Next Article in Journal
Evaluation of Solutions Containing Fluoride, Sodium Trimetaphosphate, Xylitol, and Erythritol, Alone or in Different Associations, on Dual-Species Biofilms
Previous Article in Journal
Can RNA Affect Memory Modulation? Implications for PTSD Understanding and Treatment
Previous Article in Special Issue
Impact of Environmental and Epigenetic Changes on Mesenchymal Stem Cells during Aging
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Molecular Regulatory Mechanism in Multipotency and Differentiation of Wharton’s Jelly Stem Cells

The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(16), 12909; https://doi.org/10.3390/ijms241612909
Submission received: 23 July 2023 / Revised: 6 August 2023 / Accepted: 10 August 2023 / Published: 18 August 2023
(This article belongs to the Special Issue Genomics and Epigenetics of Stem Cell)

Abstract

:
Wharton’s jelly-derived mesenchymal stem cells (WJ-MSCs) are isolated from Wharton’s jelly tissue of umbilical cords. They possess the ability to differentiate into lineage cells of three germ layers. WJ-MSCs have robust proliferative ability and strong immune modulation capacity. They can be easily collected and there are no ethical problems associated with their use. Therefore, WJ-MSCs have great tissue engineering value and clinical application prospects. The identity and functions of WJ-MSCs are regulated by multiple interrelated regulatory mechanisms, including transcriptional regulation and epigenetic modifications. In this article, we summarize the latest research progress on the genetic/epigenetic regulation mechanisms and essential signaling pathways that play crucial roles in pluripotency and differentiation of WJ-MSCs.

1. Introduction

WJ-MSCs were originally isolated as fibroblast-like cells from human umbilical cords by McElreavey [1]. Subsequent studies found that the surface of these cells express CD105, CD73, and CD90 surface markers of mesenchymal stem cells [2]. They can differentiate into bone, cartilage, and fat cells [3]. These features meet the standard of pluripotent mesenchymal stem cells established by the International Society for Cell Therapy in 2006 [4]. Therefore, WJ-MSCs have attracted massive attention as an important source of multipotent stem cells. Interestingly, pluripotency markers such as OCT4, NANOG, SOX2, and MYC are expressed in WJ-MSCs [5]. Thus, WJ-MSCs have some characteristics of both embryonic stem cells (ESCs) and adult stem cells and may be a bridge between these two types of cells [6]. WJ-MSCs are convenient to isolate and there are no ethical disputes associated with their use. Importantly, WJ-MSCs are not tumorigenic and have strong cell proliferation, differentiation, and immune regulation capabilities [7]. Therefore, WJ-MSCs have become more and more important in applications of mesenchymal stem cells. Due to these advantages, studies on WJ-MSCs and related applications have gradually increased. Hence, it is very important to study the molecular regulatory mechanisms of their biological characteristics.
Epigenetic regulation is the reversible and heritable change of gene expression levels by altering the topological structure of chromatin and regulating transcription initiation, elongation, DNA replication, and DNA repair without modifying the gene sequence [8]. Epigenetic regulation is mainly achieved through DNA methylation, histone modifications, and non-coding RNAs [9]. Epigenetic modification plays a fundamental regulatory role in the process of determining the fate of stem cells, exerting influence at multiple levels pre-, mid-, and post-transcription. Thus, epigenetic modification is an endogenous regulatory mechanism in stem cell fate decisions [10].
Gene expression directs protein synthesis and cellular functions. One of the mechanisms that regulates the complex relationship between genes and their products is transcriptional regulation inside the cell. Transcription factors (TFs) can bind to a specific gene sequence through their DNA binding domain and assist RNA polymerase II and TFII to form an effective transcription initiation complex, thereby initiating gene expression [11]. TFs play a central role in determining cell differentiation fate throughout embryonic development and maintaining the cellular properties of adult tissues [12]; the expression of even a single TF can determine the direction of cell differentiation [13].
Epigenetic regulation and transcriptional regulation can explain the unique biological characteristics of WJ-MSCs at the molecular level. Therefore, this article concisely summarizes the research progress on the epigenetic and transcriptional regulation mechanisms that maintain the properties of WJ-MSCs. We also briefly discuss the related signaling pathways that play an important role in WJ-MSCS.

2. Epigenetic Regulation

2.1. DNA Methylation

DNA methylation is catalyzed by DNA methyltransferases (DNMTs), using S-Adenosyl methionine (SAM) as a methyl donor, and transferring a methyl group to cytosine phosphate guanine dinucleotide co-transferring to CpG islands. The process results in the formation of 5-methylcytosine (5mC) through valent combination [14,15]. DNA demethylation is the process of removing a methyl group from cytosine through active or passive demethylation processes. TET (Ten-eleven translocation) is mainly involved in the active demethylation process since it can demethylate DNA by oxidizing 5mC to an unmethylated cytosine intermediate [16,17]. These two types of enzymes, DNMTs and TETs, play an important role in the process of DNA methylation/demethylation, thus regulating the maintenance, proliferation, differentiation, and apoptosis of stem cells by activating or repressing gene expression.
DNMT3A is highly expressed in differentiated cells and is involved in DNA de novo methylation, and hence it plays an important role in normal development [18]. EID3 can repress transcription and inhibit cell differentiation [19]. Studies have shown that EID3 can directly affect DNMT3A in WJ-MSCs, thereby directing MSCs to differentiate [20]. In mice, Tet2 can regulate the cell cycle of MSCs [21]. Tet1 and Tet2 can ensure the self-renewal and differentiation of MSCs by maintaining the demethylation status of the p2rx7 promoter [22]. TET1 and TET2 play the same role in human MSCs, and they affect the function of MSC differentiation by regulating lineage-determining genes [23]. Interestingly, TET1 and TET2 have different expression patterns in MSCs from different sources [24]. However, whether TET plays the same role in WJ-MSCs has not been reported and further exploration is needed.
The methylation status of the promoter regions of pluripotency genes such as OCT4 and NANOG can limit the differentiation potential of MSCs, while the methylation status of lineage-specific genes can also affect the differentiation potential of MSCs [25]. Hypermethylation in adipose, bone marrow, and muscle-derived MSCs hinders differentiation, whereas hypomethylation status has no predictive value for differentiation potential [26,27]. However, hypomethylation in WJ-MSCs has an impact on differentiation potential. Studies have shown that hypomethylation in the promoter regions of cardiac lineage-specific factors GATA4, TBX5, SERCA, and NKX2.5 in WJ-MSCs correlates with differentiation potential. Due to the hypomethylation status of cardiac lineage-specific factors in WJ-MSCs, they are ideal cell sources for differentiation into cardiomyocytes [28]. In WJ-MSCs, adipose differentiation is also associated with hypomethylation status, and the decrease of CpG methylation in the MEST promoter may promote the binding of SOX6, thereby enhancing the expression of MEST and stimulating the differentiation of WJ-MSCs into adipocytes [29]. It is noteworthy that this difference between WJ-MSCs and MSCs derived from other sources may also be regulated by DNA methylation [30].

2.2. Histone Modifications

Histones are the basic structural proteins in eukaryotic chromatin and are mainly divided into H2A, H2B, H3, and H4. Histones can be modified in a variety of ways, such as histone methylation, acetylation, phosphorylation, ubiquitination, and ADP-ribosylation [31]. These diverse histone modifications can dynamically switch chromatin states from transcriptionally active to transcriptionally silent, and vice versa.
It has been found that the changes of histone H3K9ac and histone H3K4me2 in the OCT4 and NANOG promoters were consistent with the corresponding gene expression levels in WJ-MSCs. By affecting histone H3K9 or H3K14 acetylation and histone H3K4 dimethylation, the mRNA expression of pluripotency-related genes and proliferation genes can be increased and the spontaneous differentiation of MSCs can be inhibited [32]. RUNX2 is an important factor in regulating osteogenic differentiation [33], and the P1 promoter region of RUNX2 regulates the transcription of RUNX2/p57 which are the main regulators of osteogenic differentiation [34]. In undifferentiated WJ-MSCs, the P1 promoter region of RUNX2 lacks the histone marks H3ac, H3K27ac, and H3K4me3, which are associated with transcriptionally active genes. When the histone marks H3ac, H3K27ac, and H3K4me3 were enriched in the P1 promoter region, RUNX2/P57 gene expression was activated and WJ-MSCs differentiated toward osteoblasts [35]. H3K4 methylation, H3K27 acetylation, and DNA demethylases also have a strong correlation with upregulation of gene expression, and they can jointly regulate gene expression [36] (Figure 1).
Histone modification is inseparable from the action of related enzymes. Histone deacetylases (HDACs) mediate the deacetylation of histone, compacting chromatin, impeding the ability of transcription factors to bind to DNA, and thus inhibiting gene transcription. There are currently 18 HDACs found in humans, which are mainly divided into 4 types [37]. In embryonic stem cells, inhibition of HDACs reduces the self-renewal capacity of the cells and increases the expression of differentiation markers. Likewise, HDACs are also critical for maintaining the self-renewal and differentiation of MSCs. It has been reported that inhibition of HDACs, histone H3, and H4 acetylation can block the G2/M phase of cells, inhibiting self-renewal, increasing the expression of osteogenic genes, and thus directing MSCs to differentiate into osteoblasts [38,39]. However, this is contrary to other reports [32]. The discrepancy may be due to the use of different cell types or different HDAC inhibition methods, which may produce different effects. Interestingly, inhibition of HDAC in WJ-MSCs can reduce cell proliferation [37] and promote cardiomyocyte formation by upregulating NKX2.5 [40], and inhibition of HDAC IIa can induce WJ-MSCs to differentiate into pancreatic endocrine-like cells [41].

2.3. Non-Coding RNA

Non-coding RNAs play an irreplaceable role in basic physiological processes including development, immunity, and reproduction. There are more than 100 known RNA modifications, many of which can participate in the regulation of eukaryotic gene expression.
miRNAs are a class of small, endogenous, non-coding single-stranded RNAs. miRNAs target the 3′-UTR region of mRNAs. As a result, the mRNAs are degraded or translation is inhibited, thereby affecting the protein expression level [42]. Thus, miRNAs play a crucial role in post-transcriptional regulation [43]. miRNAs are involved in many biological processes such as cell proliferation, differentiation, apoptosis, and migration, and can also participate in the regulation of stem cell pluripotency [44,45]. In WJ-MSCs, miR-196b-5p can block the G0/G1 phase, inhibit cell proliferation, and promote collagen content in the extracellular matrix. Meanwhile, this miRNA can regulate the downstream key gene SERPINB2 of osteogenic differentiation and stimulate osteogenic ability, thus promoting new bone formation [46,47]. miRNAs also play an important role in the differentiation of WJ-MSCs into neurons [48]. In mice, overexpression of miR-3099 can up-regulate genes related to neuronal differentiation: NeuN, Tuj1, NeuroD1, Sox4, Gat1, vGluT1, and vGluT2, and down-regulate genes related to astrocyte generation process including Gfap, S100β, and Slc1a3, thus playing a key role in controlling the expression of key neuronal plasticity markers and in the regulation of learning and memory. In humans, MDS21, which is highly expressed in WJ-MSCs, targets similar genes as miR-3099 [49]. This may be one of the reasons why WJ-MSCs are more advantageous in neuron differentiation. miR-20b and miR-106a can directly or indirectly regulate the expression of neuronal genes to affect neural differentiation by inhibiting the expression of NGN2, MAP2, and TUBB3 [50]. In WJ-MSCs, depletion of endogenous miR-34a expression results in enhanced motility of WJ-MSCs, which may contribute to neuronal precursor motility [51]. SOX11 and VIM can maintain the undifferentiated state of WJ-MSCs. During the differentiation of WJ-MSCs into hepatocytes, miR-122-5p can bind to the 3′-UTR regions of SOX11 and VIM and inhibit the expression of SOX11 and VIM, thus promoting the differentiation of WJ-MSCs into hepatocytes by down-regulating mesenchymal marker genes [52]. In addition, miR-146a-5p is essential for the motility and proliferation of MSCs. In WJ-MSCs, knockdown of miR-146a-5p can inhibit cell proliferation and enhance cell migration [53].
CircRNAs are novel non-coding RNA molecules that do not have a 5′ cap and 3′ poly(A) tail and are formed by covalent bonds creating a ring structure. CircRNAs are species-, tissue-, and time-specific and can regulate cell differentiation, tissue homeostasis, and disease development. CircRNA contains miRNA binding sites and can function as a miRNA sponge, thus indirectly regulating the expression of downstream target genes of miRNA. In the process of stem cell differentiation, many circRNAs are differentially expressed, and the stem cell differentiation of different lineages can be regulated through the circRNA–miRNA–mRNA interaction network. Hence, CircRNA can be used as a biomarker and target for stem cell therapy [54]. In WJ-MCSs, circRNA also plays an important role. For instance, CircRNA_05432, circRNA_08441, and circRNA_01536 are related to cell proliferation and differentiation and play a role in the differentiation of WJ-MSCs into cardiomyocyte-like cells [55]. CDR1as also participates in the proliferation and differentiation of WJ-MCSs, regulates the cell cycle and apoptosis of WJ-MSCs, and affects the expression of stem transcription factors, which plays an important role in pluripotency regulation. Interestingly, circRNA can also influence the therapeutic effect of stem cells. For example, circ6401 binds to miR-29b-1-5p in WJ-MSCs and regulates the repair of the endometrium by affecting the VEGF signaling pathway [56].
LncRNAs are non-coding RNAs with a length of more than 200 nucleotides and have no ability to encode proteins. They can participate in the methylation process of CpG islands [34] and the recruitment of related essential molecules to methylated or ubiquitinated proteins, hence playing roles in epigenetic regulation [57]. LncRNA are important factors in the regulation of MSC function [58]. lncRNA CIR can bind to EZH2 and inhibit the expression of ATOH8 and the chondrogenic differentiation of WJ-MSCs through EZH2-mediated H3K27me3 [59]. ITGA6-AS1 is the antisense non-coding RNA of ITGA6. ITGA6 can maintain the self-renewal of stem cells. The post-transcriptional regulation of ITGA6 in WJ-MSCs is controlled by ITGA6-AS1. Therefore, the regulation of ITGA6-AS1 may be associated with the cell identity of WJ-MSCs [60].
Interestingly, non-coding RNA can interact with DNA methylation and histone modification to form a precise network of gene regulation. Oct4 overexpression can downregulate miR-148a through DNA methylation. miR-148a can directly regulate DNMT1 by targeting the 3′-UTR of its transcript. Thus, reduced expression of miR-148a can induce high levels of DNMT1 [44]. OCT4 and NANOG can also directly bind to the DNMT1 promoter to increase the expression of DNMT1. Increased expression levels of DNMT1 can then lead to repressed expression of genes associated with development and lineage differentiation [61] (Figure 2).

3. Transcriptional Regulation

Transcriptional regulation changes the level of gene expression by altering the rate of transcription. Transcription factors (TFs) play a vital role in the mechanism of regulating transcription. TFs regulate chromatin structure and gene transcription by recognizing specific DNA sequences and play an important role in gene expression, cell differentiation and development, autophagy, metabolism, and apoptosis [62]. Compared with other types of stem cells, WJ-MSCs have a unique transcriptional profile. Genes related to the immune system, chemotaxis, and cell death are highly expressed in WJ-MSCs [63]. The formation of this unique transcriptional expression profile is highly associated with the intracellular TF function in WJ-MSCs.
OCT4, SOX2, and NANOG are key transcription factors regulating ESC pluripotency and reprogramming [64,65]. It is interesting that undifferentiated ESC markers can also be detected in WJ-MSCs. These genes include OCT4, SOX2, NANOG, LIN28, SSEA1, SSEA3, SSEA4, KLF4, MYC, CRIPTO, REX1, TDGF1, and ZFP42 [63,66]. Although the expression levels of OCT4, NANOG, SOX2, and LIN28 are low in WJ-MSCs [63], their expression remains detectable despite different isolation methods and preservation techniques [67]. These suggest that WJ-MSCs have similar primitive, undifferentiated properties with ESCs.
In ESCs, the pluripotency and self-renewal of ESCs are maintained through the “Oct4-centered” and “Myc-centered” transcriptional regulatory networks [68]. In MSCs, OCT4 can target genes similar to those in ESCs, promote the expression of MSCs-specific genes, and regulate the pluripotency of MSCs through similar regulatory mechanisms [69]. OCT4 can bind to the SOX2 promoter to initiate SOX2 transcription. The expression level of SOX2 in WJ-MSCs is much higher than that of OCT4 [70], and SOX2 is considered to be an important factor regulating the pluripotency and cell cycle of WJ-MSCs [71]. The transcription of SOX2 is regulated by a variety of transcription factors. In addition to OCT4, the transcription of SOX2 is also regulated by E2F1. After E2F1 binding to the SOX2 promoter, the expression of SOX2 increases and cell stemness is enhanced [72]. SOX2 can regulate cell growth by inhibiting CCND1 and CDK4 [73]. In WJ-MSCs, E2F1, as a transcription factor, can regulate the expression of SOX2. It also binds to open chromatin structures containing histone H3K27ac and H3K4me3 and activates ELOVL2 expression, leading to impaired mitochondrial oxygen utilization function and affecting cell metabolism [74].
NR2F2 (nuclear receptor family F group 2) is mainly expressed in mesenchymal cells. NR2F2 is an important regulator of differentiation and is related to tissue homeostasis. NR2F2 has been considered to be a key regulator of mesenchymal stem cell fate. The regulation of NR2F2 on stem cell fate cannot be separated from the role of miRNA. MiR-194 directly targets the 3′-UTR of NR2F2 mRNA to negatively regulate NR2F2 expression, thus regulating bone and fat formation [75]. NR2F2 also affects the formation of cartilage, and MSCs overexpressing NR2F2 show significantly enhanced cartilage formation [76]. In WJ-MSCs, NR2F2 can also regulate the expression of CCND1 and CDK4, affecting cell proliferation and inflammatory factor secretion of WJ-MSCs, and has potential roles in the treatment of diseases such as immunity and cancer [77].
p53 is a key transcription factor involved in the regulation of cell differentiation, cell stemness, and development [78]. Loss of p53 leads to accelerated proliferation and differentiation of MSCs, which are more inclined to differentiate into adipocytes and osteoblasts [79,80]. Some studies have shown that p53 inactivation plays an active role in stemness maintenance, as knocking out p53 in MSCs increases the expression of stemness markers such as OCT4, SOX2, and NANOG [81]. It is noteworthy that p53 can directly or indirectly inhibit the functions of important transcription factors in stem cells, and thus the ultimate biological outcome of p53 is dependent on the specific functions of those transcription factors in specific stem cell types [82]. p53 can also bind to OSX and prevent OSX and transcriptional cofactors from binding to Sp1/GC-rich sequences in OSX, thereby inhibiting OSX transcription. OSX is an important transcription factor regulating osteogenic differentiation, and its transcriptional repression directly affects osteogenic differentiation [83].
SNAI2 (formerly known as Slug) is a zinc finger transcription factor that is usually considered to play a role in the process of epithelial–mesenchymal transition. Studies have found that transcription factors related to epithelial–mesenchymal transition also regulate the functions of embryonic and adult stem cells, which are also the main regulatory factors for stem cell function and cell differentiation [84]. The interaction between Snail/Slug and YAP/TAZ controls the self-renewal and differentiation characteristics of mesenchymal stem cells [85]. During the differentiation of WJ-MSCs into osteoblasts, overexpression of SNAI2 can promote the deposition of cellular mineralized matrix [86].
HNF4A is a transcriptional regulator that controls the expression of liver-related genes during the transformation of endodermal cells to hepatic progenitor cells and is also associated with stem cell differentiation. In WJ-MSCs, overexpression of HNF4A can improve the hepatic differentiation capacity of WJ-MSCs [87]. Hence, overexpression of HNF4A in WJ-MSCs may be a better choice to treat acute liver failure [88].
Taken together, although some transcription factors have been shown to be important in regulating the biological characteristics of WJ-MSCs, systematic screening for essential transcription factors that are crucial for WJ-MSCs has been lacking until recently. Therefore, research which aims to decode the transcription network that governs cell WJ-MSCs’ fate will be prominent in the near future.

4. Signaling Pathways

The Wnt pathway regulates key developmental processes and tissue homeostasis by coordinating cell proliferation, differentiation, cell polarity, cell motility, and stem cell renewal. The Wnt signaling pathway can be divided into the Wnt/β-Catenin pathway, Wnt/PCP pathway, and Wnt/Ca2+ pathway. The first type is a canonical Wnt signaling pathway, and the latter two are non-canonical Wnt signaling pathways [89]. Interestingly, the Wnt signaling pathway can regulate the self-renewal and differentiation of MSCs [90]. Differences in the expression of Wnt signaling-related molecules affect the osteogenic differentiation of MSCs. Compared with BM-MSCs, the Wnt signaling inhibitor DKK1 is upregulated in WJ-MSCs, thus the Wnt/β-Catenin pathway is inhibited and the expression of its downstream target gene WISP1 is lower. The decreased expression of WISP1 may be the reason why the ability of WJ-MSCs to differentiate into osteoblasts is not as good as that of BM-MSCs [91]. The Wnt signaling pathway also promotes cardiomyocytes but inhibits cardiomyocyte differentiation at later stages of development. WNT signaling represses HDAC1 expression through the β-catenin/LEF1 pathway, resulting in decreased HDAC1 expression and increased NKX2.5 expression and differentiation toward cardiomyocytes, but it has the opposite effect at late stages [92] (Figure 3).
MAPKs are a group of evolutionarily conserved serine–threonine kinases, which can be divided into four subfamilies: ERK, p38, JNK, and ERK5, representing four different cascade pathways, respectively. The MAPK pathway can be activated by extracellular stimuli such as cytokines, cellular stress, hormones, and neurotransmitters. After the upstream activator protein binds to a specific receptor, it activates its downstream transcription factors through the gradual phosphorylation of MAP3K–MAP2K–MAPK, and then regulates diverse cellular functions including gene expression, cell proliferation, migration, differentiation, and stress response in MSCs [93,94,95]. In WJ-MSCs, CDC42 can affect the proliferation of WJ-MSCs and the secretion of inflammatory factors IL-6 and IL-8 by affecting the phosphorylation of ERK1/2 and p38 MAPK [96]. IL-1β can induce the expression of MMP-3 and enhance the migration of WJ-MSCs through ERK1/2, JNK, and p38 MAPK signaling pathways [97]. IGFBP2 promotes adipogenic differentiation of WJ-MSCs by increasing the phosphorylation of JNK/AKT and activating the JNK/AKT signaling pathway [98]. COL induces osteogenic differentiation by activating the transcription of BMP6/7 and TGFB3, and then activating the downstream target genes INS, IGF1, RUNX2 and VEGFR2 through the p38/MAPK pathway [99]. After BMP receptors are activated, phosphorylated TAK1 recruits TAB1 to initiate MKK-p38 MAPK or MKK-ERK1/2 signaling (Figure 4).
In WJ-MSCs, the SHH pathway, NF-kB pathway, and STAT pathway also play important roles. The SHH pathway can stimulate and promote the production of angiogenic factors such as activin A, angiopoietin, angiopoietin 1, granulocyte-macrophage colony-stimulating factor, matrix metalloprotein peptidase-9, and urokinase-type plasminogen activator. These factors can be secreted to enhance the pro-angiogenic ability of WJ-MSCs in vitro and in vivo [100]. Activation of the NF-kB pathway can protect WJ-MSCs from serum deprivation-induced apoptosis [101]. After the NF-kB pathway is inhibited, its downstream iNOS and cytokine signaling are inhibited, and the antioxidant enzyme system is enhanced. Hence, the anti-inflammatory effect of WJ-MSCs can be better exerted [102]. The STAT pathway can mediate the immunosuppression of B7-H1 induced by IFN-γ [103].

5. Other Important Molecules in Differentiation of WJ-MSCs

WJ-MSCs have strong differentiation ability and can be induced into different types of cells through different induction methods. Some key factors play an important role in this process.
JARID1B (Jumonji AT-rich interactive domain 1B) is a histone demethylase that can specifically remove the H3K4me3 mark and inhibit transcription of its related genes, playing an important role in cell fate determination, cancer progression, and stem cell self-renewal role [104]. In WJ-MSCs, the activity of JARID1B can regulate the expression of RUNX2 and affect the differentiation ability of osteoblasts.
KGF (keratinocyte growth factor) is a member of the fibroblast growth factor family. KGF is produced by mesenchymal cells and exerts its biological effects by binding to their receptors with high affinity. KGF plays an important role in regulating embryonic development, cell proliferation, and cell differentiation. Studies have shown that KGF can affect the proliferation of pancreatic ductal cells through the MEK-ERK1/2 pathway and induce the differentiation of pancreatic ductal cells into β cells through the PI3K/AKT pathway [105]. KGF is also a key growth factor for the differentiation of WJ-MSCs into sweat gland-like cells [106], although the specific mechanism has not yet been uncovered.
NGF (Nerve growth factor) is a neurotrophic factor that induces neurite outgrowth and promotes the survival and maintenance of neurons. NGF, its precursor molecule pro-NGF, and its different receptor systems, such as TrkA, p75NTR, and sortilin, play key roles in the development and physiological functions of the nervous and immune systems. The combination of NGF and TrkA can activate Ras and downstream MAPK pathways [107]. Interestingly, overexpression of NGF in WJ-MSCs will affect the expression levels of SOX1, SOX2, and NES, showing that overexpression of NGF can lead to the differentiation of MSCs into neural cells [108]. BDNF (Brain derived neurotrophic factor) is also an important neurotrophic factor that can guide cells into the pathway of neuronal differentiation [109]. BDNF activates the ERK pathway by binding to specific Trk receptors and participates in cell differentiation [110].

6. Combined Factors Mediate Osteogenic Differentiation of WJ-MSCs

To date, a great deal of efforts have been put into directing WJ-MSCs to differentiate into osteoblast for clinical applications. Intriguingly, the differentiation of WJ-MSCs into osteoblasts involves different genetic/epigenetic regulatory mechanisms that work together to promote WJ-MSCs to differentiate into osteoblasts. For instance, RUNX2, a downstream target gene of the p38/MAPK signaling pathway [99], is also an important regulator of osteogenic differentiation. When JARID1B is enriched in the P1 promoter region of RUNX2, the levels of activation marks, including histone H3ac and H3K4me3, are reduced, whereas increased levels of the suppressive mark histone H3K27me3 lead to the inhibition of RUNX2 transcription [34]. On the contrary, when JARID1B is inactivated, histone marks H3ac, H3K27ac, and H3K4me3 are enriched in the P1 promoter region, and SNAI2 also binds to the RUNX2 promoter to activate RUNX2 expression [111].
In addition, coupled with the activation of the MAPK signaling pathway, the transcriptional activity of RUNX2 can be further increased after phosphorylation [112]. Furthermore, RUNX2 can mediate the BMP-2-induced expression of OSX [113]. BMP2 is regulated by the downstream target genes WISP1 [114] and miR424 [115] of the Wnt signaling pathway, which enhances its induction of OSX. OSX is another important regulator of osteogenic differentiation. When the promoter region of OSX is bound by p53, its transcription is inhibited, thus affecting osteogenic differentiation [83]. Conversely, when p53 is inactivated, H2BK120ub1 and H3K4me3 are enriched in the OSX promoter region, and OSX transcription can be increased [116]. At the same time, the activation of the MAPK signaling pathway can also increase the transcriptional activity of OSX [112] and promote the transcription of downstream osteogenic genes OPN, OCN, and COL1A1. During osteogenic differentiation, the increased activity of H2AK119ub, H2BK120ub, and H3K4me3 [116], and the participation of CDR1as [117] and circ-CTTN [118] can jointly promote the differentiation of osteoblasts. Hence, many factors can induce WJ-MSCs to differentiate into osteoblasts.

7. Conclusions and Prospects

WJ-MSCs have broad application prospects due to their unique biological characteristics. Their molecular features are regulated by complicated epigenetic mechanisms, specific transcription factors, and signaling pathways. However, our current understanding of WJ-MSCs is still very limited since the majority of research has been focused only on their clinical applications. As basic research on WJ-MSCs is still relatively limited, further exploration of regulatory mechanisms of WJ-MSCs by utilizing single-cell analysis and omics methods should be conducted. The new knowledge gained will surely fully harness the great potential of WJ-MSCs in regenerative medicine.

Author Contributions

Conceptualization, Q.W.; methodology, L.M.; software, L.M.; validation, L.M., X.H. and Q.W.; formal analysis, L.M.; investigation, L.M.; resources, Q.W.; data curation, L.M.; writing—original draft preparation, L.M. and X.H.; writing—review and editing, Q.W.; visualization, L.M.; supervision, Q.W.; project administration, Q.W.; funding acquisition, Q.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Macau Science and Technology Development Fund (FDCT) (File number file number 0072/2019/A2 and SKL-QRCM (MUST)-2023-2025).

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. Mcelreavey, K.D.; Irvine, A.I.; Ennis, K.T.; Mclean, W.I. Isolation, culture and characterisation of fibroblast-like cells derived from the Wharton’s jelly portion of human umbilical cord. Biochem. Soc. Trans. 1991, 19, 29S. [Google Scholar] [CrossRef]
  2. Bakhshi, T.; Zabriskie, R.C.; Bodie, S.; Kidd, S.; Ramin, S.; Paganessi, L.A.; Gregory, S.A.; Fung, H.C.; Christopherson, K.W., 2nd. Mesenchymal stem cells from the Wharton’s jelly of umbilical cord segments provide stromal support for the maintenance of cord blood hematopoietic stem cells during long-term ex vivo culture. Transfusion 2008, 48, 2638–2644. [Google Scholar] [CrossRef]
  3. Can, A.; Karahuseyinoglu, S. Concise Review: Human Umbilical Cord Stroma with Regard to the Source of Fetus-Derived Stem Cells. Stem Cells 2007, 25, 2886–2895. [Google Scholar] [CrossRef]
  4. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.C.; Krause, D.S.; Deans, R.J.; Keating, A.; Prockop, D.J.; Horwitz, E.M. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy Position Statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  5. Zhao, G.; Liu, F.; Lan, S.; Li, P.; Wang, L.; Kou, J.; Qi, X.; Fan, R.; Hao, D.; Wu, C.; et al. Large-scale expansion of Wharton’s jelly-derived mesenchymal stem cells on gelatin microbeads, with retention of self-renewal and multipotency characteristics and the capacity for enhancing skin wound healing. Stem Cell Res. Ther. 2015, 6, 38. [Google Scholar] [CrossRef]
  6. Taghizadeh, R.; Cetrulo, K.; Cetrulo, C. Wharton’s Jelly stem cells: Future clinical applications. Placenta 2011, 32, S311–S315. [Google Scholar] [CrossRef]
  7. Kim, D.-W.; Staples, M.; Shinozuka, K.; Pantcheva, P.; Kang, S.-D.; Borlongan, C.V. Wharton’s Jelly-Derived Mesenchymal Stem Cells: Phenotypic Characterization and Optimizing Their Therapeutic Potential for Clinical Applications. Int. J. Mol. Sci. 2013, 14, 11692–11712. [Google Scholar] [CrossRef]
  8. Jaenisch, R.; Bird, A. Epigenetic regulation of gene expression: How the genome integrates intrinsic and environmental signals. Nat. Genet. 2003, 33, 245–254. [Google Scholar] [CrossRef]
  9. Skvortsova, K.; Iovino, N.; Bogdanović, O. Functions and mechanisms of epigenetic inheritance in animals. Nat. Rev. Mol. Cell Biol. 2018, 19, 774–790. [Google Scholar] [CrossRef] [PubMed]
  10. Lunyak, V.V.; Rosenfeld, M.G. Epigenetic regulation of stem cell fate. Hum. Mol. Genet. 2008, 17, R28–R36. [Google Scholar] [CrossRef] [PubMed]
  11. Lambert, S.A.; Jolma, A.; Campitelli, L.F.; Das, P.K.; Yin, Y.; Albu, M.; Chen, X.; Taipale, J.; Hughes, T.R.; Weirauch, M.T. The Human Transcription Factors. Cell 2018, 172, 650–665. [Google Scholar] [CrossRef] [PubMed]
  12. Adachi, K.; Schöler, H.R. Directing reprogramming to pluripotency by transcription factors. Curr. Opin. Genet. Dev. 2012, 22, 416–422. [Google Scholar] [CrossRef]
  13. Graf, T.; Enver, T. Forcing cells to change lineages. Nature 2009, 462, 587–594. [Google Scholar] [CrossRef] [PubMed]
  14. Bird, A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002, 16, 6–21. [Google Scholar] [CrossRef]
  15. Janke, R.; Dodson, A.E.; Rine, J. Metabolism and Epigenetics. Annu. Rev. Cell Dev. Biol. 2015, 31, 473–496. [Google Scholar] [CrossRef]
  16. He, Y.-F.; Li, B.-Z.; Li, Z.; Liu, P.; Wang, Y.; Tang, Q.; Ding, J.; Jia, Y.; Chen, Z.; Li, L.; et al. Tet-Mediated Formation of 5-Carboxylcytosine and Its Excision by TDG in Mammalian DNA. Science 2011, 333, 1303–1307. [Google Scholar] [CrossRef]
  17. Tahiliani, M.; Koh, K.P.; Shen, Y.; Pastor, W.A.; Bandukwala, H.; Brudno, Y.; Agarwal, S.; Iyer, L.M.; Liu, D.R.; Aravind, L.; et al. Conversion of 5-Methylcytosine to 5-Hydroxymethylcytosine in Mammalian DNA by MLL Partner TET1. Science 2009, 324, 930–935. [Google Scholar] [CrossRef]
  18. Okano, M.; Bell, D.W.; Haber, D.A.; Li, E. DNA Methyltransferases Dnmt3a and Dnmt3b Are Essential for De Novo Methylation and Mammalian Development. Cell 1999, 99, 247–257. [Google Scholar] [CrossRef]
  19. Sasajima, Y.; Tanaka, H.; Miyake, S.; Yuasa, Y. A novel EID family member, EID-3, inhibits differentiation and forms a homodimer or heterodimer with EID-2. Biochem. Biophys. Res. Commun. 2005, 333, 969–975. [Google Scholar] [CrossRef]
  20. Luo, L.; Chen, W.-J.; Yin, J.Q.; Xu, R.-X. EID3 directly associates with DNMT3A during transdifferentiation of human umbilical cord mesenchymal stem cells to NPC-like cells. Sci. Rep. 2017, 7, 40463. [Google Scholar] [CrossRef]
  21. Gu, J.; Wang, Y.; Gao, J.; Yuan, S.; Chu, Y.; Li, Y.; Yuan, W.; Wang, X. Tet2 regulates the function of mesenchymal stem cells. Bao Chin. J. Biotechnol. 2019, 35, 142–149. [Google Scholar]
  22. Yang, R.; Yu, T.; Kou, X.; Gao, X.; Chen, C.; Liu, D.; Zhou, Y.; Shi, S. Tet1 and Tet2 maintain mesenchymal stem cell homeostasis via demethylation of the P2rX7 promoter. Nat. Commun. 2018, 9, 2143. [Google Scholar] [CrossRef] [PubMed]
  23. Cakouros, D.; Hemming, S.; Gronthos, K.; Liu, R.; Zannettino, A.; Shi, S.; Gronthos, S. Specific functions of TET1 and TET2 in regulating mesenchymal cell lineage determination. Epigenet. Chromatin 2019, 12, 3. [Google Scholar] [CrossRef]
  24. Mahaira, L.G.; Katsara, O.; Pappou, E.; Iliopoulou, E.G.; Fortis, S.; Antsaklis, A.; Fotinopoulos, P.; Baxevanis, C.N.; Papamichail, M.; Perez, S.A. IGF2BP1 Expression in Human Mesenchymal Stem Cells Significantly Affects Their Proliferation and Is Under the Epigenetic Control of TET1/2 Demethylases. Stem Cells Dev. 2014, 23, 2501–2512. [Google Scholar] [CrossRef]
  25. Yannarelli, G.; Pacienza, N.; Cuniberti, L.; Medin, J.; Davies, J.; Keating, A. Brief Report: The Potential Role of Epigenetics on Multipotent Cell Differentiation Capacity of Mesenchymal Stromal Cells. Stem Cells 2013, 31, 215–220. [Google Scholar] [CrossRef]
  26. Sørensen, A.L.; Timoskainen, S.; West, F.D.; Vekterud, K.; Boquest, A.C.; Ährlund-Richter, L.; Stice, S.L.; Collas, P.; Ghanjati, F.; Santourlidis, S.; et al. Lineage-Specific Promoter DNA Methylation Patterns Segregate Adult Progenitor Cell Types. Stem Cells Dev. 2010, 19, 1257–1266. [Google Scholar] [CrossRef]
  27. Sørensen, A.L.; Jacobsen, B.M.; Reiner, A.H.; Andersen, I.S.; Collas, P. Promoter DNA Methylation Patterns of Differentiated Cells Are Largely Programmed at the Progenitor Stage. Mol. Biol. Cell 2010, 21, 2066–2077. [Google Scholar] [CrossRef]
  28. Govarthanan, K.; Gupta, P.K.; Ramasamy, D.; Kumar, P.; Mahadevan, S.; Verma, R.S. DNA methylation microarray uncovers a permissive methylome for cardiomyocyte differentiation in human mesenchymal stem cells. Genomics 2019, 112, 1384–1395. [Google Scholar] [CrossRef]
  29. Leow, S.C.; Poschmann, J.; Too, P.G.; Yin, J.; Joseph, R.; McFarlane, C.; Dogra, S.; Shabbir, A.; Ingham, P.W.; Prabhakar, S.; et al. The transcription factor SOX6 contributes to the developmental origins of obesity by promoting adipogenesis. Development 2016, 143, 950–961. [Google Scholar] [CrossRef] [PubMed]
  30. Feng, Z.; Yang, Y.; Liu, Z.; Zhao, W.; Huang, L.; Wu, T.; Mu, Y. Integrated analysis of DNA methylome and transcriptome reveals the differences in biological characteristics of porcine mesenchymal stem cells. BMC Genet. 2021, 22, 56. [Google Scholar] [CrossRef]
  31. Bannister, A.J.; Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 2011, 21, 381–395. [Google Scholar] [CrossRef]
  32. Wang, Y.; Chen, T.; Yan, H.; Qi, H.; Deng, C.; Ye, T.; Zhou, S.; Li, F.-R. Role of histone deacetylase inhibitors in the aging of human umbilical cord mesenchymal stem cells. J. Cell. Biochem. 2013, 114, 2231–2239. [Google Scholar] [CrossRef]
  33. Stein, G.S.; Lian, J.B.; van Wijnen, A.J.; Stein, J.L.; Montecino, M.; Javed, A.; Zaidi, S.K.; Young, D.W.; Choi, J.-Y.; Pockwinse, S.M. Runx2 control of organization, assembly and activity of the regulatory machinery for skeletal gene expression. Oncogene 2004, 23, 4315–4329. [Google Scholar] [CrossRef]
  34. Rojas, A.; Aguilar, R.; Henriquez, B.; Lian, J.B.; Stein, J.L.; Stein, G.S.; van Wijnen, A.J.; van Zundert, B.; Allende, M.L.; Montecino, M. Epigenetic Control of the Bone-master Runx2 Gene during Osteoblast-lineage Commitment by the Histone Demethylase JARID1B/KDM5B. J. Biol. Chem. 2015, 290, 28329–28342. [Google Scholar] [CrossRef] [PubMed]
  35. Sepulveda, H.; Aguilar, R.; Prieto, C.P.; Bustos, F.; Aedo, S.; Lattus, J.; van Zundert, B.; Palma, V.; Montecino, M. Epigenetic Signatures at the RUNX2-P1 and Sp7 Gene Promoters Control Osteogenic Lineage Commitment of Umbilical Cord-Derived Mesenchymal Stem Cells. J. Cell. Physiol. 2017, 232, 2519–2527. [Google Scholar] [CrossRef] [PubMed]
  36. Ghazimoradi, M.H.; Farivar, S. The role of DNA demethylation in induction of stem cells. Prog. Biophys. Mol. Biol. 2020, 153, 17–22. [Google Scholar] [CrossRef] [PubMed]
  37. Drela, K.; Sarnowska, A.; Siedlecka, P.; Szablowska-Gadomska, I.; Wielgos, M.; Jurga, M.; Lukomska, B.; Domanska-Janik, K. Low oxygen atmosphere facilitates proliferation and maintains undifferentiated state of umbilical cord mesenchymal stem cells in an hypoxia inducible factor-dependent manner. Cytotherapy 2014, 16, 881–892. [Google Scholar] [CrossRef] [PubMed]
  38. Lee, S.; Park, J.-R.; Seo, M.-S.; Roh, K.-H.; Park, S.-B.; Hwang, J.-W.; Sun, B.; Seo, K.; Lee, Y.-S.; Kang, S.-K.; et al. Histone deacetylase inhibitors decrease proliferation potential and multilineage differentiation capability of human mesenchymal stem cells. Cell Prolif. 2009, 42, 711–720. [Google Scholar] [CrossRef]
  39. Karantzali, E.; Schulz, H.; Hummel, O.; Hubner, N.; Hatzopoulos, A.; Kretsovali, A. Histone deacetylase inhibition accelerates the early events of stem cell differentiation: Transcriptomic and epigenetic analysis. Genome Biol. 2008, 9, R65. [Google Scholar] [CrossRef]
  40. Bhuvanalakshmi, G.; Arfuso, F.; Kumar, A.P.; Dharmarajan, A.; Warrier, S. Epigenetic reprogramming converts human Wharton’s jelly mesenchymal stem cells into functional cardiomyocytes by differential regulation of Wnt mediators. Stem Cell Res. Ther. 2017, 8, 185. [Google Scholar] [CrossRef]
  41. Belame Shivakumar, S.; Bharti, D.; Baregundi Subbarao, R.; Park, J.-M.; Son, Y.-B.; Ullah, I.; Choe, Y.-H.; Lee, H.-J.; Park, B.-W.; Lee, S.-L.; et al. Pancreatic endocrine-like cells differentiated from human umbilical cords Wharton’s jelly mesenchymal stem cells using small molecules. J. Cell. Physiol. 2019, 234, 3933–3947. [Google Scholar] [CrossRef] [PubMed]
  42. Cannell, I.G.; Kong, Y.W.; Bushell, M. How do microRNAs regulate gene expression? Biochem. Soc. Trans. 2008, 36, 1224–1231. [Google Scholar] [CrossRef] [PubMed]
  43. Bartel, D.P. MicroRNAs: Genomics, Biogenesis, Mechanism, and Function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [PubMed]
  44. Balzano, F.; Campesi, I.; Cruciani, S.; Garroni, G.; Bellu, E.; Giudici, S.D.; Angius, A.; Oggiano, A.; Rallo, V.; Capobianco, G.; et al. Epigenetics, Stem Cells, and Autophagy: Exploring a Path Involving miRNA. Int. J. Mol. Sci. 2019, 20, 5091. [Google Scholar] [CrossRef]
  45. Balzano, F.; Cruciani, S.; Basoli, V.; Santaniello, S.; Facchin, F.; Ventura, C.; Maioli, M. MiR200 and miR302: Two Big Families Influencing Stem Cell Behavior. Molecules 2018, 23, 282. [Google Scholar] [CrossRef]
  46. 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] [PubMed]
  47. 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]
  48. 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]
  49. Abidin, S.Z.; Fam, S.-Z.; Chong, C.-E.; Abdullah, S.; Cheah, P.-S.; Nordin, N.; Ling, K.-H. miR-3099 promotes neurogenesis and inhibits astrogliogenesis during murine neural development. Gene 2019, 697, 201–212. [Google Scholar] [CrossRef]
  50. 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]
  51. 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] [PubMed]
  52. 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]
  53. Hsieh, J.-Y.; Huang, T.-S.; Cheng, S.-M.; Lin, W.-S.; Tsai, T.-N.; Lee, O.K.; Wang, H.-W. miR-146a-5p circuitry uncouples cell proliferation and migration, but not differentiation, in human mesenchymal stem cells. Nucleic Acids Res. 2013, 41, 9753–9763. [Google Scholar] [CrossRef] [PubMed]
  54. Lin, Z.; Tang, X.; Wan, J.; Zhang, X.; Liu, C.; Liu, T. Functions and mechanisms of circular RNAs in regulating stem cell differentiation. RNA Biol. 2021, 18, 2136–2149. [Google Scholar] [CrossRef]
  55. Ruan, Z.; Chen, G.; Zhang, R.; Zhu, L. Circular RNA expression profiles during the differentiation of human umbilical cord–derived mesenchymal stem cells into cardiomyocyte-like cells. J. Cell. Physiol. 2019, 234, 16412–16423. [Google Scholar] [CrossRef] [PubMed]
  56. 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]
  57. Wang, C.; Wang, L.; Ding, Y.; Lu, X.; Zhang, G.; Yang, J.; Zheng, H.; Wang, H.; Jiang, Y.; Xu, L. LncRNA Structural Characteristics in Epigenetic Regulation. Int. J. Mol. Sci. 2017, 18, 2659. [Google Scholar] [CrossRef]
  58. Xie, Z.-Y.; Wang, P.; Wu, Y.-F.; Shen, H.-Y. Long non-coding RNA: The functional regulator of mesenchymal stem cells. World J. Stem Cells 2019, 11, 167–179. [Google Scholar] [CrossRef]
  59. Liu, F.; Song, D.-Y.; Huang, J.; Yang, H.-Q.; Di You, D.; Ni, J.-D. Long non-coding RNA CIR inhibits chondrogenic differentiation of mesenchymal stem cells by epigenetically suppressing ATOH8 via methyltransferase EZH2. Mol. Med. 2021, 27, 12. [Google Scholar] [CrossRef]
  60. Al-Obaide, M.; Ishmakej, A.; Brown, C.; Mazzella, M.; Agosta, P.; Perez-Cruet, M.; Chaudhry, G.R. The potential role of integrin alpha 6 in human mesenchymal stem cells. Front. Genet. 2022, 13, 968228. [Google Scholar] [CrossRef]
  61. Tsai, C.-C.; Su, P.-F.; Huang, Y.-F.; Yew, T.-L.; Hung, S.-C. Oct4 and Nanog Directly Regulate Dnmt1 to Maintain Self-Renewal and Undifferentiated State in Mesenchymal Stem Cells. Mol. Cell 2012, 47, 169–182. [Google Scholar] [CrossRef]
  62. Vaquerizas, J.M.; Kummerfeld, S.K.; Teichmann, S.A.; Luscombe, N.M. A census of human transcription factors: Function, expression and evolution. Nat. Rev. Genet. 2009, 10, 252–263. [Google Scholar] [CrossRef]
  63. Fong, C.-Y.; Chak, L.-L.; Biswas, A.; Tan, J.-H.; Gauthaman, K.; Chan, W.-K.; Bongso, A. Human Wharton’s Jelly Stem Cells Have Unique Transcriptome Profiles Compared to Human Embryonic Stem Cells and Other Mesenchymal Stem Cells. Stem Cell Rev. Rep. 2011, 7, 1–16. [Google Scholar] [CrossRef]
  64. Rodda, D.J.; Chew, J.-L.; Lim, L.-H.; Loh, Y.-H.; Wang, B.; Ng, H.-H.; Robson, P. Transcriptional Regulation of Nanog by OCT4 and SOX2. J. Biol. Chem. 2005, 280, 24731–24737. [Google Scholar] [CrossRef]
  65. Loh, Y.-H.; Wu, Q.; Chew, J.-L.; Vega, V.B.; Zhang, W.; Chen, X.; Bourque, G.; George, J.; Leong, B.; Liu, J.; et al. The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells. Nat. Genet. 2006, 38, 431–440. [Google Scholar] [CrossRef]
  66. Gao, L.R.; Zhang, N.K.; Ding, Q.A.; Chen, H.Y.; Hu, X.; Jiang, S.; Li, T.C.; Chen, Y.; Wang, Z.G.; Ye, Y.; et al. Common Expression of Stemness Molecular Markers and Early Cardiac Transcription Factors in Human Wharton’s Jelly-Derived Mesenchymal Stem Cells and Embryonic Stem Cells. Cell Transplant. 2013, 22, 1883–1900. [Google Scholar] [CrossRef] [PubMed]
  67. Bharti, D.; Shivakumar, S.B.; Ullah, I.; Subbarao, R.B.; Park, J.-S.; Lee, S.-L.; Park, B.-W.; Rho, G.-J. Comparative analysis of human Wharton’s jelly mesenchymal stem cells derived from different parts of the same umbilical cord. Cell Tissue Res. 2018, 372, 51–65. [Google Scholar] [CrossRef]
  68. Ng, H.-H.; Surani, M.A. The transcriptional and signalling networks of pluripotency. Nature 2011, 13, 490–496. [Google Scholar] [CrossRef] [PubMed]
  69. Greco, S.J.; Liu, K.; Rameshwar, P. Functional Similarities Among Genes Regulated by Oct4 in Human Mesenchymal and Embryonic Stem Cells. Stem Cells 2007, 25, 3143–3154. [Google Scholar] [CrossRef] [PubMed]
  70. Weiss, M.L.; Medicetty, S.; Bledsoe, A.R.; Rachakatla, R.S.; Choi, M.; Merchav, S.; Luo, Y.; Rao, M.S.; Velagaleti, G.; Troyer, D. Human Umbilical Cord Matrix Stem Cells: Preliminary Characterization and Effect of Transplantation in a Rodent Model of Parkinson’s Disease. Stem Cells 2006, 24, 781–792. [Google Scholar] [CrossRef]
  71. Gil-Kulik, P.; Świstowska, M.; Krzyżanowski, A.; Petniak, A.; Kwaśniewska, A.; Płachno, B.J.; Galkowski, D.; Bogucka-Kocka, A.; Kocki, J. Evaluation of the Impact of Pregnancy-Associated Factors on the Quality of Wharton’s Jelly-Derived Stem Cells Using SOX2 Gene Expression as a Marker. Int. J. Mol. Sci. 2022, 23, 7630. [Google Scholar] [CrossRef] [PubMed]
  72. Schaal, C.M.; Bora-Singhal, N.; Kumar, D.M.; Chellappan, S.P. Regulation of Sox2 and stemness by nicotine and electronic-cigarettes in non-small cell lung cancer. Mol. Cancer 2018, 17, 149. [Google Scholar] [CrossRef] [PubMed]
  73. Świstowska, M.; Gil-Kulik, P.; Krzyżanowski, A.; Bielecki, T.; Czop, M.; Kwaśniewska, A.; Kocki, J. Potential Effect of SOX2 on the Cell Cycle of Wharton’s Jelly Stem Cells (WJSCs). Oxidative Med. Cell. Longev. 2019, 2019, 5084689. [Google Scholar] [CrossRef] [PubMed]
  74. Tan, P.Y.; Chang, C.W.; Duan, K.; Poidinger, M.; Ng, K.L.; Chong, Y.S.; Gluckman, P.D.; Stünkel, W. E2F1 Orchestrates Transcriptomics and Oxidative Metabolism in Wharton’s Jelly-Derived Mesenchymal Stem Cells from Growth-Restricted Infants. PLoS ONE 2016, 11, e0163035. [Google Scholar] [CrossRef]
  75. Jeong, B.-C.; Kang, I.-H.; Hwang, Y.-C.; Kim, S.-H.; Koh, J.-T. MicroRNA-194 reciprocally stimulates osteogenesis and inhibits adipogenesis via regulating COUP-TFII expression. Cell Death Dis. 2014, 5, e1532. [Google Scholar] [CrossRef]
  76. Gao, G.; Zhang, X.-F.; Hubbell, K.; Cui, X. NR2F2 regulates chondrogenesis of human mesenchymal stem cells in bioprinted cartilage. Biotechnol. Bioeng. 2017, 114, 208–216. [Google Scholar] [CrossRef]
  77. Ma, L.; Huang, M.; Liao, X.; Cai, X.; Wu, Q. NR2F2 Regulates Cell Proliferation and Immunomodulation in Whartons’ Jelly Stem Cells. Genes 2022, 13, 1458. [Google Scholar] [CrossRef]
  78. Velletri, T.; Xie, N.; Wang, Y.; Huang, Y.; Yang, Q.; Chen, X.; Chen, Q.; Shou, P.; Gan, Y.; Cao, G.; et al. P53 functional abnormality in mesenchymal stem cells promotes osteosarcoma development. Cell Death Dis. 2016, 7, e2015. [Google Scholar] [CrossRef]
  79. Armesilla-Diaz, A.; Elvira, G.; Silva, A. p53 regulates the proliferation, differentiation and spontaneous transformation of mesenchymal stem cells. Exp. Cell Res. 2009, 315, 3598–3610. [Google Scholar] [CrossRef]
  80. Velletri, T.; Huang, Y.; Wang, Y.; Li, Q.; Hu, M.; Xie, N.; Yang, Q.; Chen, X.; Chen, Q.; Shou, P.; et al. Loss of p53 in mesenchymal stem cells promotes alteration of bone remodeling through negative regulation of osteoprotegerin. Cell Death Differ. 2021, 28, 156–169. [Google Scholar] [CrossRef] [PubMed]
  81. Wang, B.; Wang, L.; Mao, J.; Wen, H.; Xu, L.; Ren, Y.; Du, H.; Yang, H. Mouse bone marrow mesenchymal stem cells with distinct p53 statuses display differential characteristics. Mol. Med. Rep. 2020, 21, 2051–2062. [Google Scholar] [CrossRef]
  82. He, Y.; de Castro, L.F.; Shin, M.H.; Dubois, W.; Yang, H.H.; Jiang, S.; Mishra, P.J.; Ren, L.; Gou, H.; Lal, A.; et al. p53 Loss Increases the Osteogenic Differentiation of Bone Marrow Stromal Cells. Stem Cells 2015, 33, 1304–1319. [Google Scholar] [CrossRef]
  83. Artigas, N.; Gámez, B.; Cubillos-Rojas, M.; Diego, C.S.-D.; Valer, J.A.; Pons, G.; Rosa, J.L.; Ventura, F. p53 inhibits SP7/Osterix activity in the transcriptional program of osteoblast differentiation. Cell Death Differ. 2017, 24, 2022–2031. [Google Scholar] [CrossRef]
  84. Zhou, W.; Gross, K.M.; Kuperwasser, C. Molecular regulation of Snai2 in development and disease. J. Cell Sci. 2019, 132, jcs235127. [Google Scholar] [CrossRef]
  85. Tang, Y.; Weiss, S.J. Snail/Slug-YAP/TAZ complexes cooperatively regulate mesenchymal stem cell function and bone formation. Cell Cycle 2017, 16, 399–405. [Google Scholar] [CrossRef] [PubMed]
  86. Torreggiani, E.; Lisignoli, G.; Manferdini, C.; Lambertini, E.; Penolazzi, L.; Vecchiatini, R.; Gabusi, E.; Chieco, P.; Facchini, A.; Gambari, R.; et al. Role of Slug transcription factor in human mesenchymal stem cells. J. Cell. Mol. Med. 2012, 16, 740–751. [Google Scholar] [CrossRef]
  87. Hang, H.; Yu, Y.; Wu, N.; Huang, Q.; Xia, Q.; Bian, J. Induction of Highly Functional Hepatocytes from Human Umbilical Cord Mesenchymal Stem Cells by HNF4α Transduction. PLoS ONE 2014, 9, e104133. [Google Scholar] [CrossRef]
  88. Yu, Y.; Zhang, Q.; Wu, N.; Xia, L.; Cao, J.; Xia, Q.; Zhao, J.; Zhang, J.; Hang, H. HNF4α overexpression enhances the therapeutic potential of umbilical cord mesenchymal stem/stromal cells in mice with acute liver failure. FEBS Lett. 2022, 596, 3176–3190. [Google Scholar] [CrossRef] [PubMed]
  89. Sharma, M.; Pruitt, K. Wnt Pathway: An Integral Hub for Developmental and Oncogenic Signaling Networks. Int. J. Mol. Sci. 2020, 21, 8018. [Google Scholar] [CrossRef]
  90. Ling, L.; Nurcombe, V.; Cool, S.M. Wnt signaling controls the fate of mesenchymal stem cells. Gene 2009, 433, 1–7. [Google Scholar] [CrossRef] [PubMed]
  91. Batsali, A.K.; Pontikoglou, C.; Koutroulakis, D.; Pavlaki, K.I.; Damianaki, A.; Mavroudi, I.; Alpantaki, K.; Kouvidi, E.; Kontakis, G.; Papadaki, H.A. Differential expression of cell cycle and WNT pathway-related genes accounts for differences in the growth and differentiation potential of Wharton’s jelly and bone marrow-derived mesenchymal stem cells. Stem Cell Res. Ther. 2017, 8, 102. [Google Scholar] [CrossRef]
  92. Liu, Z.; Li, T.; Liu, Y.; Jia, Z.; Li, Y.; Zhang, C.; Chen, P.; Ma, K.; Affara, N.; Zhou, C. WNT signaling promotes Nkx2.5 expression and early cardiomyogenesis via downregulation of Hdac1. Biochim. Biophys. Acta BBA Mol. Cell Res. 2009, 1793, 300–311. [Google Scholar] [CrossRef]
  93. Biggs, M.J.; Richards, R.G.; Gadegaard, N.; Wilkinson, C.D.; Oreffo, R.O.; Dalby, M.J. The use of nanoscale topography to modulate the dynamics of adhesion formation in primary osteoblasts and ERK/MAPK signalling in STRO-1+ enriched skeletal stem cells. Biomaterials 2009, 30, 5094–5103. [Google Scholar] [CrossRef] [PubMed]
  94. He, D.; Wu, H.; Xiang, J.; Ruan, X.; Peng, P.; Ruan, Y.; Chen, Y.-G.; Wang, Y.; Yu, Q.; Zhang, H.; et al. Gut stem cell aging is driven by mTORC1 via a p38 MAPK-p53 pathway. Nat. Commun. 2020, 11, 37. [Google Scholar] [CrossRef]
  95. Berlier, J.L.; Rigutto, S.; Valle, A.D.; Lechanteur, J.; Soyfoo, M.S.; Gangji, V.; Rasschaert, J. Adenosine Triphosphate Prevents Serum Deprivation-Induced Apoptosis in Human Mesenchymal Stem Cells via Activation of the MAPK Signaling Pathways. Stem Cells 2015, 33, 211–218. [Google Scholar] [CrossRef] [PubMed]
  96. Lu, J.; Wang, Q.-H.; Huang, L.-H.; Dong, H.-Y.; Lin, L.-J.; Tan, J.-M. Correlation of CDC42 Activity with Cell Proliferation and Palmitate-Mediated Cell Death in Human Umbilical Cord Wharton’s Jelly Derived Mesenchymal Stromal Cells. Stem Cells Dev. 2017, 26, 1283–1292. [Google Scholar] [CrossRef] [PubMed]
  97. Chang, C.-H.; Lin, Y.-L.; Tyan, Y.-S.; Chiu, Y.-H.; Liang, Y.-H.; Chen, C.-P.; Wu, J.-C.; Wang, H.-S. Interleukin-1β-induced matrix metalloproteinase-3 via ERK1/2 pathway to promote mesenchymal stem cell migration. PLoS ONE 2021, 16, e0252163. [Google Scholar] [CrossRef]
  98. Wang, Y.; Liu, Y.; Fan, Z.; Liu, D.; Wang, F.; Zhou, Y. IGFBP2 enhances adipogenic differentiation potentials of mesenchymal stem cells from Wharton’s jelly of the umbilical cord via JNK and Akt signaling pathways. PLoS ONE 2017, 12, e0184182. [Google Scholar] [CrossRef]
  99. Hiew, V.V.; Teoh, P.L. Collagen Modulates the Biological Characteristics of WJ-MSCs in Basal and Osteoinduced Conditions. Stem Cells Int. 2022, 2022, 2116367. [Google Scholar] [CrossRef]
  100. Zavala, G.; Prieto, C.P.; Villanueva, A.A.; Palma, V. Sonic hedgehog (SHH) signaling improves the angiogenic potential of Wharton’s jelly-derived mesenchymal stem cells (WJ-MSC). Stem Cell Res. Ther. 2017, 8, 203. [Google Scholar] [CrossRef]
  101. Goyal, U.; Ta, M. A novel role of vitronectin in promoting survival of mesenchymal stem cells under serum deprivation stress. Stem Cell Res. Ther. 2020, 11, 181. [Google Scholar] [CrossRef]
  102. Ju, D.; Van Thao, D.; Lu, C.; Ali, A.; Shibu, M.A.; Chen, R.; Day, C.H.; Shih, T.; Tsai, C.; Kuo, C.; et al. Protective effects of CHIP overexpression and Wharton’s jelly mesenchymal-derived stem cell treatment against streptozotocin-induced neurotoxicity in rats. Environ. Toxicol. 2022, 37, 1979–1987. [Google Scholar] [CrossRef]
  103. Jang, I.K.; Jung, H.J.; Noh, O.K.; Lee, D.; Lee, K.C.; Park, J.E. B7-H1-mediated immunosuppressive properties in human mesenchymal stem cells are mediated by STAT-1 and not PI3K/Akt signaling. Mol. Med. Rep. 2018, 18, 1842–1848. [Google Scholar] [CrossRef] [PubMed]
  104. Huang, D.; Xiao, F.; Hao, H.; Hua, F.; Luo, Z.; Huang, Z.; Li, Q.; Chen, S.; Cheng, X.; Zhang, X.; et al. JARID1B promotes colorectal cancer proliferation and Wnt/β-catenin signaling via decreasing CDX2 level. Cell Commun. Signal. 2020, 18, 169. [Google Scholar] [CrossRef]
  105. Uzan, B.; Figeac, F.; Portha, B.; Movassat, J. Mechanisms of KGF Mediated Signaling in Pancreatic Duct Cell Proliferation and Differentiation. PLoS ONE 2009, 4, e4734. [Google Scholar] [CrossRef]
  106. Xu, Y.; Hong, Y.; Xu, M.; Ma, K.; Fu, X.; Zhang, M.; Wang, G. Role of Keratinocyte Growth Factor in the Differentiation of Sweat Gland-Like Cells From Human Umbilical Cord-Derived Mesenchymal Stem Cells. Stem Cells Transl. Med. 2016, 5, 106–116. [Google Scholar] [CrossRef]
  107. Bracci-Laudiero, L.; De Stefano, M.E. NGF in Early Embryogenesis, Differentiation, and Pathology in the Nervous and Immune Systems. In Current Topics in Behavioral Neurosciences; Springer: Berlin/Heidelberg, Germany, 2015; Volume 29, pp. 125–152. [Google Scholar] [CrossRef]
  108. Morys, J.; Borkowska, P.; Zielinska, A.; Kowalski, J. Study of the influence of NGF-β gene overexpression in human mesenchymal stem cells on the expression level of SOX1 and neural pathway genes. Mol. Biol. Rep. 2022, 49, 4435–4441. [Google Scholar] [CrossRef]
  109. Borkowska, P.; Morys, J.; Zielinska, A.; Sadlocha, M.; Kowalski, J. Survival and Neurogenesis-Promoting Effects of the Co-Overexpression of BCLXL and BDNF Genes on Wharton’s Jelly-Derived Mesenchymal Stem Cells. Life 2022, 12, 1406. [Google Scholar] [CrossRef]
  110. Langhnoja, J.; Buch, L.; Pillai, P. Potential role of NGF, BDNF, and their receptors in oligodendrocytes differentiation from neural stem cell: An in vitro study. Cell Biol. Int. 2020, 45, 432–446. [Google Scholar] [CrossRef]
  111. Bustos, F.; Sepúlveda, H.; Prieto, C.P.; Carrasco, M.; Díaz, L.; Palma, J.; Lattus, J.; Montecino, M.; Palma, V. Runt-Related Transcription Factor 2 Induction During Differentiation of Wharton’s Jelly Mesenchymal Stem Cells to Osteoblasts Is Regulated by Jumonji AT-Rich Interactive Domain 1B Histone Demethylase. Stem Cells 2017, 35, 2430–2441. [Google Scholar] [CrossRef]
  112. Wu, M.; Chen, G.; Li, Y.-P. TGF-β and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease. Bone Res. 2016, 4, 16009. [Google Scholar] [CrossRef] [PubMed]
  113. Celil, A.B.; Campbell, P.G. BMP-2 and Insulin-like Growth Factor-I Mediate Osterix (Osx) Expression in Human Mesenchymal Stem Cells via the MAPK and Protein Kinase D Signaling Pathways. J. Biol. Chem. 2005, 280, 31353–31359. [Google Scholar] [CrossRef]
  114. Ono, M.; Inkson, C.A.; Kilts, T.M.; Young, M.F. WISP-1/CCN4 regulates osteogenesis by enhancing BMP-2 activity. J. Bone Miner. Res. 2011, 26, 193–208. [Google Scholar] [CrossRef]
  115. Fallah, A.; Alipour, M.; Jamali, Z.; Farjadfar, A.; Roshangar, L.; Nasr, M.P.; Hashemi, P.; Aghazadeh, 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]
  116. He, S.; Yang, S.; Zhang, Y.; Li, X.; Gao, D.; Zhong, Y.; Cao, L.; Ma, H.; Liu, Y.; Li, G.; et al. LncRNA ODIR1 inhibits osteogenic differentiation of hUC-MSCs through the FBXO25/H2BK120ub/H3K4me3/OSX axis. Cell Death Dis. 2019, 10, 947. [Google Scholar] [CrossRef]
  117. Yang, L.; Bin, Z.; Hui, S.; Rong, L.; You, B.; Wu, P.; Han, X.; Qian, H.; Xu, W. The Role of CDR1as in Proliferation and Differentiation of Human Umbilical Cord-Derived Mesenchymal Stem Cells. Stem Cells Int. 2019, 2019, 2316834. [Google Scholar] [CrossRef] [PubMed]
  118. Su, C.; Zheng, X.; He, Y.; Long, L.; Chen, W. Transcriptomic profiling and functional prediction reveal aberrant expression of circular RNAs during osteogenic differentiation in human umbilical cord mesenchymal stromal cells. Sci. Rep. 2021, 11, 19881. [Google Scholar] [CrossRef]
Figure 1. Histone modifications in differentiation of WJ-MSCs.
Figure 1. Histone modifications in differentiation of WJ-MSCs.
Ijms 24 12909 g001
Figure 2. Noncoding RNAs affect cell differentiation of WJ-MSCs.
Figure 2. Noncoding RNAs affect cell differentiation of WJ-MSCs.
Ijms 24 12909 g002
Figure 3. Wnt signaling pathway influences differentiation of WJ-MSCs.
Figure 3. Wnt signaling pathway influences differentiation of WJ-MSCs.
Ijms 24 12909 g003
Figure 4. MAPK pathway directs WJ-MSCs into osteoblasts.
Figure 4. MAPK pathway directs WJ-MSCs into osteoblasts.
Ijms 24 12909 g004
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

Ma, L.; He, X.; Wu, Q. The Molecular Regulatory Mechanism in Multipotency and Differentiation of Wharton’s Jelly Stem Cells. Int. J. Mol. Sci. 2023, 24, 12909. https://doi.org/10.3390/ijms241612909

AMA Style

Ma L, He X, Wu Q. The Molecular Regulatory Mechanism in Multipotency and Differentiation of Wharton’s Jelly Stem Cells. International Journal of Molecular Sciences. 2023; 24(16):12909. https://doi.org/10.3390/ijms241612909

Chicago/Turabian Style

Ma, Li, Xuguang He, and Qiang Wu. 2023. "The Molecular Regulatory Mechanism in Multipotency and Differentiation of Wharton’s Jelly Stem Cells" International Journal of Molecular Sciences 24, no. 16: 12909. https://doi.org/10.3390/ijms241612909

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