong, J.; Wang, M.; Wang, D. Intra-articular injection of hUC-MSCs expressing miR-140-5p induces cartilage self-repairing in the rat osteoarthritis. J. Bone Miner. Metab. 2020, 38, 277–288. [Google Scholar] [CrossRef] [PubMed]
  103. Diao, L.; Zhang, Q. Transfer of lncRNA UCA1 by hUCMSCs-derived exosomes protects against hypoxia/reoxygenation injury through impairing miR-143-targeted degradation of Bcl-2. Aging 2021, 13, 5967–5985. [Google Scholar] [CrossRef] [PubMed]
  104. Xiang, J.; Jiang, T.; Zhang, W.; Xie, W.; Tang, X.; Zhang, J. Human umbilical cord-derived mesenchymal stem cells enhanced HK-2 cell autophagy through MicroRNA-145 by inhibiting the PI3K/AKT/mTOR signaling pathway. Exp. Cell Res. 2019, 378, 198–205. [Google Scholar] [CrossRef]
  105. Zhang, Z.; Zou, X.; Zhang, R.; Xie, Y.; Feng, Z.; Li, F.; Han, J.; Sun, H.; Ouyang, Q.; Hua, S.; et al. Human umbilical cord mesenchymal stem cell-derived exosomal miR-146a-5p reduces microglial-mediated neuroinflammation via suppression of the IRAK1/TRAF6 signaling pathway after ischemic stroke. Aging 2021, 13, 3060–3079. [Google Scholar] [CrossRef]
  106. Lai, X.; Wang, Y.; Wang, X.; Liu, B.; Rong, L. miR-146a-5p-modified hUCMSC-derived exosomes facilitate spinal cord function recovery by targeting neurotoxic astrocytes. Stem Cell Res. Ther. 2022, 13, 487. [Google Scholar] [CrossRef] [PubMed]
  107. 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] [PubMed]
  108. Xiu, C.; Zheng, H.; Jiang, M.; Li, J.; Zhou, Y.; Mu, L.; Liu, W. MSCs-Derived miR-150-5p-Expressing Exosomes Promote Skin Wound Healing by Activating PI3K/AKT Pathway through PTEN. Int. J. Stem Cells 2022, 15, 359–371. [Google Scholar] [CrossRef]
  109. Nguyen, T.H.; Dao, H.H.; Duong, C.M.; Nguyen, X.H.; Hoang, D.H.; Do, X.H.; Truong, T.Q.; Nguyen, T.D.; Nguyen, L.T.; Than, U.T.T. Cytokine-primed umbilical cord mesenchymal stem cells enhanced therapeutic effects of extracellular vesicles on osteoarthritic chondrocytes. Front. Immunol. 2022, 13, 1041592. [Google Scholar] [CrossRef]
  110. Li, X.; Liu, L.; Yang, J.; Yu, Y.; Chai, J.; Wang, L.; Ma, L.; Yin, H. Exosome Derived From Human Umbilical Cord Mesenchymal Stem Cell Mediates MiR-181c Attenuating Burn-induced Excessive Inflammation. EBioMedicine 2016, 8, 72–82. [Google Scholar] [CrossRef]
  111. Zhang, Q.; Cao, L.; Zou, S.; Feng, Y.; Miao, X.; Huang, L. Human Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles Carrying MicroRNA-181c-5p Promote BMP2-Induced Repair of Cartilage Injury through Inhibition of SMAD7 Expression. Stem Cells Int. 2022, 2022, 1157498. [Google Scholar] [CrossRef] [PubMed]
  112. Zhao, H.; Li, Y.; Chen, L.; Shen, C.; Xiao, Z.; Xu, R.; Wang, J.; Luo, Y. HucMSCs-Derived miR-206-Knockdown Exosomes Contribute to Neuroprotection in Subarachnoid Hemorrhage Induced Early Brain Injury by Targeting BDNF. Neuroscience 2019, 417, 11–23. [Google Scholar] [CrossRef] [PubMed]
  113. Yang, M.; Liao, M.; Liu, R.; Zhang, Q.; Zhang, S.; He, Y.; Jin, J.; Zhang, P.; Zhou, L. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles loaded with miR-223 ameliorate myocardial infarction through P53/S100A9 axis. Genomics 2022, 114, 110319. [Google Scholar] [CrossRef] [PubMed]
  114. Zheng, Y.; Li, L. circPTP4A2-miR-330-5p-PDK2 Signaling Facilitates In Vivo Survival of HuMSCs on SF-SIS Scaffolds and Improves the Repair of Damaged Endometrium. Oxidative Med. Cell. Longev. 2022, 2022, 2818433. [Google Scholar] [CrossRef]
  115. Jiang, S.; Tian, G.; Yang, Z.; Gao, X.; Wang, F.; Li, J.; Tian, Z.; Huang, B.; Wei, F.; Sang, X.; et al. Enhancement of acellular cartilage matrix scaffold by Wharton’s jelly mesenchymal stem cell-derived exosomes to promote osteochondral regeneration. Bioact. Mater. 2021, 6, 2711–2728. [Google Scholar] [CrossRef] [PubMed]
  116. Wei, X.; Yi, X.; Lv, H. MicroRNA-377-3p released by mesenchymal stem cell exosomes ameliorates lipopolysaccharide-induced acute lung injury by targeting RPTOR to induce autophagy. Cell Death Diseas 2020, 11, 657. [Google Scholar] [CrossRef]
  117. Han, J.; Yang, S.; Hao, X.; Zhang, B.; Zhang, H.; Xin, C.; Hao, Y. Extracellular Vesicle-Derived microRNA-410 From Mesenchymal Stem Cells Protects Against Neonatal Hypoxia-Ischemia Brain Damage Through an HDAC1-Dependent EGR2/Bcl2 Axis. Front. Cell Dev. Biol. 2020, 8, 579236. [Google Scholar] [CrossRef]
  118. Yan, B.; Zhang, Y.; Liang, C.; Liu, B.; Ding, F.; Wang, Y.; Zhu, B.; Zhao, R.; Yu, X.Y.; Li, Y. Stem cell-derived exosomes prevent pyroptosis and repair ischemic muscle injury through a novel exosome/circHIPK3/ FOXO3a pathway. Theranostics 2020, 10, 6728–6742. [Google Scholar] [CrossRef]
  119. Liu, J.; Xing, F.; Fu, Q.; He, B.; Jia, Z.; Du, J.; Li, Y.; Zhang, X.; Chen, X. hUC-MSCs exosomal miR-451 alleviated acute lung injury by modulating macrophage M2 polarization via regulating MIF-PI3K-AKT signaling pathway. Environ. Toxicol. 2022, 37, 2819–2831. [Google Scholar] [CrossRef]
  120. Shao, M.; Xu, Q.; Wu, Z.; Chen, Y.; Shu, Y.; Cao, X.; Chen, M.; Zhang, B.; Zhou, Y.; Yao, R.; et al. Exosomes derived from human umbilical cord mesenchymal stem cells ameliorate IL-6-induced acute liver injury through miR-455-3p. Stem Cell Res. Ther. 2020, 11, 37. [Google Scholar] [CrossRef]
  121. Sun, D.; Jiang, Z.; Chen, Y.; Shang, D.; Miao, P.; Gao, J. MiR-455-5p upregulation in umbilical cord mesenchymal stem cells attenuates endometrial injury and promotes repair of damaged endometrium via Janus kinase/signal transducer and activator of transcription 3 signaling. Bioengineered 2021, 12, 12891–12904. [Google Scholar] [CrossRef] [PubMed]
  122. Ma, N.; Li, S.; Lin, C.; Cheng, X.; Meng, Z. Mesenchymal stem cell conditioned medium attenuates oxidative stress injury in hepatocytes partly by regulating the miR-486-5p/PIM1 axis and the TGF-β/Smad pathway. Bioengineered 2021, 12, 6434–6447. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, H.; Liu, S.; Zhang, W.; Liu, M.; Deng, C. Human Umbilical Cord Mesenchymal Stem Cell-Derived Exosome Repairs Endometrial Epithelial Cells Injury Induced by Hypoxia via Regulating miR-663a/CDKN2A Axis. Oxidative Med. Cell. Longev. 2022, 2022, 3082969. [Google Scholar] [CrossRef] [PubMed]
  124. Han, C.; Zhou, J.; Liu, B.; Liang, C.; Pan, X.; Zhang, Y.; Zhang, Y.; Wang, Y.; Shao, L.; Zhu, B.; et al. Delivery of miR-675 by stem cell-derived exosomes encapsulated in silk fibroin hydrogel prevents aging-induced vascular dysfunction in mouse hindlimb. Mater. Sci. Eng. C 2019, 99, 322–332. [Google Scholar] [CrossRef] [PubMed]
  125. Zhou, H.; Shen, X.; Yan, C.; Xiong, W.; Ma, Z.; Tan, Z.; Wang, J.; Li, Y.; Liu, J.; Duan, A.; et al. Extracellular vesicles derived from human umbilical cord mesenchymal stem cells alleviate osteoarthritis of the knee in mice model by interacting with METTL3 to reduce m6A of NLRP3 in macrophage. Stem Cell Res. Ther. 2022, 13, 322. [Google Scholar] [CrossRef] [PubMed]
  126. Xie, K.; Liu, L.; Chen, J.; Liu, F. Exosomes derived from human umbilical cord blood mesenchymal stem cells improve hepatic ischemia reperfusion injury via delivering miR-1246. Cell Cycle 2019, 18, 3491–3501. [Google Scholar] [CrossRef]
  127. Xie, K.; Liu, L.; Chen, J.; Liu, F. Exosomal miR-1246 derived from human umbilical cord blood mesenchymal stem cells attenuates hepatic ischemia reperfusion injury by modulating T helper 17/regulatory T balance. IUBMB Life 2019, 71, 2020–2030. [Google Scholar] [CrossRef]
  128. Yang, B.C.; Kuang, M.J.; Kang, J.Y.; Zhao, J.; Ma, J.X.; Ma, X.L. Human umbilical cord mesenchymal stem cell-derived exosomes act via the miR-1263/Mob1/Hippo signaling pathway to prevent apoptosis in disuse osteoporosis. Biochem. Biophys. Res. Commun. 2020, 524, 883–889. [Google Scholar] [CrossRef]
  129. Shi, Q.; Wang, D.; Ding, X.; Yang, X.; Zhang, Y. Exosome-shuttled miR-7162-3p from human umbilical cord derived mesenchymal stem cells repair endometrial stromal cell injury by restricting APOL6. Arch. Biochem. Biophys. 2021, 707, 108887. [Google Scholar] [CrossRef]
  130. Liu, J.S.; Du, J.; Cheng, X.; Zhang, X.Z.; Li, Y.; Chen, X.L. Exosomal miR-451 from human umbilical cord mesenchymal stem cells attenuates burn-induced acute lung injury. J. Chin. Med.Assoc. JCMA 2019, 82, 895–901. [Google Scholar] [CrossRef]
  131. Wang, Y.; Lai, X.; Wu, D.; Liu, B.; Wang, N.; Rong, L. Umbilical mesenchymal stem cell-derived exosomes facilitate spinal cord functional recovery through the miR-199a-3p/145-5p-mediated NGF/TrkA signaling pathway in rats. Stem Cell Res. Ther. 2021, 12, 117. [Google Scholar] [CrossRef]
  132. Li, W.; Jin, L.Y.; Cui, Y.B.; Xie, N. Human umbilical cord mesenchymal stem cells-derived exosomal microRNA-17-3p ameliorates inflammatory reaction and antioxidant injury of mice with diabetic retinopathy via targeting STAT1. Int. Immunopharmacol. 2021, 90, 107010. [Google Scholar] [CrossRef] [PubMed]
  133. Ding, C.; Zhu, L.; Shen, H.; Lu, J.; Zou, Q.; Huang, C.; Li, H.; Huang, B. Exosomal miRNA-17-5p derived from human umbilical cord mesenchymal stem cells improves ovarian function in premature ovarian insufficiency by regulating SIRT7. Stem Cells 2020, 38, 1137–1148. [Google Scholar] [CrossRef] [PubMed]
  134. Xu, Z.; Tian, N.; Li, S.; Li, K.; Guo, H.; Zhang, H.; Jin, H.; An, M.; Yu, X. Extracellular vesicles secreted from mesenchymal stem cells exert anti-apoptotic and anti-inflammatory effects via transmitting microRNA-18b in rats with diabetic retinopathy. Int. Immunopharmacol. 2021, 101, 108234. [Google Scholar] [CrossRef]
  135. Yang, Z.; Shan, N.; Deng, Q.; Wang, Y.; Hou, Y.; Mei, J.; Wu, Z. Extracellular vesicle-derived microRNA-18b ameliorates preeclampsia by enhancing trophoblast proliferation and migration via Notch2/TIM3/mTORC1 axis. J. Cell. Mol. Med. 2021, 25, 4583–4595. [Google Scholar] [CrossRef] [PubMed]
  136. Shi, L.; Ren, J.; Li, J.; Wang, D.; Wang, Y.; Qin, T.; Li, X.; Zhang, G.; Li, C.; Wang, Y. Extracellular vesicles derived from umbilical cord mesenchymal stromal cells alleviate pulmonary fibrosis by means of transforming growth factor-β signaling inhibition. Stem Cell Res. Ther. 2021, 12, 230. [Google Scholar] [CrossRef]
  137. Cai, J.H.; Sun, Y.T.; Bao, S. HucMSCs-exosomes containing miR-21 promoted estrogen production in ovarian granulosa cells via LATS1-mediated phosphorylation of LOXL2 and YAP. Gen. Comp. Endocrinol. 2022, 321–322, 114015. [Google Scholar] [CrossRef]
  138. Chen, J.; Chen, J.; Cheng, Y.; Fu, Y.; Zhao, H.; Tang, M.; Zhao, H.; Lin, N.; Shi, X.; Lei, Y.; et al. Mesenchymal stem cell-derived exosomes protect beta cells against hypoxia-induced apoptosis via miR-21 by alleviating ER stress and inhibiting p38 MAPK phosphorylation. Stem Cell Res. Ther. 2020, 11, 97. [Google Scholar] [CrossRef]
  139. Du, X.; Li, H.; Han, X.; Ma, W. Mesenchymal stem cells-derived exosomal miR-24-3p ameliorates non-alcohol fatty liver disease by targeting Keap-1. Biochem. Biophys. Res. Commun. 2022, 637, 331–340. [Google Scholar] [CrossRef]
  140. Yuan, X.; Li, T.; Shi, L.; Miao, J.; Guo, Y.; Chen, Y. Human umbilical cord mesenchymal stem cells deliver exogenous miR-26a-5p via exosomes to inhibit nucleus pulposus cell pyroptosis through METTL14/NLRP3. Mol. Med. 2021, 27, 91. [Google Scholar] [CrossRef]
  141. Li, D.; Zhang, J.; Liu, Z.; Gong, Y.; Zheng, Z. Human umbilical cord mesenchymal stem cell-derived exosomal miR-27b attenuates subretinal fibrosis via suppressing epithelial-mesenchymal transition by targeting HOXC6. Stem Cell Res. Ther. 2021, 12, 24. [Google Scholar] [CrossRef]
  142. Gao, T.; Cao, Y. Human Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles Carrying MicroRNA-29a Improves Ovarian Function of Mice with Primary Ovarian Insufficiency by Targeting HMG-Box Transcription Factor/Wnt/β-Catenin Signaling. Dis. Markers 2022, 2022, 5045873. [Google Scholar] [CrossRef] [PubMed]
  143. Bier, A.; Berenstein, P.; Kronfeld, N.; Morgoulis, D.; Ziv-Av, A.; Goldstein, H.; Kazimirsky, G.; Cazacu, S.; Meir, R.; Popovtzer, R.; et al. Placenta-derived mesenchymal stromal cells and their exosomes exert therapeutic effects in Duchenne muscular dystrophy. Biomaterials 2018, 174, 67–78. [Google Scholar] [CrossRef] [PubMed]
  144. He, Y.; Zhang, Z.; Yao, T.; Huang, L.; Gan, J.; Lv, H.; Chen, J. Extracellular vesicles derived from human umbilical cord mesenchymal stem cells relieves diabetic retinopathy through a microRNA-30c-5p-dependent mechanism. Diabetes Res. Clin. Pract. 2022, 190, 109861. [Google Scholar] [CrossRef] [PubMed]
  145. Zhang, F.; Li, F.; Lu, J. microRNA-100 shuttled by human umbilical cord MSC-secreted extracellular vesicles induces endometriosis by inhibiting HS3ST2. Cell. Signal. 2023, 102, 110532. [Google Scholar] [CrossRef] [PubMed]
  146. Niu, J.; Yu, F.; Luo, X.; Chen, S. Human Umbilical Cord Mesenchymal Stem Cells Improve Premature Ovarian Failure through Cell Apoptosis of miR-100-5p/NOX4/NLRP3. BioMed Res. Int. 2022, 2022, 3862122. [Google Scholar] [CrossRef]
  147. Li, N.; Gao, Z.; Zhao, L.; Du, B.; Ma, B.; Nian, H.; Wei, R. MSC-Derived Small Extracellular Vesicles Attenuate Autoimmune Dacryoadenitis by Promoting M2 Macrophage Polarization and Inducing Tregs via miR-100-5p. Front. Immunol. 2022, 13, 888949. [Google Scholar] [CrossRef]
  148. Zhong, Z.; Tian, Y.; Luo, X.; Zou, J.; Wu, L.; Tian, J. Extracellular Vesicles Derived From Human Umbilical Cord Mesenchymal Stem Cells Protect Against DOX-Induced Heart Failure Through the miR-100-5p/NOX4 Pathway. Front. Bioeng. Biotechnol. 2021, 9, 703241. [Google Scholar] [CrossRef]
  149. Gao, H.; Yu, Z.; Li, Y.; Wang, X. miR-100-5p in human umbilical cord mesenchymal stem cell-derived exosomes mediates eosinophilic inflammation to alleviate atherosclerosis via the FZD5/Wnt/β-catenin pathway. Acta Biochim. Biophys. Sin. 2021, 53, 1166–1176. [Google Scholar] [CrossRef]
  150. Cui, J.; Chen, X.; Lin, S.; Li, L.; Fan, J.; Hou, H.; Li, P. MiR-101-containing extracellular vesicles bind to BRD4 and enhance proliferation and migration of trophoblasts in preeclampsia. Stem Cell Res. Ther. 2020, 11, 231. [Google Scholar] [CrossRef]
  151. Oh, S.J.; Lee, E.N.; Park, J.H.; Lee, J.K.; Cho, G.J.; Park, I.H.; Shin, O.S. Anti-Viral Activities of Umbilical Cord Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Against Human Respiratory Viruses. Front. Cell. Infect. Microbiol. 2022, 12, 850744. [Google Scholar] [CrossRef] [PubMed]
  152. Zhang, W.; Wang, Y.; Kong, Y. Exosomes Derived From Mesenchymal Stem Cells Modulate miR-126 to Ameliorate Hyperglycemia-Induced Retinal Inflammation Via Targeting HMGB1. Investig. Ophthalmol. Vis. Sci. 2019, 60, 294–303. [Google Scholar] [CrossRef] [PubMed]
  153. Qu, Q.; Liu, L. miR-126-3p containing exosomes derived from human umbilical cord mesenchymal stem cells promote angiogenesis and attenuate ovarian granulosa cell apoptosis in a preclinical rat model of premature ovarian failure. Stem Cell Res. Ther. 2022, 13, 352. [Google Scholar] [CrossRef] [PubMed]
  154. Chen, Z.; Zhou, S. SDF-1α Facilitates Mesenchymal Stem Cells to Induce Regulatory B Cell Differentiation from Patients with Immune Thrombocytopenia. Stem Cells Int. 2021, 2021, 3254488. [Google Scholar] [CrossRef]
  155. Wang, D.; Na, Q.; Song, G.Y.; Wang, L. Human umbilical cord mesenchymal stem cell-derived exosome-mediated transfer of microRNA-133b boosts trophoblast cell proliferation, migration and invasion in preeclampsia by restricting SGK1. Cell Cycle 2020, 19, 1869–1883. [Google Scholar] [CrossRef]
  156. Jiang, Y.; Luo, T.; Xia, Q.; Tian, J.; Yang, J. microRNA-140-5p from human umbilical cord mesenchymal stem cells-released exosomes suppresses preeclampsia development. Funct. Integr. Genom. 2022, 22, 813–824. [Google Scholar] [CrossRef]
  157. Kwak, J.; Choi, S.J.; Oh, W.; Yang, Y.S.; Jeon, H.B. Cobalt Chloride Enhances the Anti-Inflammatory Potency of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells through the ERK-HIF-1α-MicroRNA-146a-Mediated Signaling Pathway. Stem Cells Int. 2018, 2018, 4978763. [Google Scholar] [CrossRef]
  158. Wang, J.; Pei, B.; Yan, J.; Xu, X.; Fang, A.N.; Ocansey, D.K.W.; Zhang, X.; Qian, H. hucMSC-Derived Exosomes Alleviate the Deterioration of Colitis via the miR-146a/SUMO1 Axis. Mol. Pharm. 2022, 19, 484–493. [Google Scholar] [CrossRef]
  159. Liang, Y.C.; Wu, Y.P.; Li, X.D.; Chen, S.H.; Ye, X.J.; Xue, X.Y. TNF-α-induced exosomal miR-146a mediates mesenchymal stem cell-dependent suppression of urethral stricture. J. Cell. Physiol. 2019, 234, 23243–23255. [Google Scholar] [CrossRef]
  160. Song, Y.; Dou, H.; Li, X.; Zhao, X.; Li, Y.; Liu, D.; Ji, J.; Liu, F.; Ding, L.; Ni, Y.; et al. Exosomal miR-146a Contributes to the Enhanced Therapeutic Efficacy of Interleukin-1β-Primed Mesenchymal Stem Cells Against Sepsis. Stem Cells 2017, 35, 1208–1221. [Google Scholar] [CrossRef]
  161. Hua, T.; Yang, M.; Song, H.; Kong, E.; Deng, M.; Li, Y.; Li, J.; Liu, Z.; Fu, H.; Wang, Y.; et al. Huc-MSCs-derived exosomes attenuate inflammatory pain by regulating microglia pyroptosis and autophagy via the miR-146a-5p/TRAF6 axis. J. Nanobiotechnology 2022, 20, 324. [Google Scholar] [CrossRef]
  162. Chen, X.; Su, C.; Wei, Q.; Sun, H.; Xie, J.; Nong, G. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Alleviate Diffuse Alveolar Hemorrhage Associated with Systemic Lupus Erythematosus in Mice by Promoting M2 Macrophage Polarization via the microRNA-146a-5p/NOTCH1 Axis. Immunol. Investig. 2022, 51, 1975–1993. [Google Scholar] [CrossRef] [PubMed]
  163. Zhang, Y.; Le, X.; Zheng, S.; Zhang, K.; He, J.; Liu, M.; Tu, C.; Rao, W.; Du, H.; Ouyang, Y.; et al. MicroRNA-146a-5p-modified human umbilical cord mesenchymal stem cells enhance protection against diabetic nephropathy in rats through facilitating M2 macrophage polarization. Stem Cell Res. Ther. 2022, 13, 171. [Google Scholar] [CrossRef]
  164. Dong, L.; Wang, Y.; Zheng, T.; Pu, Y.; Ma, Y.; Qi, X.; Zhang, W.; Xue, F.; Shan, Z.; Liu, J.; et al. Hypoxic hUCMSC-derived extracellular vesicles attenuate allergic airway inflammation and airway remodeling in chronic asthma mice. Stem Cell Res. Ther. 2021, 12, 4. [Google Scholar] [CrossRef] [PubMed]
  165. Spinosa, M.; Lu, G.; Su, G.; Bontha, S.V.; Gehrau, R.; Salmon, M.D.; Smith, J.R.; Weiss, M.L.; Mas, V.R.; Upchurch, G.R., Jr.; et al. Human mesenchymal stromal cell-derived extracellular vesicles attenuate aortic aneurysm formation and macrophage activation via microRNA-147. FASEBJ. Off. Publ. Fed. Am. Soc. Exp. Biol. 2018, 32, fj201701138RR. [Google Scholar] [CrossRef] [PubMed]
  166. Zhou, Q.; Rong, C.; Gu, T.; Li, H.; Wu, L.; Zhuansun, X.; Zhao, X.; Xiao, Z.; Kuang, Y.; Xu, S.; et al. Mesenchymal stem cells improve liver fibrosis and protect hepatocytes by promoting microRNA-148a-5p-mediated inhibition of Notch signaling pathway. Stem Cell Res. Ther. 2022, 13, 354. [Google Scholar] [CrossRef]
  167. Li, D.; Lu, Z.; Li, X.; Xu, Z.; Jiang, J.; Zheng, Z.; Jia, J.; Lin, S. Human umbilical cord mesenchymal stem cells facilitate the up-regulation of miR-153-3p, whereby attenuating MGO-induced peritoneal fibrosis in rats. J. Cell. Mol. Med. 2018, 22, 3452–3463. [Google Scholar] [CrossRef]
  168. Li, D.; Li, X.; Duan, M.; Dou, Y.; Feng, Y.; Nan, N.; Zhang, W. MiR-153-3p induces immune dysregulation by inhibiting PELI1 expression in umbilical cord-derived mesenchymal stem cells in patients with systemic lupus erythematosus. Autoimmunity 2020, 53, 201–209. [Google Scholar] [CrossRef]
  169. Zheng, B.; Zhang, P.; Yuan, L.; Chhetri, R.K.; Guo, Y.; Deng, D. Effects of human umbilical cord mesenchymal stem cells on inflammatory factors and miR-181a in T lymphocytes from patients with systemic lupus erythematosus. Lupus 2020, 29, 126–135. [Google Scholar] [CrossRef]
  170. Zhou, C.; Wan, S.; Zhao, X.; Gu, S.; Pei, J.; Wu, Y.; Han, Z.; Che, R.; Hua, X. Exosomal miR-195 in hUC-MSCs alleviates hypoxia-induced damage of trophoblast cells through tissue factor pathway inhibitor 2. Curr. Res. Transl. Med. 2022, 70, 103352. [Google Scholar] [CrossRef]
  171. Bi, Z.M.; Zhou, Q.F.; Geng, Y.; Zhang, H.M. Human umbilical cord mesenchymal stem cells ameliorate experimental cirrhosis through activation of keratinocyte growth factor by suppressing microRNA-199. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 4905–4912. [Google Scholar] [PubMed]
  172. Cheng, T.; Ding, S.; Liu, S.; Li, Y.; Sun, L. Human umbilical cord-derived mesenchymal stem cell therapy ameliorates lupus through increasing CD4+ T cell senescence via MiR-199a-5p/Sirt1/p53 axis. Theranostics 2021, 11, 893–905. [Google Scholar] [CrossRef] [PubMed]
  173. Xu, Y.; Tang, X.; Fang, A.; Yan, J.; Kofi Wiredu Ocansey, D.; Zhang, X.; Mao, F. HucMSC-Ex carrying miR-203a-3p.2 ameliorates colitis through the suppression of caspase11/4-induced macrophage pyroptosis. Int. Immunopharmacol. 2022, 110, 108925. [Google Scholar] [CrossRef]
  174. Lee, C.; Mitsialis, S.A.; Aslam, M.; Vitali, S.H.; Vergadi, E.; Konstantinou, G.; Sdrimas, K.; Fernandez-Gonzalez, A.; Kourembanas, S. Exosomes mediate the cytoprotective action of mesenchymal stromal cells on hypoxia-induced pulmonary hypertension. Circulation 2012, 126, 2601–2611. [Google Scholar] [CrossRef] [PubMed]
  175. Zhang, Y.; Deng, W.; Wang, W.; Song, A.; Mukama, O.; Deng, S.; Han, X.; De Dieu Habimana, J.; Peng, K.; Ni, B.; et al. MicroRNA-206 down-regulated human umbilical cord mesenchymal stem cells alleviate cognitive decline in D-galactose-induced aging mice. Cell Death Discov. 2022, 8, 304. [Google Scholar] [CrossRef] [PubMed]
  176. Zhang, W.; Lin, J.; Shi, P.; Su, D.; Cheng, X.; Yi, W.; Yan, J.; Chen, H.; Cheng, F. Small Extracellular Vesicles Derived From MSCs Have Immunomodulatory Effects to Enhance Delivery of ASO-210 for Psoriasis Treatment. Front. Cell Dev. Biol. 2022, 10, 842813. [Google Scholar] [CrossRef] [PubMed]
  177. Liu, W.; Zhou, N.; Liu, Y.; Zhang, W.; Li, X.; Wang, Y.; Zheng, R.; Zhang, Y. Mesenchymal stem cell exosome-derived miR-223 alleviates acute graft-versus-host disease via reducing the migration of donor T cells. Stem Cell Res. Ther. 2021, 12, 153. [Google Scholar] [CrossRef]
  178. Liu, L.; Yin, H.; Hao, X.; Song, H.; Chai, J.; Duan, H.; Chang, Y.; Yang, L.; Wu, Y.; Han, S.; et al. Down-Regulation of miR-301a-3p Reduces Burn-Induced Vascular Endothelial Apoptosis by potentiating hMSC-Secreted IGF-1 and PI3K/Akt/FOXO3a Pathway. iScience 2020, 23, 101383. [Google Scholar] [CrossRef]
  179. Zhang, L.; Yuan, J.; Kofi Wiredu Ocansey, D.; Lu, B.; Wan, A.; Chen, X.; Zhang, X.; Qiu, W.; Mao, F. Exosomes derived from human umbilical cord mesenchymal stem cells regulate lymphangiogenesis via the miR-302d-3p/VEGFR3/AKT axis to ameliorate inflammatory bowel disease. Int. Immunopharmacol. 2022, 110, 109066. [Google Scholar] [CrossRef]
  180. Wang, G.; Yuan, J.; Cai, X.; Xu, Z.; Wang, J.; Ocansey, D.K.W.; Yan, Y.; Qian, H.; Zhang, X.; Xu, W.; et al. HucMSC-exosomes carrying miR-326 inhibit neddylation to relieve inflammatory bowel disease in mice. Clin. Transl. Med. 2020, 10, e113. [Google Scholar] [CrossRef]
  181. Qiu, Z.; Zhong, Z.; Zhang, Y.; Tan, H.; Deng, B.; Meng, G. Human umbilical cord mesenchymal stem cell-derived exosomal miR-335-5p attenuates the inflammation and tubular epithelial-myofibroblast transdifferentiation of renal tubular epithelial cells by reducing ADAM19 protein levels. Stem Cell Res. Ther. 2022, 13, 373. [Google Scholar] [CrossRef] [PubMed]
  182. Pan, Z.; Chen, Q.; Ding, H.; Li, H. MicroRNA-342-3p loaded by human umbilical cord mesenchymal stem cells-derived exosomes attenuates deep vein thrombosis by downregulating EDNRA. J. Thromb. Thrombolysis 2022, 54, 411–419. [Google Scholar] [CrossRef] [PubMed]
  183. Chen, W.; Zhang, F.; Hou, X.; Xu, H.; Tang, D. Ameliorating role of microRNA-378 carried by umbilical cord mesenchymal stem cells-released extracellular vesicles in mesangial proliferative glomerulonephritis. Cell Commun. Signal. CCS 2022, 20, 28. [Google Scholar] [CrossRef] [PubMed]
  184. Cai, X.; Zhang, Z.Y.; Yuan, J.T.; Ocansey, D.K.W.; Tu, Q.; Zhang, X.; Qian, H.; Xu, W.R.; Qiu, W.; Mao, F. hucMSC-derived exosomes attenuate colitis by regulating macrophage pyroptosis via the miR-378a-5p/NLRP3 axis. Stem Cell Res. Ther. 2021, 12, 416. [Google Scholar] [CrossRef]
  185. Zhou, Q.; Gu, T. Human Umbilical Cord Mesenchymal Stem Cells Ameliorate Hepatic Stellate Cell Activation and Liver Fibrosis by Upregulating MicroRNA-455-3p through Suppression of p21-Activated Kinase-2. BioMed Res. Int. 2021, 2021, 6685605. [Google Scholar] [CrossRef] [PubMed]
  186. Park, S.E.; Jeong, J.B.; Oh, S.J. Wharton’s Jelly-Derived Mesenchymal Stem Cells Reduce Fibrosis in a Mouse Model of Duchenne Muscular Dystrophy by Upregulating microRNA 499. Biomedicines 2021, 9, 1089. [Google Scholar] [CrossRef]
  187. Cheng, L.; Yu, P.; Li, F.; Jiang, X.; Jiao, X.; Shen, Y. Human umbilical cord-derived mesenchymal stem cell-exosomal miR-627-5p ameliorates non-alcoholic fatty liver disease by repressing FTO expression. Hum. Cell 2021, 34, 1697–1708. [Google Scholar] [CrossRef]
  188. Wang, Z.; Gao, D.; Wang, S.; Lin, H.; Wang, Y.; Xu, W. Exosomal microRNA-1246 from human umbilical cord mesenchymal stem cells potentiates myocardial angiogenesis in chronic heart failure. Cell Biol. Int. 2021, 45, 2211–2225. [Google Scholar] [CrossRef] [PubMed]
  189. Zhang, X.; Zhou, Y.; Ye, Y.; Wu, R.; Li, W.; Yao, C.; Wang, S. Human umbilical cord mesenchymal stem cell-derived exosomal microRNA-148a-3p inhibits neointimal hyperplasia by targeting Serpine1. Arch. Biochem. Biophys. 2022, 719, 109155. [Google Scholar] [CrossRef]
  190. Zhang, Y.; Pan, Y.; Liu, Y.; Li, X.; Tang, L.; Duan, M.; Li, J.; Zhang, G. Exosomes derived from human umbilical cord blood mesenchymal stem cells stimulate regenerative wound healing via transforming growth factor-β receptor inhibition. Stem Cell Res. Ther. 2021, 12, 434. [Google Scholar] [CrossRef]
  191. Zhang, X.-F.; Wang, T.; Wang, Z.-X.; Huang, K.-P.; Zhang, Y.-W.; Wang, G.-L.; Zhang, H.-J.; Chen, Z.-H.; Wang, C.-Y.; Zhang, J.-X.; et al. Hypoxic ucMSC-secreted exosomal miR-125b promotes endothelial cell survival and migration during wound healing by targeting TP53INP1. Mol. Ther.-Nucleic Acids 2021, 26, 347–359. [Google Scholar] [CrossRef] [PubMed]
  192. Li, K.; Yan, G.; Huang, H.; Zheng, M.; Ma, K.; Cui, X.; Lu, D.; Zheng, L.; Zhu, B.; Cheng, J.; et al. Anti-inflammatory and immunomodulatory effects of the extracellular vesicles derived from human umbilical cord mesenchymal stem cells on osteoarthritis via M2 macrophages. J. Nanobiotechnology 2022, 20, 38. [Google Scholar] [CrossRef] [PubMed]
  193. Chen, P.; Tang, S.; Gao, H.; Zhang, H.; Chen, C.; Fang, Z.; Peng, G.; Weng, H.; Chen, A.; Zhang, C.; et al. Wharton’s jelly mesenchymal stem cell-derived small extracellular vesicles as natural nanoparticles to attenuate cartilage injury via microRNA regulation. Int. J. Pharm. 2022, 623, 121952. [Google Scholar] [CrossRef] [PubMed]
  194. Du, L.; Tao, X.; Shen, X. Human umbilical cord mesenchymal stem cell-derived exosomes inhibit migration and invasion of breast cancer cells via miR-21-5p/ZNF367 pathway. Breast Cancer 2021, 28, 829–837. [Google Scholar] [CrossRef] [PubMed]
  195. Yuan, L.; Liu, Y.; Qu, Y.; Liu, L.; Li, H. Exosomes Derived From MicroRNA-148b-3p-Overexpressing Human Umbilical Cord Mesenchymal Stem Cells Restrain Breast Cancer Progression. Front. Oncol. 2019, 9, 1076. [Google Scholar] [CrossRef] [PubMed]
  196. Khazaei-Poul, Y.; Shojaei, S.; Koochaki, A.; Ghanbarian, H.; Mohammadi-Yeganeh, S. Evaluating the influence of Human Umbilical Cord Mesenchymal Stem Cells-derived exosomes loaded with miR-3182 on metastatic performance of Triple Negative Breast Cancer cells. Life Sci. 2021, 286, 120015. [Google Scholar] [CrossRef]
  197. Khazaei-Poul, Y.; Mirmotalebisohi, S.A.; Zali, H.; Molavi, Z.; Mohammadi-Yeganeh, S. Identification of miR-3182 and miR-3143 target genes involved in the cell cycle as a novel approach in TNBC treatment: A systems biology approach. Chem. Biol. Drug Des. 2023, 101, 662–677. [Google Scholar] [CrossRef]
  198. Wang, Y.; Wang, P.; Zhao, L.; Chen, X.; Lin, Z.; Zhang, L.; Li, Z. miR-224-5p Carried by Human Umbilical Cord Mesenchymal Stem Cells-Derived Exosomes Regulates Autophagy in Breast Cancer Cells via HOXA5. Front. Cell Dev. Biol. 2021, 9, 679185. [Google Scholar] [CrossRef]
  199. Pan, Y.; Wang, X.; Li, Y.; Yan, P.; Zhang, H. Human umbilical cord blood mesenchymal stem cells-derived exosomal microRNA-503-3p inhibits progression of human endometrial cancer cells through downregulating MEST. Cancer Gene Ther. 2022, 29, 1130–1139. [Google Scholar] [CrossRef]
  200. Li, X.; Liu, L.L.; Yao, J.L.; Wang, K. Human Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles Inhibit Endometrial Cancer Cell Proliferation and Migration through Delivery of Exogenous miR-302a. Stem Cells Int. 2019, 2019, 8108576. [Google Scholar] [CrossRef]
  201. Zhang, Y.; Wang, J.; Wu, D.; Li, M.; Zhao, F.; Ren, M.; Cai, Y.; Dou, J. IL-21-secreting hUCMSCs combined with miR-200c inhibit tumor growth and metastasis via repression of Wnt/β-catenin signaling and epithelial-mesenchymal transition in epithelial ovarian cancer. OncoTargets Ther. 2018, 11, 2037–2050. [Google Scholar] [CrossRef] [PubMed]
  202. Qiu, L.; Wang, J.; Chen, M.; Chen, F.; Tu, W. Exosomal microRNA-146a derived from mesenchymal stem cells increases the sensitivity of ovarian cancer cells to docetaxel and taxane via a LAMC2-mediated PI3K/Akt axis. Int. J. Mol. Med. 2020, 46, 609–620. [Google Scholar] [CrossRef] [PubMed]
  203. Lee, H.K.; Finniss, S.; Cazacu, S.; Bucris, E.; Ziv-Av, A.; Xiang, C.; Bobbitt, K.; Rempel, S.A.; Hasselbach, L.; Mikkelsen, T.; et al. Mesenchymal stem cells deliver synthetic microRNA mimics to glioma cells and glioma stem cells and inhibit their cell migration and self-renewal. Oncotarget 2013, 4, 346–361. [Google Scholar] [CrossRef] [PubMed]
  204. Sharif, S.; Ghahremani, M.H.; Soleimani, M. Delivery of Exogenous miR-124 to Glioblastoma Multiform Cells by Wharton’s Jelly Mesenchymal Stem Cells Decreases Cell Proliferation and Migration, and Confers Chemosensitivity. Stem Cell Rev. Rep. 2018, 14, 236–246. [Google Scholar] [CrossRef]
  205. Hao, S.C.; Ma, H.; Niu, Z.F.; Sun, S.Y.; Zou, Y.R.; Xia, H.C. hUC-MSCs secreted exosomes inhibit the glioma cell progression through PTENP1/miR-10a-5p/PTEN pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 10013–10023. [Google Scholar] [CrossRef]
  206. Li, N.; Wang, B. Suppressive effects of umbilical cord mesenchymal stem cell-derived exosomal miR-15a-5p on the progression of cholangiocarcinoma by inhibiting CHEK1 expression. Cell Death Discov. 2022, 8, 205. [Google Scholar] [CrossRef]
  207. Ma, H.; Weng, F.; Wang, L.; Tong, X.; Yao, Y.; Li, H. Extracellular vesicle-mediated delivery of miR-127-3p inhibits the proliferation and invasion of choriocarcinoma cells by targeting ITGA6. Exp. Cell Res. 2022, 414, 113098. [Google Scholar] [CrossRef]
  208. Xie, H.; Wang, J. MicroRNA-320a-containing exosomes from human umbilical cord mesenchymal stem cells curtail proliferation and metastasis in lung cancer by binding to SOX4. J. Recept. Signal Transduct. Res. 2022, 42, 268–278. [Google Scholar] [CrossRef]
  209. Dong, L.; Pu, Y.; Zhang, L.; Qi, Q.; Xu, L.; Li, W.; Wei, C.; Wang, X.; Zhou, S.; Zhu, J.; et al. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles promote lung adenocarcinoma growth by transferring miR-410. Cell Death Dis. 2018, 9, 218. [Google Scholar] [CrossRef]
  210. Liu, J.; Zhu, M.; Tang, Q. Human umbilical cord mesenchymal stem cells-derived exosomal microRNA-181a retards nasopharyngeal carcinoma development by mediating KDM5C. J. Cancer Res. Clin. Oncol. 2021, 147, 2867–2877. [Google Scholar] [CrossRef]
  211. Chen, M.; Xia, Z.; Deng, J. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles carrying miR-655-3p inhibit the development of esophageal cancer by regulating the expression of HIF-1α via a LMO4/HDAC2-dependent mechanism. Cell Biol. Toxicol. 2022. [Google Scholar] [CrossRef]
  212. He, Z.; Li, W.; Zheng, T.; Liu, D.; Zhao, S. Human umbilical cord mesenchymal stem cells-derived exosomes deliver microRNA-375 to downregulate ENAH and thus retard esophageal squamous cell carcinoma progression. J. Exp. Clin. CancerRes. CR 2020, 39, 140. [Google Scholar] [CrossRef]
  213. Qu, M.; Li, J.; Hong, Z.; Jia, F.; He, Y.; Yuan, L. The role of human umbilical cord mesenchymal stem cells-derived exosomal microRNA-431-5p in survival and prognosis of colorectal cancer patients. Mutagenesis 2022, 37, 164–171. [Google Scholar] [CrossRef] [PubMed]
  214. Chen, Z.; Xie, Y.; Chen, W.; Li, T.; Chen, X.; Liu, B. microRNA-6785-5p-loaded human umbilical cord mesenchymal stem cells-derived exosomes suppress angiogenesis and metastasis in gastric cancer via INHBA. Life Sci. 2021, 284, 119222. [Google Scholar] [CrossRef] [PubMed]
  215. Jia, Y.; Ding, X.; Zhou, L.; Zhang, L.; Yang, X. Mesenchymal stem cells-derived exosomal microRNA-139-5p restrains tumorigenesis in bladder cancer by targeting PRC1. Oncogene 2021, 40, 246–261. [Google Scholar] [CrossRef]
  216. Zheng, T.; Zhou, Y.; Xu, X.; Qi, X.; Liu, J.; Pu, Y.; Zhang, S.; Gao, X.; Luo, X.; Li, M.; et al. MiR-30c-5p loss-induced PELI1 accumulation regulates cell proliferation and migration via activating PI3K/AKT pathway in papillary thyroid carcinoma. J. Transl. Med. 2022, 20, 20. [Google Scholar] [CrossRef]
  217. Gan, J.; Liu, S.; Zhang, Y.; He, L.; Bai, L.; Liao, R.; Zhao, J.; Guo, M.; Jiang, W.; Li, J.; et al. MicroRNA-375 is a therapeutic target for castration-resistant prostate cancer through the PTPN4/STAT3 axis. Exp. Mol. Med. 2022, 54, 1290–1305. [Google Scholar] [CrossRef] [PubMed]
  218. Xu, Y.; Lai, Y.; Cao, L.; Li, Y.; Chen, G.; Chen, L.; Weng, H.; Chen, T.; Wang, L.; Ye, Y. Human umbilical cord mesenchymal stem cells-derived exosomal microRNA-451a represses epithelial-mesenchymal transition of hepatocellular carcinoma cells by inhibiting ADAM10. RNA Biol. 2021, 18, 1408–1423. [Google Scholar] [CrossRef]
  219. Ding, Y.; Cao, F.; Sun, H.; Wang, Y.; Liu, S.; Wu, Y.; Cui, Q.; Mei, W.; Li, F. Exosomes derived from human umbilical cord mesenchymal stromal cells deliver exogenous miR-145-5p to inhibit pancreatic ductal adenocarcinoma progression. Cancer Lett. 2019, 442, 351–361. [Google Scholar] [CrossRef] [PubMed]
  220. Huang, H.; Zhong, P.; Zhang, J.; Chen, X.; Chen, J.; Lin, T.; Wu, Q. Human umbilical cord-mesenchymal stem cells-derived exosomes carrying microRNA-15a-5p possess therapeutic effects on Wilms tumor via regulating septin 2. Bioengineered 2022, 13, 6136–6149. [Google Scholar] [CrossRef]
  221. Chen, X.; Chen, Y.; Zhang, M.; Cheng, H.; Mai, H.; Yi, M.; Xu, H.; Yuan, X.; Liu, S.; Wen, F. HucMSC exosomes promoted imatinib-induced apoptosis in K562-R cells via a miR-145a-5p/USP6/GLS1 axis. Cell Death Dis. 2022, 13, 92. [Google Scholar] [CrossRef] [PubMed]
  222. Ding, Y.; Mei, W.; Zheng, Z.; Cao, F.; Liang, K.; Jia, Y.; Wang, Y.; Liu, D.; Li, J.; Li, F. Exosomes secreted from human umbilical cord mesenchymal stem cells promote pancreatic ductal adenocarcinoma growth by transferring miR-100-5p. Tissue Cell 2021, 73, 101623. [Google Scholar] [CrossRef] [PubMed]
  223. Chang, Y.H.; Vuong, C.K.; Ngo, N.H.; Yamashita, T.; Ye, X.; Futamura, Y.; Fukushige, M.; Obata-Yasuoka, M.; Hamada, H.; Osaka, M.; et al. Extracellular vesicles derived from Wharton’s Jelly mesenchymal stem cells inhibit the tumor environment via the miR-125b/HIF1α signaling pathway. Sci. Rep. 2022, 12, 13550. [Google Scholar] [CrossRef] [PubMed]
  224. Li, H.; Huang, H.; Chen, X.; Chen, S.; Yu, L.; Wang, C.; Liu, Y.; Zhang, K.; Wu, L.; Han, Z.C.; et al. The delivery of hsa-miR-11401 by extracellular vesicles can relieve doxorubicin-induced mesenchymal stem cell apoptosis. Stem Cell Res. Ther. 2021, 12, 77. [Google Scholar] [CrossRef]
  225. Joerger-Messerli, M.S.; Oppliger, B.; Spinelli, M.; Thomi, G.; di Salvo, I.; Schneider, P.; Schoeberlein, A. Extracellular Vesicles Derived from Wharton’s Jelly Mesenchymal Stem Cells Prevent and Resolve Programmed Cell Death Mediated by Perinatal Hypoxia-Ischemia in Neuronal Cells. Cell Transplant. 2018, 27, 168–180. [Google Scholar] [CrossRef] [PubMed]
  226. Lu, C.H.; Chen, Y.A. Multiplexed Molecular Imaging Strategy Integrated with RNA Sequencing in the Assessment of the Therapeutic Effect of Wharton’s Jelly Mesenchymal Stem Cell-Derived Extracellular Vesicles for Osteoporosis. Int. J. Nanomed. 2021, 16, 7813–7830. [Google Scholar] [CrossRef]
  227. Zhu, Z.; Zhang, Y.; Zhang, Y.; Zhang, H.; Liu, W.; Zhang, N.; Zhang, X.; Zhou, G.; Wu, L.; Hua, K.; et al. Exosomes derived from human umbilical cord mesenchymal stem cells accelerate growth of VK2 vaginal epithelial cells through MicroRNAs in vitro. Hum. Reprod. 2019, 34, 248–260. [Google Scholar] [CrossRef]
  228. Yang, K.; Li, D.; Wang, M.; Xu, Z.; Chen, X.; Liu, Q.; Sun, W.; Li, J.; Gong, Y.; Liu, D.; et al. Exposure to blue light stimulates the proangiogenic capability of exosomes derived from human umbilical cord mesenchymal stem cells. Stem Cell Res. Ther. 2019, 10, 358. [Google Scholar] [CrossRef]
  229. Motawi, T.M.K.; Sabry, D.; Maurice, N.W.; Rizk, S.M. Role of mesenchymal stem cells exosomes derived microRNAs; miR-136, miR-494 and miR-495 in pre-eclampsia diagnosis and evaluation. Arch. Biochem. Biophys. 2018, 659, 13–21. [Google Scholar] [CrossRef]
  230. Yang, W.; Zhang, J.; Xu, B.; He, Y.; Liu, W.; Li, J.; Zhang, S.; Lin, X.; Su, D.; Wu, T.; et al. HucMSC-Derived Exosomes Mitigate the Age-Related Retardation of Fertility in Female Mice. Mol.Ther. J. Am. Soc. Gene Ther. 2020, 28, 1200–1213. [Google Scholar] [CrossRef]
  231. Yang, C.; Lim, W.; Park, J.; Park, S.; You, S.; Song, G. Anti-inflammatory effects of mesenchymal stem cell-derived exosomal microRNA-146a-5p and microRNA-548e-5p on human trophoblast cells. Mol. Hum. Reprod. 2019, 25, 755–771. [Google Scholar] [CrossRef] [PubMed]
  232. Chen, Q.; Lin, J.; Deng, Z.; Qian, W. Exosomes derived from human umbilical cord mesenchymal stem cells protect against papain-induced emphysema by preventing apoptosis through activating VEGF-VEGFR2-mediated AKT and MEK/ERK pathways in rats. Regen. Ther. 2022, 21, 216–224. [Google Scholar] [CrossRef] [PubMed]
  233. Sun, E.; Karaoz, E. Can Wharton jelly derived or adipose tissue derived mesenchymal stem cell can be a treatment option for duchenne muscular dystrophy? Answers as transcriptomic aspect. Am. J. Stem Cells 2020, 9, 57–67. [Google Scholar] [PubMed]
  234. Fong, C.Y.; Tam, K.; Cheyyatraivendran, S.; Gan, S.U.; Gauthaman, K.; Arunmozhiarasi, A.; Jeyaseelan, K.; Choolani, M.; Biswas, A.; Bongso, A. Erratum: Human Wharton’s Jelly Stem Cells and Its Conditioned Medium Enhance Healing of Excisional and Diabetic Wounds. J. Cell. Biochem. 2017, 118, 3016. [Google Scholar] [CrossRef]
  235. Zhao, C.; Sun, X.; Li, L. Biogenesis and function of extracellular miRNAs. ExRNA 2019, 1, 38. [Google Scholar] [CrossRef]
  236. Stepanov, G.; Zhuravlev, E.; Shender, V.; Nushtaeva, A.; Balakhonova, E.; Mozhaeva, E.; Kasakin, M.; Koval, V.; Lomzov, A.; Pavlyukov, M.; et al. Nucleotide Modifications Decrease Innate Immune Response Induced by Synthetic Analogs of snRNAs and snoRNAs. Genes 2018, 9, 531. [Google Scholar] [CrossRef]
  237. Zhang, Z.; Qin, Y.W.; Brewer, G.; Jing, Q. MicroRNA degradation and turnover: Regulating the regulators. RNA 2012, 3, 593–600. [Google Scholar] [CrossRef]
  238. Biscans, A.; Coles, A.; Haraszti, R.; Echeverria, D.; Hassler, M.; Osborn, M.; Khvorova, A. Diverse lipid conjugates for functional extra-hepatic siRNA delivery in vivo. Nucleic Acids Res. 2019, 47, 1082–1096. [Google Scholar] [CrossRef]
  239. Suter, S.R.; Ball-Jones, A.; Mumbleau, M.M.; Valenzuela, R.; Ibarra-Soza, J.; Owens, H.; Fisher, A.J.; Beal, P.A. Controlling miRNA-like off-target effects of an siRNA with nucleobase modifications. Org. Biomol. Chem. 2017, 15, 10029–10036. [Google Scholar] [CrossRef]
  240. Yu, H.R.; Huang, L.H.; Li, S.C. Roles of microRNA in the immature immune system of neonates. Cancer Lett. 2018, 433, 99–106. [Google Scholar] [CrossRef]
Figure 1. The implication of distinct microRNAs (miRNAs) in the differentiation process of umbilical cord-derived mesenchymal stem cells (UCMSCs). MiRNAs that promote the differentiation of UCMSCs are shown in green, while those that inhibit this process are designated in red. IPCs: Insulin-Producing Cells.
Figure 1. The implication of distinct microRNAs (miRNAs) in the differentiation process of umbilical cord-derived mesenchymal stem cells (UCMSCs). MiRNAs that promote the differentiation of UCMSCs are shown in green, while those that inhibit this process are designated in red. IPCs: Insulin-Producing Cells.
Ijms 24 09189 g001
Figure 2. Clinical applications of umbilical cord-derived stem cells in the context of microRNA (miRNA)-directed regenerative/healing potential.
Figure 2. Clinical applications of umbilical cord-derived stem cells in the context of microRNA (miRNA)-directed regenerative/healing potential.
Ijms 24 09189 g002
Figure 3. Summary of microRNA (miRNA)-guided therapeutic applications of umbilical cord-derived mesenchymal stem cells (UCMSCs). SLE: Systemic Lupus Erythematosus; POI: Primary Ovarian Insufficiency; IBD: Inflammatory Bowel Disease; NAFLD: Non-alcoholic Fatty Liver Disease.
Figure 3. Summary of microRNA (miRNA)-guided therapeutic applications of umbilical cord-derived mesenchymal stem cells (UCMSCs). SLE: Systemic Lupus Erythematosus; POI: Primary Ovarian Insufficiency; IBD: Inflammatory Bowel Disease; NAFLD: Non-alcoholic Fatty Liver Disease.
Ijms 24 09189 g003
Table 1. Detailed roles of distinct microRNAs (miRNAs) in the differentiation process of mesenchymal stem cells (MSCs).
Table 1. Detailed roles of distinct microRNAs (miRNAs) in the differentiation process of mesenchymal stem cells (MSCs).
miRNATissue OriginTarget (Gene/Pathway)Target RegulationmiRNA FunctionReference
miR-424WJMSCsBMP2UpOsteogenic[26]
miR-140-5pUCMSCs, ATMSCs, BMMSCsBMP2DownAnti-Osteogenic[27]
miR-342-3pUCMSCsSufu/ShhDown/UpOsteogenic[28]
miR-196a-5pWJMSCsSERPINB2DownOsteogenic[29]
miR-21UCMSCsKLF12/Wnt/β-cateninDown/UpOsteogenic[30]
miR-21UCMSCsPI3K/β-cateninUpOsteogenic[31]
miR-2110UCMSCs (Exosomes)(TNF)(Up)Osteogenic[32]
miR-328-3pUCMSCs (Exosomes)CHRDDownOsteogenic[32]
miR-25-3pUCMSCsSMAD5/(Dlx5/OSX)Down/(Down)Anti-Osteogenic[33]
miR-33b-5pUCMSCsWnt10b/(Dlx5/OSX)Down/(Down)Anti-Osteogenic[33]
miR-132UCMSCsOsterix/Wnt/β-cateninDown/DownAnti-Osteogenic[34]
miR-216aUCMSCs, ATMSCsc-Cbl/PI3K/AKTDown/UpOsteogenic[35]
miR-122-5pUCMSCsSOX11, VIM, (CUTL1, CAT1)DownHepatogenic[36]
miR-106a, miR-574-3p, miR-451UCMSCsSox17, FoxA2, AFPUpHepatogenic[37]
miR-20b, miR-106aWJMSCsNgn2, MAP2, TUBB3DownAnti-Neurogenic[38]
miR-203UCBMSCsDKK1, CRX, NRLDownAnti-Neurogenic[39]
miR-29b-3pUCMSCsSOX9DownAnti-Chondrogenic[40]
miR-381UCMSCs (sEVs)SOX9/TAOK1Up/DownChondrogenic[41]
miR-410UCBMSCsMITF, LRAT, RPE65, Bestrophin, EMMPRINDownAnti-Epithelial[42]
miR-145UCMSCsTGFβRIIDownEpithelial[43]
miR-200b-3pUCMSCsZEB2DownIPCs Differentiation[44]
miR-375, miR-26aN-UCMSCs (chicken)MTPN, SOX6, BHLHE22, CCND1DownIPCs Differentiation[45]
miR-218UCMSCsMITF, (HoxB4, NF-Ya)Down, (Up)Hematopoietic[46]
miR-301b/miR-130bUCMSCs, ATMSCs, BMMSCsPPARγDownAnti-Adipogenic[47]
miR-21, miR-23a, miR-125b, miR-145UCMSCs (Exosomes)TGF-β2, TGF-βR2, SMAD2DownAnti-Myofibroblastic[48]
miR-503-5pUCMSCsSMAD7DownMyogenetic[49]
miR-222-5pUCMSCsROCK2, αSMADownAnti-Myogenetic[49]
WJMSCs: Wharton’s Jelly-derived Mesenchymal Stem Cells, UCMSCs: Umbilical Cord-derived Mesenchymal Stem Cells, UCBMSCs: Umbilical Cord Blood-derived Mesenchymal Stem Cells, ATMSCs: Adipose Tissue-derived Mesenchymal Stem Cells, BMMSCs: Bone Marrow-derived Mesenchymal Stem Cells, N-UCMSCs: Nestin-positive Umbilical Cord-derived Mesenchymal Stem Cells, sEVs: Small Extracellular Vesicles, IPC: Insulin-Producing Cells.
Table 2. miRNA-guided applications of perinatal and neonatal MSCs in regenerative medicine.
Table 2. miRNA-guided applications of perinatal and neonatal MSCs in regenerative medicine.
miRNATissue OriginVehicle TypeTarget (Gene/Pathway)FunctionClinical ApplicationReference
miR-17-5pUCMSCsEVsPTEN/AKT/HIF-1α/VEGFAngiogenesis/Proliferation/Migration/
Tube Formation Promotion
Diabetic Wound Healing[64]
miR-19aUCMSCsExosomesSOX6, AKT/JNK3/caspase-3Hypoxic Damage ReductionAMI[65]
miR-20aUCMSCsExosomesBeclin-I, FAS, Caspase-3, mTOR, P62, LC3IIApoptosis/Autophagy InhibitionHepatic IRI[66]
miR-21UCMSCsMVsPDCD4Apoptosis InhibitionRenal IRI[67]
UCBMSCs-CHIP/HIF-1αProliferation/Migration/Angiogenesis/
Neovascularization Promotion
CLI[68]
UCMSCsEVsPI3K/AKTProliferation PromotionPeripheral Nerve Injury[69]
UCMSCsEVsPTEN/PI3K/AktProliferation/Migration PromotionCorneal Wound Healing[70]
WJMSCsExosomesPTEN/AktAngiogenesis/Osteogenesis/Cell Survival PromotionONFH[71]
UCMSCsExosomesNOTCH1/DLL4Angiogenesis PromotionBone Regeneration[72]
miR-21-3pUCMSCsExosomesp65, COX2Fibrosis/Inflammation InhibitionTendon Injury[73]
miR-21-5pUCMSCsExosomesSOX5, EZH2Angiogenesis/Osteogenesis PromotionONFH[74]
miR-22-3pUCBMSCsExosomesFZD6Inflammation/Oxidative Stress/Apoptosis Suppression, Proliferation PromotionAcute Lung Injury[75]
miR-23a-3pUCBMSCsExosomes-M2 Macrophage Polarization Promotion, Microglia Activation InhibitionCerebral Infarction[76]
UCBMSCsExosomesDMT1Ferroptosis InhibitionAMI[77]
UCMSCsEVsPTEN/AKTDifferentiation/Proliferation/Migration PromotionCartilage Regeneration[78]
miR-24UCMSCsEVsAQP4/P38 MAPK/ERK1/2/P13K/AKTApoptosis/Inflammation Inhibition, Proliferation/Migration PromotionCerebral IRI[79]
UCMSCsExosomesBimImmune Rejection PreventionIschemic Hindlimb Injury[80]
miR-26b-5pUCMSCsExosomesCH25H-/TLRMicroglia M1 Polarization InhibitionCerebral IRI[81]
UCMSCsExosomesMAT2A/MAPK/STAT3Apoptosis/Inflammatory InhibitionEBI[82]
miR-27bUCMSCsEVsITCH/JUNB/IRE1αProliferation/Migration PromotionSkin Wound Healing[83]
miR-27b-3pUCMSCsExosomesARHGAP5/RhoAProliferation/Invasion PromotionTendon Injury[84]
miR-29aWJMSCs-TIMP-2Angiogenesis, Blood Perfusion PromotionSkeletal Muscle Ischemic Injury[85]
miR-29a-3pUCMSCsExosomesPTEN/mTOR/TGF-β1Differentiation/Healing PromotionTendon Injury[86]
miR-29b-3pUCMSCsEVsPTEN/Akt/mTORNecrosis Reduction, Nerve Function PromotionSCI[87]
UCMSCsEVsPTEN/PI3K/AKTNeuronal Apoptosis InhibitionSCI[88]
UCMSCsExosomesFoxO3Apoptosis/Senescence Promotion, Migration/Inflammation InhibitionCartilage Defect[89]
miR-29b-1-5pWJMSCs-RAP1B/VEGFAngiogenesis Promotion/Tissue RepairDamaged Endometrium[90]
miR-100WJMSCsMVsmTORAutophagy PromotionAcute Lung Injury[91]
miR-100-5pUCMSCsExosomesFOXO3, NLRPΙnflammasome/Cytokine Activation Suppression, Pyroptosis InhibitionAMI[92]
UCMSCsExosomesNOX4ROS Production/Apoptosis InhibitionOsteoarthritis[93]
miR-124UCBMSCsExosomesFoxg1Proliferation/Regeneration Promotion, Injury InhibitionLiver Regeneration[94]
miR-125bUCMSCsExosomesTP53INP1Cell Survival/Migration Promotion, Apoptosis InhibitionSkin Wound Healing[75]
miR-125b-5pUCMSCsExosomesSMAD7Hypoxic Injury/Apoptosis PromotionAMI[95]
UCMSCsExosomesp53Cell cycle arrest/Apoptosis InhibitionIschemic AKI[96]
miR-126UCMSCsExosomesSPRED1, PIK3R2Neurogenesis/Angiogenesis Promotion, Apoptosis InhibitionSCI[97]
miR-133a-3pUCMSCsMIF-ExosomesAKT/VEGFAngiogenesis/Proliferation Promotion, Apoptosis/Fibrosis InhibitionAMI[98]
miR-136UCMSCsExosomesApaf1Apoptosis/Fibrosis/Senescence Suppression, Angiogenesis PromotionAge-associated MI[99]
anti-miR-139-5pUCBMSCs/UCMSCs--Proliferation/Migration/Angiogenesis PromotionPeripheral Arterial Disease (Limb Ischemia)[100]
miR-140-3pUCMSCsEVsSGK1Inflammation/Oxidative Stress/Fibrosis SuppressionRheumatoid Arthritis[101]
miR-140-5pUCMSCs--Chondrogenesis/Cartilage Self-Repair PromotionOsteoarthritis[102]
anti-miR-143UCMSCsExosomesBcl-2/Beclin-1Apoptosis/Autophagy InhibitionCardiac IRI[103]
miR-145UCMSCs-PI3K/AKT/mTORAutophagy PromotionAKI[104]
miR-146a-5pUCMSCsExosomesIRAK1/TRAF6Neuroinflammation SuppressionIschemic Stroke[105]
UCMSCsExosomesTraf6/Irak1/NFκBNeurotoxic Astrocytes ReductionSCI[106]
miR-146bUCMSCs-IRAK1/NF-κBApoptosis/Inflammation InhibitionSepsis-associated AKI[107]
miR-150-5pUCMSCsExosomesPTEN/PI3K/AktGrowth/Migration Promotion, Apoptosis InhibitionSkin Wound Healing[108]
miR-181b-3pUCMSCsEVs-Chondrocyte Proliferation/Cartilage Regeneration InhibitionOsteoarthritis[109]
miR-181cWJMSCsExosomesTLR4/NF-κB/p65Burn-induced Inflammation/Macrophage Inflammation SuppressionBurn Injury[110]
miR-181c-5pUCMSCsEVsSMAD7Proliferation/Migration/Chondrogenesis PromotionCartilage Injury[111]
anti-miR-206UCMSCsExosomesBDNF/TrkB/CREBApoptosis InhibitionEBI[112]
miR-223UCMSCsEVsP53/S100A9Angiogenesis Promotion, Inflammation/Fibrosis SuppressionMI[113]
miR-330-5pUCMSCsSF-SIS ScaffoldCircPTP4A2, PDK2Mitochondrial Metabolism Impairment, Fibrosis PromotionEndometrial Hypoxic Injury[114]
miR-340-5pWJMSCsExosomesIL4Osteochondral RegenerationArticular Cartilage Defect[115]
miR-377-3pUCMSCsExosomesRPTORAutophagy PromotionAcute Lung Injury[116]
miR-410UCMSCsEVsHDAC1/EGR2/Bcl2Apoptosis InhibitionHIBD[117]
miR-421UCMSCsExosomesFOXO3aPyroptosis/Ischemic Injury PromotionAcute Lower Limb Ischemic Injury[118]
miR-451UCMSCsExosomesTLR4/NF-κB, MIF/PI3K/AktInflammation Suppression, M2 Macrophage Polarization ActivationAcute Lung Injury[119]
miR-455-3pUCMSCsExosomesPIK3r1/IL-6Macrophages Activation Inhibition, Inflammatory Cytokine SuppressionAcute Liver Injury[120]
miR-455-5pUCMSCs-SOCS3/JAK/STAT3Cell Cycle/Proliferation Promotion, Endometrial Glands Increase, Fibrosis SuppressionEndometrial Injury[121]
anti-miR-486-5pUCBMSCs-PIM1/TGF-β/SmadApoptosis/Inflammation Suppression, Proliferation PromotionOxidative Stress Injury[122]
miR-663aUCMSCsExosomesCDKN2AProliferation Promotion, Apoptosis/Migration/EMT InhibitionEndometrial Hypoxic Injury[123]
miR-675UCMSCsExosomesp21, TGF-β1Blood Perfusion Promotion, Senescence InhibitionIschemia-induced Vascular Dysfunction[124]
miR-1208UCMSCsEVsMETTLE3Proliferation/Migration Promotion, Apoptosis/Inflammation InhibitionOsteoarthritis[125]
miR-1246UCBMSCsExosomesGSK3β-Wnt/β-cateninApoptosis InhibitionHepatic IRI[126]
UCMSCsExosomesIL-6/gp130/STAT3Th17/Treg Balance RegulationHepatic IRI[127]
miR-1263UCMSCsExosomesMob1/HippoApoptosis InhibitionDisuse Osteoporosis[128]
miR-7162-3pUCMSCsExosomesAPOL6Apoptosis Inhibition, Cell Repair/RegenerationEndometrial Injury[129]
UCMSCs: Umbilical Cord-derived Mesenchymal Stem Cells, UCBMSCs: Umbilical Cord Blood-derived Mesenchymal Stem Cells, WJMSCs: Wharton’s Jelly-derived Mesenchymal Stem Cells, EVs: Extracellular vesicles, MVs: Microvesicles, MIF: Macrophage Migration Inhibitor, SF: Silk Fibroin, SIS, Small Intestinal Submucosa, AMI: Acute Myocardial Infarction, IRI: Ischemic/Reperfusion Injury, CLI: Critical Limb Ischemia, ONFH: Osteonecrosis of the Femoral Head, EBI: Early Brain Injury, SCI: Spinal Cord Injury, AKI: Acute Kidney Injury, HIBD: Hypoxia-Ischemia Brain Damage.
Table 5. Multiple miRNAs/miRNA signatures that mediate MSC-based regenerative/healing applications.
Table 5. Multiple miRNAs/miRNA signatures that mediate MSC-based regenerative/healing applications.
miRNAsTissue OriginVehicle TypeTarget (Gene/Pathway)FunctionClinical ApplicationReference
miR-199a-3p/145-5pUCMSCsExosomesCblb, Cbl, NGF/TrkANeuronal Differentiation PromotionSCI[131]
miR-let-7a, miR-let-7eWJMSCsEVsCasp3Apoptosis InhibitionHypoxic-ischemic Brain Injury[225]
miR-21-5p, miR-125b-5pUCBMSCsExosomesTGF-βI, TGF-βIIWound Closure Acceleration, Scar Formation Inhibition, Skin/Nerve/Vessel RegenerationSkin Wound Healing[190]
miR-21, miR-29, miR-221, let-7aWJMSCsEVsBMP, PI3K/AKTOsteogenic Differentiation/Osteoblast Activity PromotionOsteoporosis[226]
miR-122-5p, miR-148a-3p, miR-486-5p, miR-let-7a-5p, miR-100-5pUCMSCsEVsPI3K/AktCartilage Degradation/Inflammation Suppression, M2 Macrophage Polarization PromotionOsteoarthritis[192]
miR-92b-3p, miR-32-5p, let-7b-5p, miR-19a-3p, miR-19b-3pWJMSCsExosomesDKK3Osteochondral RegenerationArticular Cartilage Defect[115]
miR-23a-3p, miR-221-3p, miR-23b-3p, miR-141-3p, miR-144-3p, miR-200a-3p, miR-454-3p, miR-23c, miR-320bWJMSCsExosomesCXCL12Osteochondral RegenerationArticular Cartilage Defect[115]
miR-374a-5p, miR-495-3p, miR-323a-3pWJMSCsExosomesCCL2Osteochondral RegenerationArticular Cartilage Defect[115]
miR-19b-3p, miR-185-5pWJMSCsExosomesPOSTNOsteochondral RegenerationArticular Cartilage Defect[115]
let-7e-5p, miR-423-5p, miR-199a-3p, miR-125b-5p, miR-142-3p, miR-92a-3pWJMSCsEVsECM-receptor interaction, NOTCH, (STAT3, IGF1R)Proliferation/Migration/M2 Infiltration Promotion, Homeostasis Maintenance, Injury Degradation SuppressionOsteoarthritis[193]
miR-100, miR-146a, (miR-21, miR-221, miR-143)UCMSCsExosomes-Cell Cycle/Proliferation Promotion, Apoptosis InhibitionVaginal Reconstruction[227]
miR-135b-5p, miR-499a-3pUCMSCsExosomesMEF2CAngiogenesis PromotionTissue Regeneration[228]
miR-136, miR-494, miR-495UCMSCsExosomes-Biomarkers (Diagnosis, Evaluation)Pre-Eclampsia[229]
miR-21, miR-146a-5pUCMSCsExosomesPI3K/mTORFertility Recovery, Oocyte Production PromotionPremature Ovarian Insufficiency[230]
miR-146a-5p, miR-548e-5pUCMSCsExosomesNF-κB, AKT, MAPKInflammation Suppression, Proliferation/Migration PromotionInflammatory Diseases/Preterm Birth[231]
miR-17 SuperfamilyWJMSCs, BMMSCsExosomes(BMPR2)Proliferation PromotionPulmonary Hypertension[174]
miR-10a-5p, miR-146a-5UCMSCsExosomesVEGF-VEGFR2/AKT, MEK/ERKApoptosis InhibitionChronic Obstructive Pulmonary Disease[232]
miR-24, miR-199a-5pWJMSCsExosomes-Muscle RegenerationDuchenne Muscular Dystrophy[233]
miR-124, miR-145Bone Marrow/Adipose Tissue/Placenta/Umbilical Cord MSCs-SCP-1, Sox2Migration/Cell Self-Renewal InhibitionGlioma[203]
let-7a-2-3p, let-7d, let-7e, let-7f, let-7f-1-3p, let-7g, let-7i, let-7i-3p, miR-100, miR-106a, miR-106b, miR-125a-3p, miR-125a-5p, miR-126, miR-146a, miR-17, miR-181a, miR-18b, miR-196a-3p, miR-19a, miR-19b, miR-200b, miR-20a, miR-20b, miR-21, miR-210, miR-25, miR-27a-5p, miR-29b, miR-302a, miR-302b-5p, miR-302c-5p, miR-30c, miR-31, miR-32, miR-335, miR-34a, miR-374a, miR-378 miR-3915, miR-3924, miR-601, miR-622, miR-920, miR-92a, miR-93, miR-98WJMSCs--Wound Healing PromotionExcisional/Diabetic Wounds[234]
UCMSCs: Umbilical Cord-derived Mesenchymal Stem Cells, UCBMSCs: Umbilical Cord Blood-derived Mesenchymal Stem Cells, WJMSCs: Wharton’s Jelly-derived Mesenchymal Stem Cells, MSCs: Mesenchymal Stem Cells EVs: Extracellular Vesicles SCI: Spinal Cord Injury.
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

Thomaidou, A.C.; Goulielmaki, M.; Tsintarakis, A.; Zoumpourlis, P.; Toya, M.; Christodoulou, I.; Zoumpourlis, V. miRNA-Guided Regulation of Mesenchymal Stem Cells Derived from the Umbilical Cord: Paving the Way for Stem-Cell Based Regeneration and Therapy. Int. J. Mol. Sci. 2023, 24, 9189. https://doi.org/10.3390/ijms24119189

AMA Style

Thomaidou AC, Goulielmaki M, Tsintarakis A, Zoumpourlis P, Toya M, Christodoulou I, Zoumpourlis V. miRNA-Guided Regulation of Mesenchymal Stem Cells Derived from the Umbilical Cord: Paving the Way for Stem-Cell Based Regeneration and Therapy. International Journal of Molecular Sciences. 2023; 24(11):9189. https://doi.org/10.3390/ijms24119189

Chicago/Turabian Style

Thomaidou, Arsinoe C., Maria Goulielmaki, Antonis Tsintarakis, Panagiotis Zoumpourlis, Marialena Toya, Ioannis Christodoulou, and Vassilis Zoumpourlis. 2023. "miRNA-Guided Regulation of Mesenchymal Stem Cells Derived from the Umbilical Cord: Paving the Way for Stem-Cell Based Regeneration and Therapy" International Journal of Molecular Sciences 24, no. 11: 9189. https://doi.org/10.3390/ijms24119189

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