Next Article in Journal
Molecular Descriptors Property Prediction Using Transformer-Based Approach
Previous Article in Journal
Hmgb1 Silencing in the Amygdala Inhibits Pain-Related Behaviors in a Rat Model of Neuropathic Pain
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mesodermal Derivatives of Pluripotent Stem Cells Route to Scarless Healing

by
Yulia Suzdaltseva
* and
Sergey L. Kiselev
Department of Epigenetics, Vavilov Institute of General Genetics of the Russian Academy of Sciences, 119333 Moscow, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(15), 11945; https://doi.org/10.3390/ijms241511945
Submission received: 16 June 2023 / Revised: 7 July 2023 / Accepted: 24 July 2023 / Published: 26 July 2023
(This article belongs to the Section Molecular Biology)

Abstract

:
Scar formation during normal tissue regeneration in adults may result in noticeable cosmetic and functional defects and have a significant impact on the quality of life. In contrast, fetal tissues in the mid-gestation period are known to be capable of complete regeneration with the restitution of the initial architecture, organization, and functional activity. Successful treatments that are targeted to minimize scarring can be realized by understanding the cellular and molecular mechanisms of fetal wound regeneration. However, such experiments are limited by the inaccessibility of fetal material for comparable studies. For this reason, the molecular mechanisms of fetal regeneration remain unknown. Mesenchymal stromal cells (MSCs) are central to tissue repair because the molecules they secrete are involved in the regulation of inflammation, angiogenesis, and remodeling of the extracellular matrix. The mesodermal differentiation of human pluripotent stem cells (hPSCs) recapitulates the sequential steps of embryogenesis in vitro and provides the opportunity to generate the isogenic cell models of MSCs corresponding to different stages of human development. Further investigation of the functional activity of cells from stromal differon in a pro-inflammatory microenvironment will procure the molecular tools to better understand the fundamental mechanisms of fetal tissue regeneration. Herein, we review recent advances in the generation of clonal precursors of primitive mesoderm cells and MSCs from hPSCs and discuss critical factors that determine the functional activity of MSCs-like cells in a pro-inflammatory microenvironment in order to identify therapeutic targets for minimizing scarring.

1. Introduction

Wound healing is a well-orchestrated process that occurs in three overlapping phases (inflammation, proliferation, and remodeling), the sequential path of which is supported by the spatial and temporal synchronization of cellular and paracrine activity. In adults, the remodeling phase results in scar formation with a partial loss of the structure and functional activity of the tissue [1]. At the same time, the impact of certain factors can interfere with successful healing and lead to the development of chronic inflammation, keloids, and hypertrophic scars [2,3]. Patients with scars suffer from functional complications and psychological problems and represent a great burden on the healthcare system. Although the cellular and molecular mechanisms underlying wound healing and scar formation are well described, and modern medications and certain techniques allow wounds to heal faster with less scarring than before, truly effective anti-scarring treatments still do not exist [4,5]. Conversely, mammalian fetal tissues throughout the first and second trimesters of development are known to heal rapidly without scar formation [6,7]. This phenomenon has been demonstrated experimentally in animal models including lamb [8], rabbit [9], rat [10], and mouse [11]. Furthermore, the experimental model for human fetal skin wounds was described by Lorenz in 1992 [12]. These studies demonstrated that damaged fetal tissues were able to restore their original structure with normal three-dimensional collagen architecture, complete epithelial covering with normal differentiation, and epidermal appendages [9,13]. Several mechanisms have been proposed to be involved in fetal scarless wound healing, including the influence of the sterile uterine environment, the composition of secreted growth factors and extracellular matrix, the specific fibroblast phenotype, and a reduced inflammatory response [14]. Obviously, the complete regeneration of fetal tissues occurs due to the coordinated action of these mechanisms, the regulation of which is disturbed in fibrosis. Since such studies are strongly limited by ethical problems in relation to humans, the physiological, cellular, and molecular mechanisms of fetal scarless regeneration remain poorly understood.
Comparative studies of the molecular mechanisms of tissue regeneration in adults and fetuses could identify a specific molecular or cellular target in the scarring pathway to generate a new drug treatment for scars, which could potentially improve the lives of many patients. Human pluripotent stem cells (hPSCs) provide the possibility to generate isogenic cellular models corresponding to different stages of human development due to their ability to reproduce the processes of human embryogenesis in vitro. Implementation of this approach seems to be the optimal solution for this purpose.
Tissue regeneration is impossible without the contribution of mesenchymal stromal cells (MSCs) since the restoration of the parenchyma occurs on the molecular framework they generate. Furthermore, the immunomodulatory activity of MSCs has been shown to contribute to the reduction of acute and chronic inflammation. MSCs perform a regulatory function at the site of damage due to their ability to dynamically change their phenotypes, functional activity, expression, and secretory profile under external signals from the microenvironment, thus promoting successful healing [15]. Scarless healing of fetal tissues is logically assumed to occur with the contribution of fetal MSCs (mesodermal progenitors), which phenotypically and functionally differ from those in adults. However, the differences in the molecular mechanisms involving soluble factors and receptor and adhesive intercellular communications, which determine the functional activity of mesodermal cells in a pro-inflammatory microenvironment in fetuses and adults, remain virtually unknown.
Below, we discuss the known mechanisms by which adult MSCs promote wound healing with a focus on the inflammatory response, extracellular matrix (ECM) synthesis, and remodeling. This is followed by a review of the physiological functions of MSCs in the regulation of tissue regeneration described in animal research reports and clinical studies. Probable pathways and molecules involved in physiological, cellular, and molecular mechanisms of fetal tissue regeneration that differ from adults will also be highlighted. Finally, recent advances in the generation of isogenic cellular models corresponding to different stages of human development are described, and new pathways to investigate phenotypic and functional features of adult and fetal mesodermal cells to elucidate the mechanisms responsible for scarless healing are proposed.

2. MSCs-Dependent Mechanisms Involved in Regulation of Inflammation and Tissue Regeneration in Adults

Adult MSCs are now widely accepted to contribute to tissue repair due to their multiple paracrine and immunomodulatory effects. MSCs perform a regulatory function in all phases of wound healing due to their ability to dynamically change their expression and secretory profile under the influence of signals from the microenvironment. The cytokines, chemokines, growth factors, and extracellular vesicles secreted by MSCs are involved in the regulation of intracellular signaling cascades that stimulate angiogenesis, recruitment of cells to the site of inflammation, and remodeling of the extracellular matrix.
MSCs can be isolated and successfully expanded in vitro from different tissue sources. Cultured MSCs are characterized by their adherence to plastic, the expression of the specific surface markers CD73, CD90, and CD105, and their capacity to differentiate into adipocytes, chondrocytes, and osteoblasts [16].
During the early inflammatory response, the secretion of pro-inflammatory cytokines interleukin-1 (IL-1), IL-6, and IL-8 is upregulated in MSCs through Toll-like receptor (TLR) activation. However, TLR stimulation may polarize MSCs into differently acting phenotypes. TLR4-primed MSCs are characterized by the expression of transforming growth factor beta (TGF-beta), indoleamine 2,3-dioxygenase (IDO), prostaglandin E2 (PGE2), and collagen deposition, while TLR3 activation leads to elevated secretion of chemokine (C-C motif) ligand 10 (CCL10), CCL5 (RANTES), IL-10, and fibronectin [17]. MSCs have also been shown to exhibit immunomodulatory effects when interacting with immune cells [18,19]. MSCs possess the ability to inhibit T cell proliferation in mixed lymphocyte reactions [20]. They also suppress the differentiation and maturation of dendritic cells (DCs) and promote their polarization towards the anti-inflammatory M2 phenotype [21]. MSCs mediate the induction of a regulatory phenotype in conventional T cells (Treg) [22,23]. We and others have demonstrated that MSCs can induce functional changes in immune cells and thus have the ability to regulate the balance between pro-inflammatory and anti-inflammatory factors produced, generating a cytokine microenvironment necessary for successful healing [24,25,26]. This immunomodulatory activity of MSCs controls the course of inflammation and its transition to the subsequent proliferative phase of tissue repair. The granulation tissue consisting of a provisional extracellular matrix is characterized by intense angiogenesis. MSCs secrete proangiogenic factors (hypoxia-inducible factor (HIF), vascular endothelial growth factor (VEGF), angiopoietin, monocyte chemoattractant protein (MCP), hepatocyte growth factor (HGF), etc.), which promote migration and proliferation of endothelial cells [27,28,29]. MSCs also support the stabilization and maturation of newly formed vessels [30,31,32]. Extracellular matrix proteins such as fibronectin, collagen, glycosaminoglycans, and proteoglycans synthesized by MSCs are also involved in the process of granulation tissue formation [33,34,35]. During the remodeling phase, metalloproteinases secreted by MSCs regulate the reorganization of collagen fibrils [36,37,38]. MSCs can also stimulate the migration and proliferation of keratinocytes due to the secretion of keratinocyte growth factor (KGF) [39]. The differentiation of MSCs into myofibroblasts contributes to wound contraction [40,41].

3. Effectiveness of Adult MSCs in Tissue Repair In Vivo

Functional and phenotypic peculiarities of MSCs have been widely characterized in vitro. However, evidence that these data can be extrapolated to the properties of endogenous MSCs in vivo is still poor and fragmentary.
Nevertheless, Kramann et al. used genetic lineage fate tracing in murine models to demonstrate that, upon injury, tissue-resident Gli1+ MSCs are recruited from the endosteal and perivascular niche to become fibrosis-driving myofibroblasts and substantially contribute to organ fibrosis. Genetic ablation of Gli1+ cells substantially ameliorated bone marrow, kidney, and heart fibrosis [42,43].
Preclinical studies in animal models and clinical trials have also demonstrated the therapeutic applicability of MSCs, confirming their crucial role in tissue repair. MSCs have been shown to promote the healing of full-thickness wounds in mice [44], rats [45,46,47], rabbits [48], and sheep [49]. Reduced synovitis and articular destruction were observed in mice with collagen-induced rheumatoid arthritis after systemic delivery of MSCs in comparison with a control group [50,51]. MSCs were shown to improve left ventricle function, increase vascular density, and decrease scar size, left ventricle stroke volumes, and ejection fractions in a rat model of myocardial infarction [52,53]. MSC treatment effectively improved renal function, resulting in reduced albuminuria, glomerular injury, and renal fibrosis, and promoted wound healing in streptozotocin-induced diabetic mice [54,55].
Currently, the therapeutic potential of human MSCs has been widely investigated in clinical trials. These studies show promising results in MSC treatment of different diseases including immunologic disorders, cardiovascular diseases, diabetes mellitus, chronic wounds, burns, aging frailty, etc. A randomized study conducted by us demonstrated the ability of MSCs to reduce chronic inflammation in long-term nonhealing wounds [56]. Other published clinical trial data demonstrated that the administration of MSCs had beneficial effects resulting in accelerated healing and improved functional capacities and quality of life. The potent mechanisms of MSC-based therapy include stimulation of neovascularization, immunomodulatory activity, reduction of fibrosis, and stimulation of endogenous tissue regeneration acting in concert [15,56,57].

4. Molecular and Cellular Mechanisms of Fetal Tissue Regeneration

Physiological, cellular, and molecular mechanisms of fetal tissue regeneration are currently poorly understood because the fetus develops in the isolated environment of the uterus. Nevertheless, several studies of wound repair in animal embryos have identified histological differences between wound healing in fetuses and adults. Healing fetal wounds were shown to contain significantly fewer inflammatory cells compared to adult wounds [14,58]. It was demonstrated that reduced migration of circulating neutrophils into the fetal wound is associated with downregulation of adhesion molecule expression [59,60]. Wulff BC et al., 2012, observed that in mice, dermal mast cells in scarless wounds generated at embryonic day 15 (E15) were fewer in number, less mature, and did not degranulate in response to wounding as effectively as mast cells of fibrotic wounds made at embryonic day 18 (E18) [61]. Other researchers noted that murine fetal wounds had fewer activated macrophages, which appeared and disappeared more rapidly compared to adult wounds. They also found the absence of B cells at the fetal wound site [62].
Inflammatory cells attracted to healing fetal wounds generate a specific microenvironment shown to have a diminished IL-6 and IL-8 response compared to that in adults [63,64]. During fetal scarless wound healing, the ratio of TGF-β3/TGF-β1 isoforms was also shown to be much higher than that in adults [65,66]. In addition, fetal wounds are hypoxic compared to adult wounds and have lower levels of VEGF and reduced vascularity [67]. Therefore, the inflammatory response to injury is significantly reduced in fetal wounds.
ECM composition in fetal and adult wounds undergoing healing also differs. Studies have demonstrated that fetal wounds predominantly regenerate with the deposition of type III collagen, which exhibits a basket-weave network of fine fibrils in a glycosaminoglycan-rich matrix resembling the collagen architecture of uninjured skin. In contrast, after wounding of postnatal skin, the disorganized deposition of type I collagen fibers leads to scarring [68,69,70]. Temporal differences in glycoprotein composition were also found to occur during wound healing in fetuses and adults. Tenascin and fibromodulin are deposited more rapidly in fetal wounds compared to adult wounds [71,72,73]. In contrast, decorin expression is downregulated in fetal wounds and increases rapidly with increasing gestational age [74]. Chondroitin sulfate has a diffuse distribution in the ECM during collagen fibril formation in fetal wounds but is not detected at this stage in adult wounds, except for a very small area immediately below the regenerated basement membrane [71]. Fetal wound fluid was also found to have the prolonged presence of significantly elevated levels of hyaluronic acid, up to 3 weeks, compared with adult wounds where it disappeared within 1 week [75,76]. In addition, a higher ratio of matrix metalloproteinases (MMPs) to their tissue-derived inhibitors (TIMPs) in scarless wounds compared to scarring wounds facilitates the migration of fetal cells and promotes extracellular matrix turnover [77].
The ECM not only represents the tridimensional structural environment of tissues but also may regulate the dynamic interplay between resident and recruited inflammatory cells through the conversion of mechanical stimuli into biochemical signals [78]. MSCs were shown to express various components of ECM, including collagens, glycoproteins, and proteoglycans [33,79,80]. Therefore, changes in MSC populations, phenotype, and functional activity can directly influence wound healing processes.
A differential pattern of gene expression for procollagen 1α1 and 3 was demonstrated in mid- and late-gestational fetal fibroblasts (MSC-like cells) in response to TGF-β1. Mid-gestational fetal cells showed decreased expression of procollagen 1α1, while late-gestational cells showed increased procollagen 1α1 and decreased procollagen 3 expression, demonstrating an increased type 3 to 1 collagen ratio in mid-gestational age and upregulation of type I collagen and decreased type III collagen synthesis with advancing gestational age [81,82]. Cell surface discoidin domain receptors-1 and 2 (DDR1 and DDR2) directly bind collagen and may also regulate collagen deposition by transmitting the signals intracellularly. The elucidation of the role of DDRs in scarless fetal wound repair revealed that DDR1 expression levels were downregulated in fetal fibroblasts with increasing gestational age and were inversely correlated with collagen production, in contrast to DDR2, which showed a similar level of expression throughout gestation [81].
Integrins mediate adhesive interactions with specific ECM proteins and can trigger signaling pathways that regulate cellular adhesion, motility, contractile capacity, proliferation, and differentiation in response to the rigidity of the substrate [83]. Variations in α-smooth actin and integrin expression patterns were demonstrated between fetal and adult fibroblasts after treatment with different isoforms of TGF-β. The expression of α-smooth actin and α3 and β1 integrin subunits was increased in adult fibroblasts. In contrast, fetal fibroblast cells showed a decrease in α1, α2, and β1 integrin expression but no change in α3 integrin and α-smooth actin expression. Fetal fibroblasts also showed an inhibition of their contractile capacity in comparison with adult fibroblasts [84].
On stimulation with pro-inflammatory cytokines, fetal and adult fibroblasts differentially express the hyaluronan synthase (HAS1-3) responsible for hyaluronic acid production. Exposure to IL-1 and tumor necrosis factor-alpha (TNF-α) induced a marked increase in HAS-1 and HAS-3 transcript levels in adult fibroblasts compared to the relatively muted response of fetal fibroblasts [85]. WNT3a and TGF-β1 treatment resulted in the induction of HAS2 and HAS3 gene expression in fetal fibroblasts and HAS1 and hyaluronidase-2 in postnatal fibroblasts [86]. Fetal fibroblasts were also found to have an approximately four-fold greater density of hyaluronic acid receptors (CD44) than those of adults [87].
Some functional differences have also been observed between adult and fetal fibroblasts in the pro-inflammatory microenvironment. After wounding, fetal fibroblasts appear to migrate in wounds and proliferate there much faster than adult fibroblasts [88,89]. A remarkable feature of fetal wounds is the essential absence of myofibroblasts, which contrasts with their emergence in the later fetal and postnatal periods [90,91]. The smooth muscle alpha-actin (α-SMA) levels have been found to be significantly elevated in adult fibroblasts in comparison to fetal fibroblasts. Thus, fetal fibroblasts do not possess myofibroblasts phenotype [92]. Fetal fibroblasts also exhibit a deficient contractile response to the rigid extracellular matrix and transforming growth factor-β1 in comparison with their adult dermal counterparts [84,93].
More recently, functionally diverse lineages of fibroblasts were discovered to coexist in the mouse back skin and oral cavity. Embryonic cells that have expressed engrailed 1 (En1) are responsible for the bulk of connective tissue deposition during embryonic development and wound healing and contribute to scarring in various models of wounds. Conversely, En1-lineage-naive fibroblasts were shown to drive dermal development and regeneration, do not participate in scar production, and their numbers decline with age [94,95]. Transplantation of En1-negative fibroblasts into back-skin wounds of adult mice resulted in a more reticular lattice arrangement of ECM and reduced scarring [95].

5. Isogenic Cellular Models of Different Stages of Human Development

The generation of isogenic cellular models corresponding to different stages of human development will empower opportunities for investigating phenotypic and functional features of adult and fetal mesodermal cells to elucidate the mechanisms responsible for scarless healing. Due to the inaccessibility of human tissues from different stages of embryonic development, the only way to investigate the mechanisms at these stages and recapitulate the ontogenetic processes in vitro is through the use of hPSCs. The promising technology of hPSCs has expanded our strategies in both basic research and clinical applications, enabling the modeling of human development and genetic diseases and the generation of differentiated cells for transplantation into patients. The pluripotent state of embryonic stem cells (Thomson JA et al., 1998) and somatic cells reprogrammed with transcription factors Oct3/4, Sox2, and Klf4, as well as c-Myc (Takahashi K et al., 2007) provides a feasible approach to obtaining cells corresponding to various stages of human development through directed differentiation in vitro [96,97]. Real-time tracking of hPSC differentiation provides a level of experimental availability of different cell models that is unattainable in vivo.
Currently, multiple strategies have been developed for spontaneous and directed differentiation of hPSCs into specialized cells from all three germ layers [98,99,100]. In the case of mesoderm, recent advances have demonstrated the opportunity to derive various types of mesenchymal cell populations from hPSCs. These include MSCs, hematopoietic cells, tenocytes, smooth muscle cells, osteoblasts, and cardiomyocytes [101,102,103,104,105,106,107,108]. Except for hematopoietic cells, MSCs have provided a differentiation potential with the capacity to generate tissues including bone, cartilage, tendon, muscle, adipose tissue, and bone marrow [109]. Although adult MSCs isolated from various tissues possess similar phenotypes and similar proliferative and differentiation potentials, they originate from developmentally diverse cell populations of lineage-specific mesodermal progenitors, which originate from the neural crest, paraxial mesoderm, and lateral plate mesoderm [101,110,111].
Several studies have reported that MSC-like cells can be obtained directly from hPSCs using serum-containing media without any signal control of intermediate stages [112,113,114,115,116,117]. Advanced methods for generating MSC-like cells and mesodermal progenitors follow a linear approach in which hPSCs are differentiated in discrete steps that mimic the sequence of events that occur during development. The identification of both inductive and repressive signal cues that define the sequential steps, through which hPSCs elaborate diversity of mesodermal progeny, facilitates the generation of mesodermal precursors for MSCs. These can be derived through several intermediate stages including primitive streak cell types [118,119], a neural crest lineage [102,120], neuromesodermal progenitors [121], lateral plate mesoderm cells [119,122], paraxial mesoderm, and somites [105].
Mesoderm development in vivo occurs during early gastrulation when the pluripotent cells of the epiblast begin to differentiate into a primitive streak, which then divides into paraxial and lateral mesoderm. Numerous experimental studies in vertebrate embryo models have yielded important insights into the molecules required for triggering mesoderm formation, maintaining mesoderm state, and mesoderm patterning [123,124,125]. The efforts to differentiate hPSCs into various mesoderm cell types in vitro have demonstrated that signaling molecules WNT, fibroblast growth factor (FGF), bone morphogenetic protein (BMP), and ACTIVIN/NODAL are involved in the induction of mesoderm differentiation and alternative mesodermal path navigation of hPSCs [101,126,127,128,129,130]. Primitive streak formation from hPSCs was found to be initiated following the activation of TGFβ, WNT, and FGF signaling. Co-expression of both BRACHYURY and MIXL1 was observed in these primitive streak-like cells [118,128]. Bifurcation of the primitive streak into the lateral and paraxial mesoderm can be induced by countervailing BMP and WNT signals. WNT activation and blocking BMP signaling were shown to abrogate lateral mesoderm and expand paraxial mesoderm. In contrast, WNT inhibition and exogenous BMP induced the lateral mesoderm. Lateral mesoderm cells were shown to express HAND1 and FOXF1. Paraxial mesoderm cells were defined by the expression of DLL3 and MSGN1 [105,122,128,131].
The paraxial mesoderm in embryos is segmented into somitomeres. The inhibition of FGF/ERK and WNT drive paraxial mesoderm cells derived from hPSCs toward early somite precursors expressing MEOX1 and FOXC2. Additionally, a homeodomain-containing transcriptional cofactor HOPX was identified as a somite segmentation marker, which was specifically expressed in a subset of somitomere cells but in neither paraxial mesoderm nor early somites [105,128,129].
In vivo, early somites are patterned to generate sclerotome and dermomyotome. Specification of hPSCs-derived somitomere cells in vitro was found to be regulated by the cross-antagonized effects of Hedgehog (Hh) and WNT. Hh activation together with WNT inhibition induced a sclerotome cell population that expressed markers PAX1, PAX9, NKX3.2/BAPX1, FOXC2, SOX9, and TWIST1. Conversely, WNT activation together with Hh blockade exclusively specified dermomyotome from somitomere cells [105,125,128].
Neuromesodermal progenitors in the posterior region of the embryo have the bipotential ability to differentiate into both ectodermal and mesodermal cell types and give rise to the paraxial mesoderm. Several studies have demonstrated that neuromesodermal progenitors can be generated from hPSCs by the activation of TGFβ, WNT, and FGF signal pathways. These cells transiently co-express mesodermal and neural crest markers Brachyury, Sox2, SOX9, SOX10, and Hox genes and can differentiate into neurons, melanocytes, MSCs, and osteogenic progenitors [120,121,132].
Moreover, in addition to the above mesodermal precursors existing transiently in these in vitro culture systems, cloned mesenchymal progenitors have been described. Using multifactorial high-throughput screening technology to engineer in vitro microenvironments that allow expansion of an hPSC-derived mesodermally restricted progenitor population, Kumar N. et al. described intermediate mesoderm cells expressing the pan-mesodermal markers MESP1, MIXL1, and LHX, which were capable of differentiating into cell types with renal gene expression patterns but failed to differentiate into lateral plate mesodermal lineages, such as blood and cardiac muscle [133].
A novel clonal progenitor for endotheliocytes and MSCs, designated as mesenchymoangioblast, was identified during mesendodermal differentiation of hPSCs in the semisolid medium in the presence of FGF2. The population of mesenchymoangioblast cells formed compact spheroid colonies and was positive for the expression of mesenchymal cell surface markers including platelet-derived growth factor receptor (PDGFR), CD146, CD90, and CD56 but did not express CD31, CD43, and CD73. Mesenchymal cells from mesenchymoangioblast colonies were able to differentiate into different types of mesenchymal lineages, including MSCs, pericytes, and smooth muscle cells but not hematopoietic cells and cardiomyocytes. Mesenchymoangioblast was shown to be a transient cell population arising from the apelin receptor (APLNR)+ mesodermal cells at the primitive streak stage. Identification of the lineage tree of mesenchymoangioblast-derived mesenchymal cells demonstrated that APLNR+ cells derived from hPSC express the FOXF1, IRX3, BMP4, WNT5A, HAND1, and HAND2 genes, representative of lateral plate/extraembryonic mesoderm but not the markers of paraxial/myogenic (MEOX1, TCF15, PAX3, PAX7) and intermediate (PAX2, PAX8) mesoderm in the embryo [134,135].
Another group utilized the hemangioblast generated from hPSCs as an intermediate cell type in the derivation of a highly potent and replenishable population of MSCs. The comparison of immunophenotype between hemangioblasts and the resulting MSCs highlighted the differences in the expression of surface markers. Most hemangioblast cells were positive in expression for hematopoietic marker CD45 and only part of them was positive for MSC markers CD90, CD105, or CD73, in contrast to MSCs, which were positive for CD90, CD105, or CD73 and negative for CD45 [136].
Following lineage-specific differentiation, hPSC-derived MSCs acquired some epigenetic, phenotypic, and functional features distinct from adult MSCs though they showed a typical MSC surface marker profile and differentiation potential. hPSC-derived MSCs showed higher CD10 and CD24 expression levels than those in adult MSCs [136]. A grained comparison of transcriptomes between hPSC-derived and adult MSCs revealed that developmental transcription factors HOXD1, NKX2-5, LHX2, and FGF12 as well as CD markers leukemia inhibitory factor receptor (LIFR), prostaglandin F2 receptor negative regulator (PTGFRN), and poliovirus receptor (PVR) were specifically upregulated in hPSC-derived MSCs [137]. hPSC-derived MSCs were also found to acquire a rejuvenation-associated gene signature, specifically, the expression of INHBE, DNMT3B, POU5F1P1, CDKN1C, and GCNT2, which are also expressed in hPSCs but not in the parental adult MSCs [138,139]. Epigenetic rejuvenation of MSCs derived from hPSC was confirmed using DNA methylation analysis. DNA methylation profiles of iPSCs and hPSC-derived MSCs were shown to maintain donor-specific characteristics, but tissue-specific, senescence-associated, and age-related DNA methylation patterns were erased during reprogramming. The DNA methylation pattern of hPSC-derived MSCs remained rejuvenated with regard to the DNA methylation pattern of parental MSCs during culture expansion [140]. Several studies indicate that MSCs derived from hPSCs exhibit stronger proliferation than adult MSCs [121,122,136,141]. Depth characterization of hPSC-derived MSCs, by comparing them to adult MSCs using transcriptomics (RNA-seq) and quantitative proteomics, highlighted biological processes relating to their source: cell cycle and nuclear division-related gene pools were enriched in ESC-MSC, consistent with their higher proliferation rate, whereas BM-MSCs showed enhanced extracellular organization related genes [136]. When induced to differentiate into osteoblasts, chondrocytes, and adipocytes in vitro, MSCs derived from hPSC through both lateral and paraxial mesoderm showed significantly improved osteogenic and chondrogenic potential but less adipogenic potential in comparison with adult MSCs [105,119,122]. The HGF expression level was found to be higher in MSCs derived through paraxial mesoderm progenitors compared to lateral mesoderm-derived MSCs, while VEGF-A and bFGF were more strongly expressed in MSCs derived through paraxial mesoderm progenitors compared to lateral mesoderm-derived MSCs [141].

6. Immunomodulatory Activity of Mesodermal Stromal Cells

The functional heterogeneity of MSCs can be determined not only by their ontogenetic origin but also by the inflammatory microenvironment. The immunomodulatory and fibrogenic activity of MSCs is mediated by the autocrine and paracrine effects of cytokines and growth factors as well as cell–cell and cell–matrix interactions through several positive and negative feedback loops occurring during the interaction of resident or migrated MSCs with immune cells in the site of acute and chronic inflammation [15,142,143,144].
Studies to date have shown that in vitro stimulation of human adult MSCs through TLR receptors induces the expression of genes involved in chemotaxis and inflammatory responses [145,146,147,148]. Other researchers demonstrated that TNF-α is also involved in the functional reprogramming of MSCs towards enhanced angiogenic and osteogenic activity in vitro and in vivo [149,150,151,152]. Alternatively, IFN-γ treatment was found to prime MSC immunosuppressive activity through the induction of the IDO enzyme, which catabolizes tryptophan into kynurenine and promotes the accumulation of tryptophan-derived catabolites contributing to the inhibition of immune cell proliferation [153,154,155,156]. We have shown that direct intercellular contact can also affect the functional status of MSCs [157].
However, the ability of hPSC-derived MSCs to respond to signals from the microenvironment and influence the function of immune cells has not been described in detail for adult MSCs. Nevertheless, several studies have confirmed the close overlapping of secreting immunomodulatory cytokines (IL-6, IL-8, MCP-1, CCL2) and anti-inflammatory mediators (cyclooxygenases-2 (COX-2), IDO, TGF-beta, programmed death-ligand 1 (PDL1)) between adult and hPSC-derived MSCs in a pro-inflammatory microenvironment [121,136,158]. Adult and hPSC-derived MSCs were also shown to possess a similar capacity to inhibit antigen-induced proliferation of peripheral blood mononuclear cells in a mixed culture system and promote the Treg response [121,136,159,160].
Several studies have reported that hPSC-derived MSCs possess improved therapeutic efficacy compared to tissue-specific MSCs in different animal models of diseases. hPSC-derived MSCs were demonstrated to outperform bone marrow MSCs in their ability to extravasate and migrate into inflamed tissues and inhibit T cell infiltration accompanied by an increasing Treg ratio in a murine encephalomyelitis model. Intraperitoneal administration of hPSC-derived MSCs resulted in the prevention of neural demyelination and a reduction in clinical symptoms [161]. hPSC-derived MSCs had a superior neuroprotective capacity over amniotic MSCs in a mouse model of brain ischemia. The increased anti-inflammatory potential of hPSC-derived MSCs was attributed to NF-κB-induced IL-13 production [162]. Compared to human adult bone marrow MSCs, transplantation of hPSC-derived MSCs into mice exerted a greater effect on vascular and muscle regeneration and achieved better attenuation of severe hind-limb ischemia [163]. MSCs derived from hPSC through neuromesodermal progenitors displayed much stronger immunomodulatory activity compared to bone marrow MSCs in vivo, as revealed by decreased inflammatory cell infiltration and diminished production of pro-inflammatory cytokines in inflamed tissue in a mouse model of contact hypersensitivity [121]. Moreover, the difference in therapeutic potential between MSCs derived from hPSCs through paraxial and lateral mesoderm progenitors was discovered in mouse models of skin wounds and pressure ulcers. In a mouse model of skin wounds, MSCs derived from hPSCs through lateral mesoderm accelerated wound healing faster than MSCs of paraxial origin. In contrast, treatment with MSCs originated from paraxial mesoderm progenitors more effectively cured pressure ulcers than treatment with lateral-mesoderm-derived MSCs [141].
The known mechanisms underlying the immunomodulatory activity of MSCs cannot perfectly describe the complex and multifactorial processes that support successful wound healing. Fibrotic disorders in various tissues are known to be associated with chronic activation of MSCs by pro-inflammatory mediators, which induce their transdifferentiation into myofibroblasts with scar-producing, proliferative, migratory, contractile, immunomodulatory, and phagocytic properties [143]. However, the antifibrotic activity of MSCs is poorly understood. Recently, we reported that equilibrity between inflammatory factors determines human MSC-mediated immunosuppressive effects, and MSC stimulation through different receptor-dependent pathways can induce some synergistic and overlapping functionalities with integrative effects [24]. The important signaling pathways such as Wnt, BMP, TGF-β, FGF, Notch, and Hh, which play critical roles in tissue patterning during embryonic development and maintaining tissue homeostasis in adults, may also be involved in wound healing and fibrosis [164]. The activation and crosstalk of these signaling pathways in MSCs can generate a gene regulatory network that determines cell fate specification, polarization, proliferation, differentiation, migration, and apoptosis. Thus, these signaling pathways may also represent potential therapeutic targets for attenuating MSC fibrotic activity. The generation of isogenic cellular models corresponding to different stages of human development discloses new pathways to investigate phenotypic and functional features of adult and fetal mesodermal cells in pro-inflammatory microenvironments to elucidate the mechanisms responsible for scarless healing.

7. Conclusions

Successful or impaired wound healing, as well as the degree of scarring, largely depends on the phenotypic and functional state of MSCs recruited into the wound, which is regulated by pro-inflammatory signals from the external microenvironment. The current knowledge of the antifibrogenic activity of MSCs in fetal wounds does not allow us to identify the mechanisms responsible for scarless healing. We believe that a comparative analysis of functional activity and gene/protein expression in isogenic mesodermal cells of various stages of human development differentiated from iPSCs under receptor-dependent activation of pro-inflammatory signaling pathways will reveal key molecules and therapeutic targets that will minimize scar formation. By elucidating the molecular mechanisms underlying the functional activity of adult and fetal mesodermal cells in pro-inflammatory microenvironments, we can achieve important advances in our understanding of the processes of wound healing and scarring. Continued work in this field will provide new insights into the mechanisms of wound healing and enable the development of novel therapeutics based on precise inhibition or stimulation of elements of the wound healing pathway.

Author Contributions

Conceptualization, Y.S.; writing—original draft preparation, Y.S.; writing—review and editing, S.L.K.; supervision, S.L.K.; project administration, S.L.K.; and funding acquisition, Y.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Russian Science Foundation, grant 23-24-00026.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created for this review.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Shaw, T.J.; Martin, P. Wound repair at a glance. J. Cell Sci. 2009, 122 Pt 18, 3209–3213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Marshall, C.D.; Hu, M.S.; Leavitt, T.; Barnes, L.A.; Lorenz, H.P.; Longaker, M.T. Cutaneous Scarring: Basic Science, Current Treatments, and Future Directions. Adv. Wound Care 2018, 7, 29–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef] [PubMed]
  4. Hu, M.S.; Maan, Z.N.; Wu, J.C.; Rennert, R.C.; Hong, W.X.; Lai, T.S.; Cheung, A.T.; Walmsley, G.G.; Chung, M.T.; McArdle, A.; et al. Tissue engineering and regenerative repair in wound healing. Ann. Biomed. Eng. 2014, 42, 1494–1507. [Google Scholar] [CrossRef]
  5. Kim, E.Y.; Hussain, A.; Khachemoune, A. Evidence-based management of keloids and hypertrophic scars in dermatology. Arch. Dermatol. Res. 2022; online ahead of print. [Google Scholar] [CrossRef]
  6. Colwell, A.S.; Longaker, M.T.; Lorenz, H.P. Fetal wound healing. Front. Biosci. 2003, 8, s1240–s1248. [Google Scholar] [CrossRef] [Green Version]
  7. Moore, A.L.; Marshall, C.D.; Barnes, L.A.; Murphy, M.P.; Ransom, R.C.; Longaker, M.T. Scarless wound healing: Transitioning from fetal research to regenerative healing. Wiley Interdiscip. Rev. Dev. Biol. 2018, 7, e309. [Google Scholar] [CrossRef]
  8. Burrington, J.D. Wound healing in the fetal lamb. J. Pediatr. Surg. 1971, 6, 523–528. [Google Scholar] [CrossRef] [PubMed]
  9. Somasundaram, K.; Prathap, K. Intra-uterine healing of skin wounds in rabbit foetuses. J. Pathol. 1970, 100, 81–86. [Google Scholar] [CrossRef]
  10. Goss, A.N. Intra-uterine healing of fetal rat oral mucosal, skin and cartilage wounds. J. Oral. Pathol. 1977, 6, 35–43. [Google Scholar] [CrossRef]
  11. Gnyawali, S.C.; Sinha, M.; El Masry, M.S.; Wulff, B.; Ghatak, S.; Soto-Gonzalez, F.; Wilgus, T.A.; Roy, S.; Sen, C.K. High resolution ultrasound imaging for repeated measure of wound tissue morphometry, biomechanics and hemodynamics under fetal, adult and diabetic conditions. PLoS ONE 2020, 15, e0241831. [Google Scholar] [CrossRef] [PubMed]
  12. Lorenz, H.P.; Longaker, M.T.; Perkocha, L.A.; Jennings, R.W.; Harrison, M.R.; Adzick, N.S. Scarless wound repair: A human fetal skin model. Development 1992, 114, 253–259. [Google Scholar] [CrossRef]
  13. Beanes, S.R.; Hu, F.Y.; Soo, C.; Dang, C.M.; Urata, M.; Ting, K.; Atkinson, J.B.; Benhaim, P.; Hedrick, M.H.; Lorenz, H.P. Confocal microscopic analysis of scarless repair in the fetal rat: Defining the transition. Plast. Reconstr. Surg. 2002, 109, 160–170. [Google Scholar] [CrossRef] [PubMed]
  14. Moretti, L.; Stalfort, J.; Barker, T.H.; Abebayehu, D. The interplay of fibroblasts, the extracellular matrix, and inflammation in scar formation. J. Biol. Chem. 2022, 298, 101530. [Google Scholar] [CrossRef] [PubMed]
  15. Pittenger, M.F.; Discher, D.E.; Péault, B.M.; Phinney, D.G.; Hare, J.M.; Caplan, A.I. Mesenchymal stem cell perspective: Cell biology to clinical progress. NPJ Regen. Med. 2019, 4, 22. [Google Scholar] [CrossRef] [Green Version]
  16. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop Dj 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]
  17. Waterman, R.S.; Tomchuck, S.L.; Henkle, S.L.; Betancourt, A.M. A new mesenchymal stem cell (MSC) paradigm: Polarization into a pro-inflammatory MSC1 or an Immunosuppressive MSC2 phenotype. PLoS ONE 2010, 5, e10088. [Google Scholar] [CrossRef] [Green Version]
  18. Le Blanc, K.; Davies, L.C. Mesenchymal stromal cells and the innate immune response. Immunol. Lett. 2015, 168, 140–146. [Google Scholar] [CrossRef] [Green Version]
  19. Weiss, A.R.R.; Dahlke, M.H. Immunomodulation by Mesenchymal Stem Cells (MSCs): Mechanisms of Action of Living, Apoptotic, and Dead MSCs. Front. Immunol. 2019, 10, 1191. [Google Scholar] [CrossRef] [Green Version]
  20. Yoo, K.H.; Jang, I.K.; Lee, M.W.; Kim, H.E.; Yang, M.S.; Eom, Y.; Lee, J.E.; Kim, Y.J.; Yang, S.K.; Jung, H.L.; et al. Comparison of immunomodulatory properties of mesenchymal stem cells derived from adult human tissues. Cell Immunol. 2009, 259, 150–156. [Google Scholar] [CrossRef]
  21. Takizawa, N.; Okubo, N.; Kamo, M.; Chosa, N.; Mikami, T.; Suzuki, K.; Yokota, S.; Ibi, M.; Ohtsuka, M.; Taira, M.; et al. Bone marrow-derived mesenchymal stem cells propagate immunosuppressive/anti-inflammatory macrophages in cell-to-cell contact-independent and -dependent manners under hypoxic culture. Exp. Cell Res. 2017, 358, 411–420. [Google Scholar] [CrossRef] [PubMed]
  22. Melief, S.M.; Schrama, E.; Brugman, M.H.; Tiemessen, M.M.; Hoogduijn, M.J.; Fibbe, W.E.; Roelofs, H. Multipotent stromal cells induce human regulatory T cells through a novel pathway involving skewing of monocytes toward anti-inflammatory macrophages. Stem Cells 2013, 31, 1980–1991. [Google Scholar] [CrossRef] [PubMed]
  23. Khosravi, M.; Bidmeshkipour, A.; Moravej, A.; Hojjat-Assari, S.; Naserian, S.; Karimi, M.H. Induction of CD4(+)CD25(+)Foxp3(+) regulatory T cells by mesenchymal stem cells is associated with RUNX complex factors. Immunol. Res. 2018, 66, 207–218. [Google Scholar] [CrossRef] [PubMed]
  24. Suzdaltseva, Y.; Goryunov, K.; Silina, E.; Manturova, N.; Stupin, V.; Kiselev, S.L. Equilibrium among Inflammatory Factors Determines Human MSC-Mediated Immunosuppressive Effect. Cells 2022, 11, 1210. [Google Scholar] [CrossRef] [PubMed]
  25. Guillén, M.I.; Platas, J.; Pérez Del Caz, M.D.; Mirabet, V.; Alcaraz, M.J. Paracrine Anti-inflammatory Effects of Adipose Tissue-Derived Mesenchymal Stem Cells in Human Monocytes. Front. Physiol. 2018, 9, 661. [Google Scholar] [CrossRef]
  26. Mareschi, K.; Castiglia, S.; Sanavio, F.; Rustichelli, D.; Muraro, M.; Defedele, D.; Bergallo, M.; Fagioli, F. Immunoregulatory effects on T lymphocytes by human mesenchymal stromal cells isolated from bone marrow, amniotic fluid, and placenta. Exp. Hematol. 2016, 44, 138–150.e1. [Google Scholar] [CrossRef] [Green Version]
  27. Kwon, H.M.; Hur, S.M.; Park, K.Y.; Kim, C.K.; Kim, Y.M.; Kim, H.S.; Shin, H.C.; Won, M.H.; Ha, K.S.; Kwon, Y.G.; et al. Multiple paracrine factors secreted by mesenchymal stem cells contribute to angiogenesis. Vasc. Pharmacol. 2014, 63, 19–28. [Google Scholar] [CrossRef]
  28. Ratushnyy, A.; Ezdakova, M.; Buravkova, L. Secretome of Senescent Adipose-Derived Mesenchymal Stem Cells Negatively Regulates Angiogenesis. Int. J. Mol. Sci. 2020, 21, 1802. [Google Scholar] [CrossRef] [Green Version]
  29. Du, W.J.; Chi, Y.; Yang, Z.X.; Li, Z.J.; Cui, J.J.; Song, B.Q.; Li, X.; Yang, S.G.; Han, Z.B.; Han, Z.C. Heterogeneity of proangiogenic features in mesenchymal stem cells derived from bone marrow, adipose tissue, umbilical cord, and placenta. Stem Cell Res. Ther. 2016, 7, 163. [Google Scholar] [CrossRef] [Green Version]
  30. Rubina, K.; Kalinina, N.; Efimenko, A.; Lopatina, T.; Melikhova, V.; Tsokolaeva, Z.; Sysoeva, V.; Tkachuk, V.; Parfyonova, Y. Adipose stromal cells stimulate angiogenesis via promoting progenitor cell differentiation, secretion of angiogenic factors, and enhancing vessel maturation. Tissue Eng. Part A 2009, 15, 2039–2050. [Google Scholar] [CrossRef] [Green Version]
  31. Freiman, A.; Shandalov, Y.; Rozenfeld, D.; Shor, E.; Segal, S.; Ben-David, D.; Meretzki, S.; Egozi, D.; Levenberg, S. Adipose-derived endothelial and mesenchymal stem cells enhance vascular network formation on three-dimensional constructs in vitro. Stem Cell Res. Ther. 2016, 7, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Yu, Y.; Situ, Q.; Jia, W.; Li, J.; Wu, Q.; Lei, J. Data driven mathematical modeling reveals the dynamic mechanism of MSC-induced neovascularization. FASEB J. 2019, 33, 3496–3509. [Google Scholar] [CrossRef] [PubMed]
  33. Novoseletskaya, E.; Grigorieva, O.; Nimiritsky, P.; Basalova, N.; Eremichev, R.; Milovskaya, I.; Kulebyakin, K.; Kulebyakina, M.; Rodionov, S.; Omelyanenko, N.; et al. Mesenchymal Stromal Cell-Produced Components of Extracellular Matrix Potentiate Multipotent Stem Cell Response to Differentiation Stimuli. Front. Cell Dev. Biol. 2020, 8, 555378. [Google Scholar] [CrossRef]
  34. Hyldig, K.; Riis, S.; Pennisi, C.P.; Zachar, V.; Fink, T. Implications of Extracellular Matrix Production by Adipose Tissue-Derived Stem Cells for Development of Wound Healing Therapies. Int. J. Mol. Sci. 2017, 18, 1167. [Google Scholar] [CrossRef] [Green Version]
  35. Baberg, F.; Geyh, S.; Waldera-Lupa, D.; Stefanski, A.; Zilkens, C.; Haas, R.; Schroeder, T.; Stühler, K. Secretome analysis of human bone marrow derived mesenchymal stromal cells. Biochim. Biophys. Acta Proteins Proteom. 2019, 1867, 434–441. [Google Scholar] [CrossRef]
  36. Lozito, T.P.; Jackson, W.M.; Nesti, L.J.; Tuan, R.S. Human mesenchymal stem cells generate a distinct pericellular zone of MMP activities via binding of MMPs and secretion of high levels of TIMPs. Matrix Biol. 2014, 7, 129. [Google Scholar] [CrossRef] [PubMed]
  37. Almalki, S.G.; Agrawal, D.K. Effects of matrix metalloproteinases on the fate of mesenchymal stem cells. Stem Cell Res. Ther. 2016, 7, 129. [Google Scholar] [CrossRef] [Green Version]
  38. Maffioli, E.; Nonnis, S.; Angioni, R.; Santagata, F.; Calì, B.; Zanotti, L.; Negri, A.; Viola, A.; Tedeschi, G. Proteomic analysis of the secretome of human bone marrow-derived mesenchymal stem cells primed by pro-inflammatory cytokines. J. Proteom. 2017, 166, 115–126. [Google Scholar] [CrossRef]
  39. Chen, X.X.; Tang, L.; Han, Z.H.; Wang, W.J.; Meng, J.G. Coculture with bone marrow-derived mesenchymal stem cells attenuates inflammation and apoptosis in lipopolysaccharide-stimulated alveolar epithelial cells via enhanced secretion of keratinocyte growth factor and angiopoietin-1 modulating the Toll-like receptor-4 signal pathway. Mol. Med. Rep. 2019, 19, 1891–1902. [Google Scholar] [CrossRef] [Green Version]
  40. Tang, W.; Zhang, Y.; Tang, L.; Zhang, J.; Xiong, L.; Wang, B. Inhibitory effect of tranilast on the myofibroblast differentiation of rat mesenchymal stem cells induced by transforming growth factor-beta1 in vitro. Mol. Med. Rep. 2018, 18, 5693–5700. [Google Scholar] [CrossRef] [Green Version]
  41. Lecarpentier, Y.; Schussler, O.; Sakic, A.; Rincon-Garriz, J.M.; Soulie, P.; Bochaton-Piallat, M.L.; Kindler, V. Human Bone Marrow Contains Mesenchymal Stromal Stem Cells That Differentiate In Vitro into Contractile Myofibroblasts Controlling T Lymphocyte Proliferation. Stem Cells Int. 2018, 2018, 6134787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Kramann, R.; Schneider, R.K.; DiRocco, D.P.; Machado, F.; Fleig, S.; Bondzie, P.A.; Henderson, J.M.; Ebert, B.L.; Humphreys, B.D. Perivascular Gli1+ progenitors are key contributors to injury-induced organ fibrosis. Cell Stem Cell 2015, 16, 51–66. [Google Scholar] [CrossRef] [Green Version]
  43. Schneider, R.K.; Mullally, A.; Dugourd, A.; Peisker, F.; Hoogenboezem, R.; Van Strien, P.M.H.; Bindels, E.M.; Heckl, D.; Büsche, G.; Fleck, D.; et al. Gli1(+) Mesenchymal Stromal Cells Are a Key Driver of Bone Marrow Fibrosis and an Important Cellular Therapeutic Target. Cell Stem Cell 2017, 20, 785–800.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Rodriguez-Menocal, L.; Shareef, S.; Salgado, M.; Shabbir, A.; Van Badiavas, E. Role of whole bone marrow, whole bone marrow cultured cells, and mesenchymal stem cells in chronic wound healing. Stem Cell Res. Ther. 2015, 6, 24. [Google Scholar] [CrossRef] [Green Version]
  45. Kato, Y.; Iwata, T.; Washio, K.; Yoshida, T.; Kuroda, H.; Morikawa, S.; Hamada, M.; Ikura, K.; Kaibuchi, N.; Yamato, M.; et al. Creation and Transplantation of an Adipose-derived Stem Cell (ASC) Sheet in a Diabetic Wound-healing Model. J. Vis. Exp. 2017, 126, 54539. [Google Scholar] [CrossRef]
  46. Silina, E.; Stupin, V.; Koreyba, K.; Bolevich, S.; Suzdaltseva, Y.; Manturova, N. Local and Remote Effects of Mesenchymal Stem Cell Administration on Skin Wound Regeneration. Pathophysiology 2021, 28, 355–372. [Google Scholar] [CrossRef]
  47. Silina, E.V.; Stupin, V.A.; Suzdaltseva, Y.G.; Aliev, S.R.; Abramov, I.S.; Khokhlov, N.V. Application of Polymer Drugs with Cerium Dioxide Nanomolecules and Mesenchymal Stem Cells for the Treatment of Skin Wounds in Aged Rats. Polymers 2021, 13, 1467. [Google Scholar] [CrossRef]
  48. Pelizzo, G.; Avanzini, M.A.; Icaro Cornaglia, A.; Osti, M.; Romano, P.; Avolio, L.; Maccario, R.; Dominici, M.; De Silvestri, A.; Andreatta, E.; et al. Mesenchymal stromal cells for cutaneous wound healing in a rabbit model: Pre-clinical study apipplicable in the pediatric surgical setting. J. Transl. Med. 2015, 13, 219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Martinello, T.; Gomiero, C.; Perazzi, A.; Iacopetti, I.; Gemignani, F.; DeBenedictis, G.M.; Ferro, S.; Zuin, M.; Martines, E.; Brun, P.; et al. Allogeneic mesenchymal stem cells improve the wound healing process of sheep skin. BMC Vet. Res. 2018, 14, 202. [Google Scholar] [CrossRef] [Green Version]
  50. Liu, L.; Farhoodi, H.P.; Han, M.; Liu, G.; Yu, J.; Nguyen, L.; Nguyen, B.; Nguyen, A.; Liao, W.; Zhao, W. Preclinical Evaluation of a Single Intravenous Infusion of hUC-MSC (BX-U001) in Rheumatoid Arthritis. Cell Transplant. 2020, 29, 963689720965896. [Google Scholar] [CrossRef]
  51. Zhang, Q.; Li, Q.; Zhu, J.; Guo, H.; Zhai, Q.; Li, B.; Jin, Y.; He, X.; Jin, F. Comparison of therapeutic effects of different mesenchymal stem cells on rheumatoid arthritis in mice. PeerJ 2019, 7, e7023. [Google Scholar] [CrossRef] [Green Version]
  52. López, Y.; Lutjemeier, B.; Seshareddy, K.; Trevino, E.M.; Hageman, K.S.; Musch, T.I.; Borgarelli, M.; Weiss, M.L. Wharton’s jelly or bone marrow mesenchymal stromal cells improve cardiac function following myocardial infarction for more than 32 weeks in a rat model: A preliminary report. Curr. Stem Cell Res. Ther. 2013, 8, 46–59. [Google Scholar] [CrossRef] [Green Version]
  53. Wang, S.; Huang, S.; Gong, L.; Yuan, Z.; Wong, J.; Lee, J.; Si, M.S. Human Neonatal Thymus Mesenchymal Stem Cells Promote Neovascularization and Cardiac Regeneration. Stem Cells Int. 2018, 2018, 8503468. [Google Scholar] [CrossRef] [PubMed]
  54. Li, H.; Rong, P.; Ma, X.; Nie, W.; Chen, Y.; Zhang, J.; Dong, Q.; Yang, M.; Wang, W. Mouse Umbilical Cord Mesenchymal Stem Cell Paracrine Alleviates Renal Fibrosis in Diabetic Nephropathy by Reducing Myofibroblast Transdifferentiation and Cell Proliferation and Upregulating MMPs in Mesangial Cells. J. Diabetes Res. 2020, 2020, 3847171. [Google Scholar] [CrossRef] [PubMed]
  55. Shrestha, C.; Zhao, L.; Chen, K.; He, H.; Mo, Z. Enhanced healing of diabetic wounds by subcutaneous administration of human umbilical cord derived stem cells and their conditioned media. Int. J. Endocrinol. 2013, 2013, 592454. [Google Scholar] [CrossRef] [Green Version]
  56. Suzdaltseva, Y.; Zhidkih, S.; Kiselev, S.L.; Stupin, V. Locally Delivered Umbilical Cord Mesenchymal Stromal Cells Reduce Chronic Inflammation in Long-Term Nonhealing Wounds: A Randomized Study. Stem Cells Int. 2020, 2020, 5308609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Rodríguez-Fuentes, D.E.; Fernández-Garza, L.E.; Samia-Meza, J.A.; Barrera-Barrera, S.A.; Caplan, A.I.; Barrera-Saldaña, H.A. Mesenchymal Stem Cells Current Clinical Applications: A Systematic Review. Arch. Med. Res. 2021, 52, 93–101. [Google Scholar] [CrossRef]
  58. Adzick, N.S.; Longaker, M.T. Animal models for the study of fetal tissue repair. J. Surg. Res. 1991, 51, 216–222. [Google Scholar] [CrossRef]
  59. Naik-Mathuria, B.; Gay, A.N.; Zhu, X.; Yu, L.; Cass, D.L.; Olutoye, O.O. Age-dependent recruitment of neutrophils by fetal endothelial cells: Implications in scarless wound healing. J. Pediatr. Surg. 2007, 42, 166–171. [Google Scholar] [CrossRef]
  60. Olutoye, O.O.; Zhu, X.; Cass, D.L.; Smith, C.W. Neutrophil recruitment by fetal porcine endothelial cells: Implications in scarless fetal wound healing. Pediatr. Res. 2005, 58, 1290–1294. [Google Scholar] [CrossRef] [Green Version]
  61. Wulff, B.C.; Parent, A.E.; Meleski, M.A.; DiPietro, L.A.; Schrementi, M.E.; Wilgus, T.A. Mast cells contribute to scar formation during fetal wound healing. J. Investig. Dermatol. 2012, 132, 458–465. [Google Scholar] [CrossRef] [Green Version]
  62. Cowin, A.J.; Brosnan, M.P.; Holmes, T.M.; Ferguson, M.W. Endogenous inflammatory response to dermal wound healing in the fetal and adult mouse. Dev. Dyn. 1998, 212, 385–393. [Google Scholar] [CrossRef]
  63. Morris, M.W., Jr.; Allukian, M., 3rd; Herdrich, B.J.; Caskey, R.C.; Zgheib, C.; Xu, J.; Dorsett-Martin, W.; Mitchell, M.E.; Liechty, K.W. Modulation of the inflammatory response by increasing fetal wound size or interleukin-10 overexpression determines wound phenotype and scar formation. Wound Repair. Regen. 2014, 22, 406–414. [Google Scholar] [CrossRef]
  64. Liechty, K.W.; Adzick, N.S.; Crombleholme, T.M. Diminished interleukin 6 (IL-6) production during scarless human fetal wound repair. Cytokine 2000, 12, 671–676. [Google Scholar] [CrossRef]
  65. Kohama, K.; Nonaka, K.; Hosokawa, R.; Shum, L.; Ohishi, M. TGF-beta-3 promotes scarless repair of cleft lip in mouse fetuses. J. Dent. Res. 2002, 81, 688–694. [Google Scholar] [CrossRef]
  66. Cowin, A.J.; Holmes, T.M.; Brosnan, P.; Ferguson, M.W. Expression of TGF-beta and its receptors in murine fetal and adult dermal wounds. Eur. J. Dermatol. 2001, 11, 424–431. [Google Scholar]
  67. Wilgus, T.A.; Ferreira, A.M.; Oberyszyn, T.M.; Bergdall, V.K.; Dipietro, L.A. Regulation of scar formation by vascular endothelial growth factor. Lab. Investig. 2008, 88, 579–590. [Google Scholar] [CrossRef] [Green Version]
  68. Lovvorn, H.N., 3rd; Cheung, D.T.; Nimni, M.E.; Perelman, N.; Estes, J.M.; Adzick, N.S. Relative distribution and crosslinking of collagen distinguish fetal from adult sheep wound repair. J. Pediatr. Surg. 1999, 34, 218–223. [Google Scholar] [CrossRef]
  69. Longaker, M.T.; Whitby, D.J.; Adzick, N.S.; Crombleholme, T.M.; Langer, J.C.; Duncan, B.W.; Bradley, S.M.; Stern, R.; Ferguson, M.W.; Harrison, M.R. Studies in fetal wound healing, VI. Second and early third trimester fetal wounds demonstrate rapid collagen deposition without scar formation. J. Pediatr. Surg. 1990, 25, 63–68. [Google Scholar] [CrossRef] [PubMed]
  70. Merkel, J.R.; DiPaolo, B.R.; Hallock, G.G.; Rice, D.C. Type I and type III collagen content of healing wounds in fetal and adult rats. Proc. Soc. Exp. Biol. Med. 1988, 187, 493–497. [Google Scholar] [CrossRef] [PubMed]
  71. Whitby, D.J.; Ferguson, M.W. The extracellular matrix of lip wounds in fetal, neonatal and adult mice. Development 1991, 112, 651–668. [Google Scholar] [CrossRef]
  72. Soo, C.; Hu, F.Y.; Zhang, X.; Wang, Y.; Beanes, S.R.; Lorenz, H.P.; Hedrick, M.H.; Mackool, R.J.; Plaas, A.; Kim, S.J.; et al. Differential expression of fibromodulin, a transforming growth factor-beta modulator, in fetal skin development and scarless repair. Am. J. Pathol. 2000, 157, 423–433. [Google Scholar] [CrossRef] [PubMed]
  73. Zheng, Z.; Zhang, X.; Dang, C.; Beanes, S.; Chang, G.X.; Chen, Y.; Li, C.S.; Lee, K.S.; Ting, K.; Soo, C. Fibromodulin Is Essential for Fetal-Type Scarless Cutaneous Wound Healing. Am. J. Pathol. 2016, 186, 2824–2832. [Google Scholar] [CrossRef] [Green Version]
  74. Beanes, S.R.; Dang, C.; Soo, C.; Wang, Y.; Urata, M.; Ting, K.; Fonkalsrud, E.W.; Benhaim, P.; Hedrick, M.H.; Atkinson, J.B.; et al. Down-regulation of decorin, a transforming growth factor-beta modulator, is associated with scarless fetal wound healing. J. Pediatr. Surg. 2001, 36, 1666–1671. [Google Scholar] [CrossRef]
  75. Longaker, M.T.; Chiu, E.S.; Adzick, N.S.; Stern, M.; Harrison, M.R.; Stern, R. Studies in fetal wound healing. V. A prolonged presence of hyaluronic acid characterizes fetal wound fluid. Ann. Surg. 1991, 213, 292–296. [Google Scholar] [CrossRef]
  76. West, D.C.; Shaw, D.M.; Lorenz, P.; Adzick, N.S.; Longaker, M.T. Fibrotic healing of adult and late gestation fetal wounds correlates with increased hyaluronidase activity and removal of hyaluronan. Int. J. Biochem. Cell Biol. 1997, 29, 201–210. [Google Scholar] [CrossRef]
  77. Dang, C.M.; Beanes, S.R.; Lee, H.; Zhang, X.; Soo, C.; Ting, K. Scarless fetal wounds are associated with an increased matrix metalloproteinase-to-tissue-derived inhibitor of metalloproteinase ratio. Plast. Reconstr. Surg. 2003, 111, 2273–2285. [Google Scholar] [CrossRef] [PubMed]
  78. Pfisterer, K.; Shaw, L.E.; Symmank, D.; Weninger, W. The Extracellular Matrix in Skin Inflammation and Infection. Front. Cell Dev. Biol. 2021, 9, 682414. [Google Scholar] [CrossRef]
  79. Burk, J.; Sassmann, A.; Kasper, C.; Nimptsch, A.; Schubert, S. Extracellular Matrix Synthesis and Remodeling by Mesenchymal Stromal Cells Is Context-Sensitive. Int. J. Mol. Sci. 2022, 23, 1758. [Google Scholar] [CrossRef]
  80. Ragelle, H.; Naba, A.; Larson, B.L.; Zhou, F.; Prijić, M.; Whittaker, C.A.; Del Rosario, A.; Langer, R.; Hynes, R.O.; Anderson, D.G. Comprehensive proteomic characterization of stem cell-derived extracellular matrices. Biomaterials 2017, 128, 147–159. [Google Scholar] [CrossRef]
  81. Chin, G.S.; Lee, S.; Hsu, M.; Liu, W.; Kim, W.J.; Levinson, H.; Longaker, M.T. Discoidin domain receptors and their ligand, collagen, are temporally regulated in fetal rat fibroblasts in vitro. Plast. Reconstr. Surg. 2001, 107, 769–776. [Google Scholar] [CrossRef]
  82. Carter, R.; Jain, K.; Sykes, V.; Lanning, D. Differential expression of procollagen genes between mid- and late-gestational fetal fibroblasts. J. Surg. Res. 2009, 156, 90–94. [Google Scholar] [CrossRef] [PubMed]
  83. Kolasangiani, R.; Bidone, T.C.; Schwartz, M.A. Integrin Conformational Dynamics and Mechanotransduction. Cells 2022, 11, 3584. [Google Scholar] [CrossRef]
  84. Moulin, V.; Tam, B.Y.; Castilloux, G.; Auger, F.A.; O’Connor-McCourt, M.D.; Philip, A.; Germain, L. Fetal and adult human skin fibroblasts display intrinsic differences in contractile capacity. J. Cell Physiol. 2001, 188, 211–222. [Google Scholar] [CrossRef] [PubMed]
  85. Kennedy, C.I.; Diegelmann, R.F.; Haynes, J.H.; Yager, D.R. Proinflammatory cytokines differentially regulate hyaluronan synthase isoforms in fetal and adult fibroblasts. J. Pediatr. Surg. 2000, 35, 874–879. [Google Scholar] [CrossRef]
  86. Carre, A.L.; James, A.W.; MacLeod, L.; Kong, W.; Kawai, K.; Longaker, M.T.; Lorenz, H.P. Interaction of wingless protein (Wnt), transforming growth factor-beta1, and hyaluronan production in fetal and postnatal fibroblasts. Plast. Reconstr. Surg. 2010, 125, 74–88. [Google Scholar] [CrossRef] [PubMed]
  87. Alaish, S.M.; Yager, D.; Diegelmann, R.F.; Cohen, I.K. Biology of fetal wound healing: Hyaluronate receptor expression in fetal fibroblasts. J. Pediatr. Surg. 1994, 29, 1040–1043. [Google Scholar] [CrossRef] [PubMed]
  88. Nath, R.K.; Parks, W.C.; Mackinnon, S.E.; Hunter, D.A.; Markham, H.; Weeks, P.M. The regulation of collagen in fetal skin wounds: mRNA localization and analysis. J. Pediatr. Surg. 1994, 29, 855–862. [Google Scholar] [CrossRef]
  89. Brink, H.E.; Miller, G.J.; Beredjiklian, P.K.; Nicoll, S.B. Serum-dependent effects on adult and fetal tendon fibroblast migration and collagen expression. Wound Repair. Regen. 2006, 14, 179–186. [Google Scholar] [CrossRef]
  90. Estes, J.M.; Vande Berg, J.S.; Adzick, N.S.; MacGillivray, T.E.; Desmoulière, A.; Gabbiani, G. Phenotypic and functional features of myofibroblasts in sheep fetal wounds. Differentiation 1994, 56, 173–181. [Google Scholar] [CrossRef]
  91. Cass, D.L.; Sylvester, K.G.; Yang, E.Y.; Crombleholme, T.M.; Adzick, N.S. Myofibroblast persistence in fetal sheep wounds is associated with scar formation. J. Pediatr. Surg. 1997, 32, 1017–1021, discussion 1021-2. [Google Scholar] [CrossRef]
  92. Satish, L.; Johnson, S.; Wang, J.H.; Post, J.C.; Ehrlich, G.D.; Kathju, S. Chaperonin containing T-complex polypeptide subunit eta (CCT-eta) is a specific regulator of fibroblast motility and contractility. PLoS ONE 2010, 5, e10063. [Google Scholar] [CrossRef] [Green Version]
  93. Jerrell, R.J.; Leih, M.J.; Parekh, A. The altered mechanical phenotype of fetal fibroblasts hinders myofibroblast differentiation. Wound Repair. Regen. 2019, 27, 29–38. [Google Scholar] [CrossRef] [Green Version]
  94. Rinkevich, Y.; Walmsley, G.G.; Hu, M.S.; Maan, Z.N.; Newman, A.M.; Drukker, M.; Januszyk, M.; Krampitz, G.W.; Gurtner, G.C.; Lorenz, H.P.; et al. Skin fibrosis. Identification and isolation of a dermal lineage with intrinsic fibrogenic potential. Science 2015, 348, aaa2151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Jiang, D.; Correa-Gallegos, D.; Christ, S.; Stefanska, A.; Liu, J.; Ramesh, P.; Rajendran, V.; De Santis, M.M.; Wagner, D.E.; Rinkevich, Y. Two succeeding fibroblastic lineages drive dermal development and the transition from regeneration to scarring. Nat. Cell Biol. 2018, 20, 422–431. [Google Scholar] [CrossRef] [Green Version]
  96. Thomson, J.A.; Itskovitz-Eldor, J.; Shapiro, S.S.; Waknitz, M.A.; Swiergiel, J.J.; Marshall, V.S.; Jones, J.M. Embryonic stem cell lines derived from human blastocysts. Science 1998, 282, 1145–1147. [Google Scholar] [CrossRef] [Green Version]
  97. Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K.; Yamanaka, S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007, 131, 861–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Nekrasov, E.D.; Vigont, V.A.; Klyushnikov, S.A.; Lebedeva, O.S.; Vassina, E.M.; Bogomazova, A.N.; Chestkov, I.V.; Semashko, T.A.; Kiseleva, E.; Suldina, L.A.; et al. Manifestation of Huntington’s disease pathology in human induced pluripotent stem cell-derived neurons. Mol. Neurodegener. 2016, 11, 27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Philonenko, E.S.; Shutova, M.V.; Khomyakova, E.A.; Vassina, E.M.; Lebedeva, O.S.; Kiselev, S.L.; Lagarkova, M.A. Differentiation of Human Pluripotent Stem Cells into Mesodermal and Ectodermal Derivatives Is Independent of the Type of Isogenic Reprogrammed Somatic Cells. Acta Naturae 2017, 9, 68–74. [Google Scholar] [CrossRef]
  100. Panova, A.V.; Klementieva, N.V.; Sycheva, A.V.; Korobko, E.V.; Sosnovtseva, A.O.; Krasnova, T.S.; Karpova, M.R.; Rubtsov, P.M.; Tikhonovich, Y.V.; Tiulpakov, A.N.; et al. Aberrant Splicing of INS Impairs Beta-Cell Differentiation and Proliferation by ER Stress in the Isogenic iPSC Model of Neonatal Diabetes. Int. J. Mol. Sci. 2022, 23, 8824. [Google Scholar] [CrossRef]
  101. Cheung, C.; Bernardo, A.S.; Trotter, M.W.; Pedersen, R.A.; Sinha, S. Generation of human vascular smooth muscle subtypes provides insight into embryological origin-dependent disease susceptibility. Nat. Biotechnol. 2012, 30, 165–173. [Google Scholar] [CrossRef] [Green Version]
  102. Chijimatsu, R.; Ikeya, M.; Yasui, Y.; Ikeda, Y.; Ebina, K.; Moriguchi, Y.; Shimomura, K.; Hart, D.A.; Hideki, Y.; Norimasa, N. Characterization of Mesenchymal Stem Cell-Like Cells Derived from Human iPSCs via Neural Crest Development and Their Application for Osteochondral Repair. Stem Cells Int. 2017, 2017, 1960965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Kimura, M.; Furukawa, H.; Shoji, M.; Shinozawa, T. Increased mesodermal and mesendodermal populations by BMP4 treatment facilitates human iPSC line differentiation into a cardiac lineage. J. Stem Cells Regen. Med. 2019, 15, 45–51. [Google Scholar] [CrossRef]
  104. Wang, Y.; Wang, H.; Guo, J.; Gao, J.; Wang, M.; Xia, M.; Wen, Y.; Su, P.; Yang, M.; Liu, M.; et al. LGR4, Not LGR5, Enhances hPSC Hematopoiesis by Facilitating Mesoderm Induction via TGF-Beta Signaling Activation. Cell Rep. 2020, 31, 107600. [Google Scholar] [CrossRef] [PubMed]
  105. Nakajima, T.; Shibata, M.; Nishio, M.; Nagata, S.; Alev, C.; Sakurai, H.; Toguchida, J.; Ikeya, M. Modeling human somite development and fibrodysplasia ossificans progressiva with induced pluripotent stem cells. Development 2018, 145, dev165431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Nakajima, T.; Ikeya, M. Development of pluripotent stem cell-based human tenocytes. Dev. Growth Differ. 2021, 63, 38–46. [Google Scholar] [CrossRef]
  107. Philonenko, E.S.; Tan, Y.; Wang, C.; Zhang, B.; Shah, Z.; Zhang, J.; Ullah, H.; Kiselev, S.L.; Lagarkova, M.A.; Li, D.; et al. Recapitulative haematopoietic development of human pluripotent stem cells in the absence of exogenous haematopoietic cytokines. J. Cell Mol. Med. 2021, 25, 8701–8714. [Google Scholar] [CrossRef]
  108. Kamatani, T.; Hagizawa, H.; Yarimitsu, S.; Morioka, M.; Koyamatsu, S.; Sugimoto, M.; Kodama, J.; Yamane, J.; Ishiguro, H.; Shichino, S.; et al. Human iPS cell-derived cartilaginous tissue spatially and functionally replaces nucleus pulposus. Biomaterials 2022, 284, 121491. [Google Scholar] [CrossRef]
  109. Singer, N.G.; Caplan, A.I. Mesenchymal stem cells: Mechanisms of inflammation. Annu. Rev. Pathol. 2011, 6, 457–478. [Google Scholar] [CrossRef] [Green Version]
  110. Isern, J.; García-García, A.; Martín, A.M.; Arranz, L.; Martín-Pérez, D.; Torroja, C.; Sánchez-Cabo, F.; Méndez-Ferrer, S. The neural crest is a source of mesenchymal stem cells with specialized hematopoietic stem cell niche function. Elife 2014, 3, e03696. [Google Scholar] [CrossRef]
  111. Sheng, G. The developmental basis of mesenchymal stem/stromal cells (MSCs). BMC Dev. Biol. 2015, 15, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Hwang, N.S.; Varghese, S.; Lee, H.J.; Zhang, Z.; Ye, Z.; Bae, J.; Cheng, L.; Elisseeff, J. In vivo commitment and functional tissue regeneration using human embryonic stem cell-derived mesenchymal cells. Proc. Natl. Acad. Sci. USA 2008, 105, 20641–20646. [Google Scholar] [CrossRef] [PubMed]
  113. Karlsson, C.; Emanuelsson, K.; Wessberg, F.; Kajic, K.; Axell, M.Z.; Eriksson, P.S.; Lindahl, A.; Hyllner, J.; Strehl, R. Human embryonic stem cell-derived mesenchymal progenitors--potential in regenerative medicine. Stem Cell Res. 2009, 3, 39–50. [Google Scholar] [CrossRef] [Green Version]
  114. Marolt, D.; Campos, I.M.; Bhumiratana, S.; Koren, A.; Petridis, P.; Zhang, G.; Spitalnik, P.F.; Grayson, W.L.; Vunjak-Novakovic, G. Engineering bone tissue from human embryonic stem cells. Proc. Natl. Acad. Sci. USA 2012, 109, 8705–8709. [Google Scholar] [CrossRef]
  115. Liu, Y.; Goldberg, A.J.; Dennis, J.E.; Gronowicz, G.A.; Kuhn, L.T. One-step derivation of mesenchymal stem cell (MSC)-like cells from human pluripotent stem cells on a fibrillar collagen coating. PLoS ONE 2012, 7, e33225. [Google Scholar] [CrossRef] [Green Version]
  116. Villa-Diaz, L.G.; Brown, S.E.; Liu, Y.; Ross, A.M.; Lahann, J.; Parent, J.M.; Krebsbach, P.H. Derivation of mesenchymal stem cells from human induced pluripotent stem cells cultured on synthetic substrates. Stem Cells 2012, 30, 1174–1181. [Google Scholar] [CrossRef] [Green Version]
  117. Diederichs, S.; Tuan, R.S. Functional comparison of human-induced pluripotent stem cell-derived mesenchymal cells and bone marrow-derived mesenchymal stromal cells from the same donor. Stem Cells Dev. 2014, 23, 1594–1610. [Google Scholar] [CrossRef]
  118. Tran, N.T.; Trinh, Q.M.; Lee, G.M.; Han, Y.M. Efficient differentiation of human pluripotent stem cells into mesenchymal stem cells by modulating intracellular signaling pathways in a feeder/serum-free system. Stem Cells Dev. 2012, 21, 1165–1175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Liu, T.M.; Yildirim, E.D.; Li, P.; Fang, H.T.; Denslin, V.; Kumar, V.; Loh, Y.H.; Lee, E.H.; Cool, S.M.; Teh, B.T.; et al. Ascorbate and Iron Are Required for the Specification and Long-Term Self-Renewal of Human Skeletal Mesenchymal Stromal Cells. Stem Cell Rep. 2020, 14, 210–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Fukuta, M.; Nakai, Y.; Kirino, K.; Nakagawa, M.; Sekiguchi, K.; Nagata, S.; Matsumoto, Y.; Yamamoto, T.; Umeda, K.; Heike, T.; et al. Derivation of mesenchymal stromal cells from pluripotent stem cells through a neural crest lineage using small molecule compounds with defined media. PLoS ONE 2014, 9, e112291. [Google Scholar] [CrossRef] [Green Version]
  121. Wang, H.; Li, D.; Zhai, Z.; Zhang, X.; Huang, W.; Chen, X.; Huang, L.; Liu, H.; Sun, J.; Zou, Z.; et al. Characterization and Therapeutic Application of Mesenchymal Stem Cells with Neuromesodermal Origin from Human Pluripotent Stem Cells. Theranostics 2019, 9, 1683–1697. [Google Scholar] [CrossRef] [PubMed]
  122. Wei, Y.; Wang, B.; Jia, L.; Huang, W.; Xiang, A.P.; Fang, C.; Liang, X.; Li, W. Lateral Mesoderm-Derived Mesenchymal Stem Cells With Robust Osteochondrogenic Potential and Hematopoiesis-Supporting Ability. Front. Mol. Biosci. 2022, 9, 767536. [Google Scholar] [CrossRef] [PubMed]
  123. Kimelman, D. Mesoderm induction: From caps to chips. Nat. Rev. Genet. 2006, 7, 360–372. [Google Scholar] [CrossRef]
  124. Tam, P.P.; Loebel, D.A. Gene function in mouse embryogenesis: Get set for gastrulation. Nat. Rev. Genet. 2007, 8, 368–381. [Google Scholar] [CrossRef]
  125. Tani, S.; Chung, U.I.; Ohba, S.; Hojo, H. Understanding paraxial mesoderm development and sclerotome specification for skeletal repair. Exp. Mol. Med. 2020, 52, 1166–1177. [Google Scholar] [CrossRef] [PubMed]
  126. Gertow, K.; Hirst, C.E.; Yu, Q.C.; Ng, E.S.; Pereira, L.A.; Davis, R.P.; Stanley, E.G.; Elefanty, A.G. WNT3A promotes hematopoietic or mesenchymal differentiation from hESCs depending on the time of exposure. Stem Cell Rep. 2013, 1, 53–65. [Google Scholar] [CrossRef] [Green Version]
  127. Umeda, K.; Zhao, J.; Simmons, P.; Stanley, E.; Elefanty, A.; Nakayama, N. Human chondrogenic paraxial mesoderm, directed specification and prospective isolation from pluripotent stem cells. Sci. Rep. 2012, 2, 455. [Google Scholar] [CrossRef] [Green Version]
  128. Loh, K.M.; Chen, A.; Koh, P.W.; Deng, T.Z.; Sinha, R.; Tsai, J.M.; Barkal, A.A.; Shen, K.Y.; Jain, R.; Morganti, R.M.; et al. Mapping the Pairwise Choices Leading from Pluripotency to Human Bone, Heart, and Other Mesoderm Cell Types. Cell 2016, 166, 451–467. [Google Scholar] [CrossRef] [Green Version]
  129. Xi, H.; Fujiwara, W.; Gonzalez, K.; Jan, M.; Liebscher, S.; Van Handel, B.; Schenke-Layland, K.; Pyle, A.D. In Vivo Human Somitogenesis Guides Somite Development from hPSCs. Cell Rep. 2017, 18, 1573–1585. [Google Scholar] [CrossRef] [Green Version]
  130. Kishimoto, K.; Iwasawa, K.; Sorel, A.; Ferran-Heredia, C.; Han, L.; Morimoto, M.; Wells, J.M.; Takebe, T.; Zorn, A.M. Directed differentiation of human pluripotent stem cells into diverse organ-specific mesenchyme of the digestive and respiratory systems. Nat. Protoc. 2022, 17, 2699–2719. [Google Scholar] [CrossRef]
  131. Smith, C.A.; Humphreys, P.A.; Naven, M.A.; Woods, S.; Mancini, F.E.; O’Flaherty, J.; Meng, Q.J.; Kimber, S.J. Directed differentiation of hPSCs through a simplified lateral plate mesoderm protocol for generation of articular cartilage progenitors. PLoS ONE 2023, 18, e0280024. [Google Scholar] [CrossRef] [PubMed]
  132. Kidwai, F.; Mui, B.W.H.; Arora, D.; Iqbal, K.; Hockaday, M.; de Castro Diaz, L.F.; Cherman, N.; Martin, D.; Myneni, V.D.; Ahmad, M.; et al. Lineage-specific differentiation of osteogenic progenitors from pluripotent stem cells reveals the FGF1-RUNX2 association in neural crest-derived osteoprogenitors. Stem Cells 2020, 38, 1107–1123. [Google Scholar] [CrossRef] [PubMed]
  133. Kumar, N.; Richter, J.; Cutts, J.; Bush, K.T.; Trujillo, C.; Nigam, S.K.; Gaasterland, T.; Brafman, D.; Willert, K. Generation of an expandable intermediate mesoderm restricted progenitor cell line from human pluripotent stem cells. Elife 2015, 4, e08413. [Google Scholar] [CrossRef]
  134. Vodyanik, M.A.; Yu, J.; Zhang, X.; Tian, S.; Stewart, R.; Thomson, J.A.; Slukvin, I.I. A mesoderm-derived precursor for mesenchymal stem and endothelial cells. Cell Stem Cell 2010, 7, 718–729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Slukvin, I.I.; Kumar, A. The mesenchymoangioblast, mesodermal precursor for mesenchymal and endothelial cells. Cell Mol. Life Sci. 2018, 75, 3507–3520. [Google Scholar] [CrossRef]
  136. Kimbrel, E.A.; Kouris, N.A.; Yavanian, G.J.; Chu, J.; Qin, Y.; Chan, A.; Singh, R.P.; McCurdy, D.; Gordon, L.; Levinson, R.D.; et al. Mesenchymal stem cell population derived from human pluripotent stem cells displays potent immunomodulatory and therapeutic properties. Stem Cells Dev. 2014, 23, 1611–1624. [Google Scholar] [CrossRef]
  137. Billing, A.M.; Ben Hamidane, H.; Dib, S.S.; Cotton, R.J.; Bhagwat, A.M.; Kumar, P.; Hayat, S.; Yousri, N.A.; Goswami, N.; Suhre, K.; et al. Comprehensive transcriptomic and proteomic characterization of human mesenchymal stem cells reveals source specific cellular markers. Sci. Rep. 2016, 6, 21507. [Google Scholar] [CrossRef] [Green Version]
  138. Spitzhorn, L.S.; Megges, M.; Wruck, W.; Rahman, M.S.; Otte, J.; Degistirici, Ö.; Meisel, R.; Sorg, R.V.; Oreffo, R.O.C.; Adjaye, J. Human iPSC-derived MSCs (iMSCs) from aged individuals acquire a rejuvenation signature. Stem Cell Res. Ther. 2019, 10, 100. [Google Scholar] [CrossRef] [Green Version]
  139. Wruck, W.; Graffmann, N.; Spitzhorn, L.S.; Adjaye, J. Human Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells Acquire Rejuvenation and Reduced Heterogeneity. Front. Cell Dev. Biol. 2021, 9, 717772. [Google Scholar] [CrossRef]
  140. Frobel, J.; Hemeda, H.; Lenz, M.; Abagnale, G.; Joussen, S.; Denecke, B.; Sarić, T.; Zenke, M.; Wagner, W. Epigenetic rejuvenation of mesenchymal stromal cells derived from induced pluripotent stem cells. Stem Cell Rep. 2014, 3, 414–422. [Google Scholar] [CrossRef] [Green Version]
  141. Eto, S.; Goto, M.; Soga, M.; Kaneko, Y.; Uehara, Y.; Mizuta, H.; Era, T. Mesenchymal stem cells derived from human iPS cells via mesoderm and neuroepithelium have different features and therapeutic potentials. PLoS ONE 2018, 13, e0200790. [Google Scholar] [CrossRef] [Green Version]
  142. Prockop, D.J. Concise review: Two negative feedback loops place mesenchymal stem/stromal cells at the center of early regulators of inflammation. Stem Cells 2013, 31, 2042–2046. [Google Scholar] [CrossRef]
  143. Pellicoro, A.; Ramachandran, P.; Iredale, J.P.; Fallowfield, J.A. Liver fibrosis and repair: Immune regulation of wound healing in a solid organ. Nat. Rev. Immunol. 2014, 14, 181–194. [Google Scholar] [CrossRef] [PubMed]
  144. Desai, O.; Winkler, J.; Minasyan, M.; Herzog, E.L. The Role of Immune and Inflammatory Cells in Idiopathic Pulmonary Fibrosis. Front. Med. 2018, 5, 43. [Google Scholar] [CrossRef] [PubMed]
  145. Pevsner-Fischer, M.; Morad, V.; Cohen-Sfady, M.; Rousso-Noori, L.; Zanin-Zhorov, A.; Cohen, S.; Cohen, I.R.; Zipori, D. Toll-like receptors and their ligands control mesenchymal stem cell functions. Blood 2007, 109, 1422–1432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Tomchuck, S.L.; Zwezdaryk, K.J.; Coffelt, S.B.; Waterman, R.S.; Danka, E.S.; Scandurro, A.B. Toll-like receptors on human mesenchymal stem cells drive their migration and immunomodulating responses. Stem Cells 2008, 26, 99–107. [Google Scholar] [CrossRef] [Green Version]
  147. Kim, S.H.; Das, A.; Chai, J.C.; Binas, B.; Choi, M.R.; Park, K.S.; Lee, Y.S.; Jung, K.H.; Chai, Y.G. Transcriptome sequencing wide functional analysis of human mesenchymal stem cells in response to TLR4 ligand. Sci. Rep. 2016, 6, 30311. [Google Scholar] [CrossRef] [Green Version]
  148. Jafari, M.; Asghari, A.; Delbandi, A.A.; Jalessi, M.; Jazayeri, M.H.; Samarei, R.; Tajik, N. Priming TLR3 and TLR4 in human adipose- and olfactory mucosa-derived mesenchymal stromal cells and comparison of their cytokine secretions. Cytotechnology 2020, 72, 57–68. [Google Scholar] [CrossRef]
  149. Kwon, Y.W.; Heo, S.C.; Jeong, G.O.; Yoon, J.W.; Mo, W.M.; Lee, M.J.; Jang, I.H.; Kwon, S.M.; Lee, J.S.; Kim, J.H. Tumor necrosis factor-α-activated mesenchymal stem cells promote endothelial progenitor cell homing and angiogenesis. Biochim. Biophys. Acta 2013, 1832, 2136–2144. [Google Scholar] [CrossRef] [Green Version]
  150. Lu, Z.; Wang, G.; Dunstan, C.R.; Chen, Y.; Lu, W.Y.; Davies, B.; Zreiqat, H. Activation and promotion of adipose stem cells by tumour necrosis factor-alpha preconditioning for bone regeneration. J. Cell Physiol. 2013, 228, 1737–1744. [Google Scholar] [CrossRef]
  151. Zubkova, E.S.; Beloglazova, I.B.; Makarevich, P.I.; Boldyreva, M.A.; Sukhareva, O.Y.; Shestakova, M.V.; Dergilev, K.V.; Parfyonova, Y.V.; Menshikov, M.Y. Regulation of Adipose Tissue Stem Cells Angiogenic Potential by Tumor Necrosis Factor-Alpha. J. Cell Biochem. 2016, 117, 180–196. [Google Scholar] [CrossRef]
  152. Li, W.; Liu, Q.; Shi, J.; Xu, X.; Xu, J. The role of TNF-α in the fate regulation and functional reprogramming of mesenchymal stem cells in an inflammatory microenvironment. Front. Immunol. 2023, 14, 1074863. [Google Scholar] [CrossRef] [PubMed]
  153. DelaRosa, O.; Lombardo, E.; Beraza, A.; Mancheño-Corvo, P.; Ramirez, C.; Menta, R.; Rico, L.; Camarillo, E.; García, L.; Abad, J.L.; et al. Requirement of IFN-gamma-mediated indoleamine 2,3-dioxygenase expression in the modulation of lymphocyte proliferation by human adipose-derived stem cells. Tissue Eng. Part A 2009, 15, 2795–2806. [Google Scholar] [CrossRef] [PubMed]
  154. Torres Crigna, A.; Uhlig, S.; Elvers-Hornung, S.; Klüter, H.; Bieback, K. Human Adipose Tissue-Derived Stromal Cells Suppress Human, but Not Murine Lymphocyte Proliferation, via Indoleamine 2,3-Dioxygenase Activity. Cells 2020, 9, 2419. [Google Scholar] [CrossRef] [PubMed]
  155. Burand, A.J., Jr.; Di, L.; Boland, L.K.; Boyt, D.T.; Schrodt, M.V.; Santillan, D.A.; Ankrum, J.A. Aggregation of Human Mesenchymal Stromal Cells Eliminates Their Ability to Suppress Human T Cells. Front. Immunol. 2020, 11, 143. [Google Scholar] [CrossRef]
  156. Herzig, M.C.; Delavan, C.P.; Jensen, K.J.; Cantu, C.; Montgomery, R.K.; Christy, B.A.; Cap, A.P.; Bynum, J.A. A streamlined proliferation assay using mixed lymphocytes for evaluation of human mesenchymal stem cell immunomodulation activity. J. Immunol. Methods 2021, 488, 112915. [Google Scholar] [CrossRef]
  157. Suzdaltseva, Y.G.; Goryunov, K.V.; Rubtsov, Y.P. The Role of Intercellular Contacts in Induction of Indolamine-2,3-Dioxygenase Synthesis in MMSC from Adipose Tissue. Cell Tissue Biol. 2018, 12, 391–401. [Google Scholar] [CrossRef]
  158. Wang, X.; Lazorchak, A.S.; Song, L.; Li, E.; Zhang, Z.; Jiang, B.; Xu, R.H. Immune modulatory mesenchymal stem cells derived from human embryonic stem cells through a trophoblast-like stage. Stem Cells 2016, 34, 380–391. [Google Scholar] [CrossRef] [Green Version]
  159. Fu, Q.L.; Chow, Y.Y.; Sun, S.J.; Zeng, Q.X.; Li, H.B.; Shi, J.B.; Sun, Y.Q.; Wen, W.; Tse, H.F.; Lian, Q.; et al. Mesenchymal stem cells derived from human induced pluripotent stem cells modulate T-cell phenotypes in allergic rhinitis. Allergy 2012, 67, 1215–1222. [Google Scholar] [CrossRef]
  160. Ozay, E.I.; Vijayaraghavan, J.; Gonzalez-Perez, G.; Shanthalingam, S.; Sherman, H.L.; Garrigan, D.T., Jr.; Chandiran, K.; Torres, J.A.; Osborne, B.A.; Tew, G.N.; et al. Cymerus™ iPSC-MSCs significantly prolong survival in a pre-clinical, humanized mouse model of Graft-vs-host disease. Stem Cell Res. 2019, 35, 101401. [Google Scholar] [CrossRef]
  161. Wang, X.; Kimbrel, E.A.; Ijichi, K.; Paul, D.; Lazorchak, A.S.; Chu, J.; Kouris, N.A.; Yavanian, G.J.; Lu, S.J.; Pachter, J.S.; et al. Human ESC-derived MSCs outperform bone marrow MSCs in the treatment of an EAE model of multiple sclerosis. Stem Cell Rep. 2014, 3, 115–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Hawkins, K.E.; Corcelli, M.; Dowding, K.; Ranzoni, A.M.; Vlahova, F.; Hau, K.L.; Hunjan, A.; Peebles, D.; Gressens, P.; Hagberg, H.; et al. Embryonic Stem Cell-Derived Mesenchymal Stem Cells (MSCs) Have a Superior Neuroprotective Capacity Over Fetal MSCs in the Hypoxic-Ischemic Mouse Brain. Stem Cells Transl. Med. 2018, 7, 439–449. [Google Scholar] [CrossRef] [PubMed]
  163. Lian, Q.; Zhang, Y.; Zhang, J.; Zhang, H.K.; Wu, X.; Zhang, Y.; Lam, F.F.; Kang, S.; Xia, J.C.; Lai, W.H.; et al. Functional mesenchymal stem cells derived from human induced pluripotent stem cells attenuate limb ischemia in mice. Circulation 2010, 121, 1113–1123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Hu, H.H.; Cao, G.; Wu, X.Q.; Vaziri, N.D.; Zhao, Y.Y. Wnt signaling pathway in aging-related tissue fibrosis and therapies. Ageing Res. Rev. 2020, 60, 101063. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Suzdaltseva, Y.; Kiselev, S.L. Mesodermal Derivatives of Pluripotent Stem Cells Route to Scarless Healing. Int. J. Mol. Sci. 2023, 24, 11945. https://doi.org/10.3390/ijms241511945

AMA Style

Suzdaltseva Y, Kiselev SL. Mesodermal Derivatives of Pluripotent Stem Cells Route to Scarless Healing. International Journal of Molecular Sciences. 2023; 24(15):11945. https://doi.org/10.3390/ijms241511945

Chicago/Turabian Style

Suzdaltseva, Yulia, and Sergey L. Kiselev. 2023. "Mesodermal Derivatives of Pluripotent Stem Cells Route to Scarless Healing" International Journal of Molecular Sciences 24, no. 15: 11945. https://doi.org/10.3390/ijms241511945

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