Next Article in Journal
Isolated Gastric Metastases of Pancreatic Ductal Adenocarcinoma following Radical Resection—Impact of Endosonography-Guided Fine Needle Aspiration Tract Seeding
Next Article in Special Issue
The Impact of Inflammatory Stimuli on Xylosyltransferase-I Regulation in Primary Human Dermal Fibroblasts
Previous Article in Journal
Modulatory Effects of Estradiol and Its Mixtures with Ligands of GPER and PPAR on MAPK and PI3K/Akt Signaling Pathways and Tumorigenic Factors in Mouse Testis Explants and Mouse Tumor Leydig Cells
Previous Article in Special Issue
Targeting of Janus Kinases Limits Pro-Inflammatory but Also Immunosuppressive Circuits in the Crosstalk between Synovial Fibroblasts and Lymphocytes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role and Function of Mesenchymal Stem Cells on Fibroblast in Cutaneous Wound Healing

1
Division of Plastic and Reconstructive Surgery, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan
2
Department of Life Science, Faculty of Medicine, Shimane University, 89-1 Enya-cho, Izumo 693-8501, Japan
3
Fukuoka Tokushukai Hospital, 4-5 Sugukita, Kasuga 816-0864, Japan
4
Department of Frontier Science for Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(6), 1391; https://doi.org/10.3390/biomedicines10061391
Submission received: 7 May 2022 / Revised: 6 June 2022 / Accepted: 10 June 2022 / Published: 12 June 2022
(This article belongs to the Special Issue Fibroblasts: Key Mediators of Regeneration, Inflammation and Fibrosis)

Abstract

:
Skin wounds often repair themselves completely over time; however, this is true only for healthy individuals. Although various studies are being conducted to improve wound-healing therapy outcomes, the mechanisms of wound healing and regeneration are not completely understood yet. In recent years, mesenchymal stem cells (MSCs) have been reported to contribute significantly to wound healing and regeneration. Understanding the function of MSCs will help to elucidate the fundamentals of wound healing. MSCs are multipotent stem cells that are used in regenerative medicine for their ability to self-renew and differentiate into bone, fat, and cartilage, with few ethical problems associated with cell harvesting. Additionally, they have anti-inflammatory and immunomodulatory properties and antifibrotic effects via paracrine signaling, and many studies have been conducted to use them to treat graft-versus-host disease, inflammatory bowel disease, and intractable cutaneous wounds. Many substances derived from MSCs are involved in the wound-healing process, and specific cascades and pathways have been elucidated. This review aims to explain the fundamental role of MSCs in wound healing and the effects of MSCs on fibroblasts.

1. Introduction

Mesenchymal stem cells (MSCs) are self-renewing multipotent tissue stem cells. MSCs renew themselves and differentiate into bone [1,2], cartilage [3,4], and fat [5,6], which in turn transform into cells that constitute each of these tissues. MSCs can be harvested from bone marrow [7,8], adipose tissue [9,10], placenta [11,12], and dental pulp [13,14]. On the other hand, embryonic stem (ES) cells are pluripotent stem cells derived from the inner cell mass of a blastocyst, an early-stage pre-implantation embryo. ES cells for clinical use have always raised ethical issues. However, researchers and physicians can avoid ethical problems in MSCs transplantation because MSCs can be harvested and cultured from adult tissues. They are one of the stem cells that are expected to be applied in regenerative medicine. One of the classes of cells that MSCs can differentiate into is fibroblasts [15]. Fibroblasts participate in the structure of all tissues in the body (skin, muscle, nerve, etc.) and are the primary cells of the connective tissue. Furthermore, fibroblasts not only constitute tissues but are also involved in the proliferation of cancer cells [16,17]. Thus, fibroblasts are essential for the maintenance of human life and are also involved in the malignant transformation of cells. Fibroblasts are also important cells for both clinical and academic purposes. In skin wound healing, fibroblasts play a major role in damage repair, and successful control of fibroblast collagen production not only promotes wound healing but also prevents scar formation [18,19,20]. The promotion of skin wound healing has been extensively studied as a form of regenerative medicine using MSCs [21,22]. Reportedly, MSCs migrate and proliferate in the damaged area to fill in the defect and properly manage fibroblast proliferation and collagen production. This review summarizes the basic description of MSCs as a regenerative medicine, the interaction between MSCs and fibroblasts, how the two cells interact in wound healing, and relevant problems.

2. Cutaneous Wound-Healing Process

The skin wound-healing process consists of three phases: inflammation, proliferation, and remodeling [23]. When external stimuli are applied to the skin causing damage, the skin tissue breaks down, and its destruction occurs. Simultaneously, blood vessels in the path of the injury are severed, causing blood to leak [24]. Platelets in the blood adhere to damaged and exposed collagen fibers and get activated [25]. Activated platelets subsequently activate coagulation factors, which convert prothrombin to thrombin [26,27], which in turn converts fibrinogen to fibrin [28]. Fibrin engulfs platelets and red blood cells to form clots. This series of events is the hemostatic effect of the blood coagulation reaction. In addition to clotting factors, various chemicals are released into the blood that leaks out due to vascular damage [29]. Additionally, cell membranes of the destroyed cells undergo chain reactions in the arachidonic acid cascade [30], releasing a variety of chemicals. These released chemicals create gaps in the endothelial cells of the capillaries and dilate them, allowing spherical polymorphonuclear leukocytes and mononuclear cells to escape from the vessels as effusions and migrate toward the wound [31]. At this time, proinflammatory cytokines are released by the phagocytosis of mononuclear cells. This induces four clinical symptoms: redness, swelling, pain, and fever. In the second proliferative phase, the released inflammatory cytokines cause fibroblasts to migrate to the wound and produce collagen fibers [32]. They also act on the endothelial cells of the capillaries to promote angiogenesis [33]. The newly created blood vessels supply the wound with blood, which in turn supplies fibroblasts with the oxygen and nutrients they need for proliferation. Through this sequence of events, the skin defect is filled, and the wound heals. The tissue that fills these defects is called the granulation tissue, and by combining with substances other than collagen, it transforms into a solid tissue such as the dermis. At this stage, it is called scar tissue [18,34,35]. In the final stage of skin wound healing, fibroblast function and collagen production decrease, but the amount of collagen produced is equal to the amount degraded and absorbed. Although the tissue formed in this manner appears to be unchanged, it is an active tissue that is constantly being produced and broken down. The reconstructed subepidermal tissue does not replace the dermis but remains a permanent scar tissue [19]. The epidermis looks almost the same as before the injury, but the scar tissue looks somewhat different from the dermis because of the irregular arrangement of collagen. This is called a scar. Once scar tissue matures, the surrounding tissue cannot regenerate normal dermal tissue [21,22,36]. Therefore, one of the challenges of wound healing is not to eliminate scar tissue but to reduce the amount of scar tissue and make it resemble the normal dermis. In recent years, MSCs have been widely applied to promote wound healing [37]. The treatment and prevention of aberrant scarring using MSCs have been studied extensively in basic studies but are not yet in clinical applications.

3. Mesenchymal Stem Cells (MSCs)

MSCs were first isolated from bone marrow in 1960–1970 and were characterized as cells with fibroblastoid shape and osteogenic potential, which form clonogenic colonies [22]. Depending on their tissue of origin, MSCs exhibit different characteristics. They are stem cells that can self-renew and proliferate [38] and differentiate into fat [5,6], bone [1,2], and/or cartilage [3,4] in the human body. They are present in all tissues and can be harvested from the placenta [11,12], bone marrow [7,8], fat tissues [9,10], and dental pulp [13,14]. MSCs reside in a non-hematopoietic population and can be cultured to form fibroblast-like colonies in vitro [39,40]. They are expected to make a major contribution to regenerative medicine because they can be isolated using a relatively simple technique [41,42] and because there are very few ethical issues, they can be easily applied to clinical trials [43]. There have been over 1000 clinical trials exists with MSCs over 25 years [44], and their numbers are growing day by day. MSCs are being studied for the treatment of fractures [45,46], arthritis [47], osteonecrosis of the jaw [48,49,50], graft-versus-host disease [51], Crohn’s disease [52], multiple sclerosis [53], stroke [54], myocardial infarction [55], liver cirrhosis [56], and chronic lung disease [57]. Furthermore, they have immunosuppressive functions, and many studies are underway to develop therapies that take advantage of this function. For that reason, MSCs have been reported to be particularly effective in the treatment of inflammatory bowel disease [58] and autoimmune disorders [59]. However, there are some issues regarding the methods of isolation and collection of MSCs. The MSCs that are currently available are obtained by seeding bone marrow mononuclear cells on culture dishes and collecting attached cells after 2–3 weeks of culture [7]. Regarding differentiation potential, the presence of hematopoietic cells and foreign cells with low or completely lost differentiation potential is inevitable, and undifferentiated cells with actual differentiation and proliferation potential (high-quality MSCs) are present only at a low frequency [60]. Regarding proliferative ability, MSCs, which are primary cultured cells, have a limited number of proliferation cycles and usually lose proliferative potential after 3–5 passages. At this time, their differentiation potential was significantly reduced. In addition, MSCs cannot be used for the treatment of systemic diseases by intravenous administration because the cells transform during culture amplification. When MSCs pass through the blood vessels of the lungs, they should be trapped in the small capillaries of the lungs [61,62]. Therefore, for safe and effective cell therapy, it is necessary to guarantee cellular functions of the MSCs, such as proliferation and differentiation capacity, as well as cell uniformity and migration capacity. Many studies have been conducted to resolve these problems, to recognize proteins expressed on the cell surface of MSCs as antigenic markers, and to isolate high-quality MSCs by analyzing their expression patterns using several types of markers [63]. Therefore, the collection of high-purity, higher-quality MSCs and the management of cultured MSCs for their use in regenerative medicine is a problem that needs to be solved as soon as possible [64,65]. Although MSCs can be used for many diseases and have significant advantages, there still are issues in establishing collection methods in clinical settings.

3.1. Bone Marrow-Derived MSCs

Bone marrow has long been used to harvest MSCs in vivo. In general, when MSCs are used to promote wound healing in basic experiments and clinical applications, they are most likely bone marrow-derived. Therefore, the characteristics of bone marrow-derived MSCs should be known and described first. The International Society for Cellular Therapy proposed the following criteria for the identification of MSCs: adherence to plastic; differentiation into chondrocytes, osteoblasts, and adipocytes under standard in vitro differentiation conditions; and expression of surface markers CD105, CD73, and CD90, in the absence of CD45, CD34, CD14, CD11b, CD79α, CD19, and HLA-DR [66]. MSCs, particularly bone marrow-derived MSCs, are found in a variety of tissues in vivo and have been extensively studied [67]. MSCs in human bone marrow were initially thought to constitute the microenvironment of hematopoietic stem cells (HSCs) and play a role in maintaining tissue homeostasis [68]. The bone marrow niche is a vital microenvironment that regulates many stem cell activities, including self-renewal, mobilization, engraftment, and lineage differentiation [69]. Niches are the local tissue microenvironments that maintain and regulate stem cells. Hematopoiesis provides a paradigm for understanding mammalian stem cells and their niches; however, the HSC niche remains incompletely defined and beset by competing models [70]. Bone marrow-derived MSCs lose their proliferative, differentiation, and migratory abilities with age because the bone marrow changes from red marrow to fat marrow with age in humans. As MSCs age, their cell size increases, and they eventually undergo apoptosis and die [71]. These characteristics have been reported to have similar results in mouse MSCs as well as human MSCs [72]. MSC fractions were collected from bone marrow cells by measuring the expression pattern of proteins on the cell surface and the fluorescence intensity by flow cytometry. Various MSC fractions have been defined from the bone marrow, and the expression patterns of markers have been investigated, but the question remains as to which is the true MSC fraction. It is assumed that all fractions contain at least some true MSCs, but a large percentage of cells other than MSCs (erythrocytes, mononuclear cells, and fibroblasts) are thought to be present. For example, if the MSC fraction contains a high proportion of fibroblasts, cells taken and cultured from the bone marrow would be fibroblasts rather than MSCs and would not differentiate into bone or fat. Thus, MSCs and fibroblasts are largely related, and there is an urgent need to develop a method that can completely distinguish between MSC and fibroblasts. Sudo et al. considered various primary fibroblast-like cultures and showed that all cells studied expressed CD44, CD90, and CD105, whereas none of them expressed CD14, CD34, and CD45 [73]. Although fibroblasts are increasingly being reported to have an MSC-like cell surface immunophenotype (cell surface protein), MSCs have also been perceived to be positive for fibroblast markers such as collagen, vimentin, fibroblast surface protein, heat shock protein 47 (HSP47), and α smooth muscle actin (αSMA) [74,75]. Although several markers have been identified for the two cell types, one of the key problems associated with the proper characterization of both cell types is that they are generally nonspecific. The two types of cells might be categorized by a combination of nonspecific markers rather than a few specific markers. There are also many conflicting results regarding the characterization of cell surface markers on MSCs and differences between MSCs and fibroblasts. The variability in results may be due to a variety of factors, including differences in the tissue of origin, donor heterogeneity, and lack of adequate studies to specifically identify differences between these two related cell types [76]. Thus, MSCs harvested from bone marrow have great potential for clinical application owing to their high proliferative and differentiation potential, but problems arise when harvesting bone marrow from a living person because of the small quantity extracted and the labor-intensive nature of the process. In most cases, bone marrow is harvested from the iliac bone, from which MSCs are isolated and cultured. However, because only a small number of bone marrow cells can be obtained from a single bone marrow biopsy, the number of MSCs that retain multilineage differentiation potential is small. In addition, the technology and environment for culturing and managing MSCs in a completely clean environment are required to increase their numbers, which is time-consuming and costly. In addition, the risk of side effects from bone marrow biopsy must be considered.

3.2. Adipose Tissue-Derived MSCs

Understanding adipose-derived MSCs compared to bone marrow-derived MSCs also provides a deeper understanding of the characteristics of MSCs. In particular, plastic surgeons involved in wound healing will be able to obtain adipose-derived MSCs through liposuction procedures; therefore, research is expected to advance further in the future. As adipose tissue is one of the factors contributing to obesity, and many people may not have a suitable perception of fat. Liposuction is becoming increasingly common worldwide and a common procedure in the field of plastic surgery [77,78]. In addition, in regenerative medicine, subcutaneous adipose tissue is attracting attention as a new tissue stem cell source to replace bone marrow because it can be harvested in large volumes, has a high percentage of multipotent cells, can be easily cultured in large volumes, and has characteristics similar to those of bone marrow-derived MSCs [79]. Although these are few reports, adipose tissue-derived MSCs can differentiate into numerous cell forms such as osteocytes, adipocytes, neural precursor cells, vascular endothelial cells, myocytes, pancreatic β cells, and hepatocytes [80,81,82,83,84]. Human adipose tissue-derived MSCs express conventional MSC surface markers, including the cell adhesion molecules CD29, CD44, CD146, and CD166, the receptor molecules CD90 and CD105, and the GPI-anchored enzyme CD73. Furthermore, adipose tissue-derived MSCs should be negative for hematopoietic cell surface antigens, including CD11b, CD13, CD14, CD19, and CD45. They are also negative for endothelial markers CD31 and HLA-DR [85]. From a clinical standpoint, adipose tissue-derived MSCs and bone marrow-derived MSCs are considered to have similar capabilities with some molecular differences (immunophenotype, differentiation potential, transcriptome, proteome, and immunomodulatory activity). Furthermore, adipose tissue-derived MSCs may be more suitable for the treatment of several diseases than bone marrow-derived MSCs because of their less invasive and safe technique [86]. Bone marrow was the first tissue used in vivo for MSC collection; however, in recent years, adipose tissue has been used more frequently because of its simplicity. Adipose tissue-derived MSCs are more easily obtained from the donor sites than bone marrow-derived MSCs and are extensively proliferative ex vivo and in vitro. However, the question arises as to whether MSCs derived from adipose tissue are fully immature and undifferentiated. If MSCs in adipose tissue are adipocyte-prone MSCs, they may revert from adipocytes to a more undifferentiated state as MSCs. The mechanisms of maintenance of MSC multipotency are still unrevealed. We believe that the differences in MSC characteristics are affected by the differences in their origin. Further research is needed to investigate the characteristic of MSCs multipotency and apply them to regenerative medicine. Specifically, MSCs derived from adipose tissue increased renal blood flow and improved renal function when administered to patients with focal segmental glomerulosclerosis [87]. The in vivo kinetics of the MSCs used in this clinical trial have not been elucidated, and the mechanism may be investigated using gene modification techniques.

3.3. MSCs Promote Wound Healing

Understanding the interaction between MSCs and wound healing is important because we believe that understanding this interaction can help prevent scarring and promote reparative healing. The mechanisms by which MSCs treatments promote wound healing include secretion of angiogenesis-promoting factors, differentiation into wound-healing cells such as fibroblasts, mobilization of intrinsic stem cells, generation and remodeling of the extracellular matrix, the polarization of M2 macrophages, and immunosuppressive effects. MSCs in the stem cell niche are known to repair damaged or dead cells by proliferating and differentiating into skin cells, but they also activate cell recovery and healing processes through an autocrine and paracrine pathway [88]. In addition, cell-cell interactions between MSCs and macrophages influence skin wound healing during MSC transplantation, but the detailed mechanisms are still unclear [89]. Exosomes play an important role in paracrine functions and are involved in wound healing by MSCs. MSC-derived exosomes have attracted attention not only for wound healing but also for their immunomodulatory functions [90]. Many studies have confirmed the anti-inflammatory [91] and wound-healing effects of MSC exosomes under various conditions. MSC exosomes may be an excellent alternative to MSC cell therapy because they are more stable and less immunogenic than derived cells while having the same biological functions as derived cells [92,93]. The use of MSC-derived exosomes is being investigated as a possible treatment modality for future clinical applications. Current research on MSC exosomes in wound healing and skin regeneration has also focused on the role of MSC exosomes in the three phases (inflammation, proliferation, and remodeling) [94]. Prolonged inflammation delays wound healing and cause scarring. Therefore, it is important to control inflammation and the transition to the proliferative phase [95]. During the inflammatory phase of the wound-healing process, MSC-derived exosomes can change the polarity of M1 macrophages to M2 macrophages, which in turn have been reported to have anti-inflammatory and immunomodulatory properties that promote wound healing [96,97]. Induced M2 macrophages secrete anti-inflammatory factors, such as IL-10 and transforming growth factor-β (TGF-β), and M2 markers, such as IL-1RA, CD163, and C-C motif chemokine 22 (CCL22) [98]. These actions are important for wound repair. These actions suppress inflammation and promote wound healing. Second, both MSC exosomes and MSCs play a role in the proliferative phase. MSC-derived exosomes enhance the proliferation and migration of human keratinocytes and dermal fibroblasts [99,100]. In addition, MSC has been shown to migrate into wounds and differentiate into fibroblasts, keratinocytes, and other wound-healing cells in the skin [15]. Finally, in the last remodeling phase, the extracellular matrix composed of type III collagen is converted to type I collagen [101]. Dermal fibroblasts produce collagen and extracellular matrix, which are ultimately important in tissue remodeling. However, if fibroblasts fail to function properly and produce excessive collagen fibers, they may eventually cause scarring [20]. The antifibrotic effects and regulation of collagen-producing fibroblast function of MSC-derived exosomes are important for the prevention of scarring, but they are not well understood. Although this review discusses MSCs and fibrosis with a focus on skin wound healing, treatments for fibrosis in other organs have also been developed using the antifibrotic properties of MSCs [102]. The extent to which MSCs influence fibroblasts during wound healing is also important (Figure 1).
When normal skin tissue is damaged by external factors, resident and/or infused MSCs migrate to the damaged area [88,103]. The migrating MSCs act on fibroblasts in the dermis to stimulate the production of appropriate amounts of collagen fibers to repair the damaged area. In addition, exosomes secreted by MSCs prevent scar formation and help the skin to return to tissue similar to normal skin tissue.

3.4. MSCs and Fibroblasts

MSCs are closely related to fibroblasts during the wound-healing process, and appropriate proliferation control is necessary because collagen regeneration by fibroblasts, especially during the proliferative and remodeling phases, can cause scar formation [20]. Fibroblasts are cells present in all tissues in the body and are largely responsible for the production of the extracellular matrix. Human bone marrow-derived MSCs express CD90, and fibroblasts express CD90′ detected in many tissues, including the human myometrium, orbit, and lung. As both cells express CD90, analysis of the expression pattern of the antigen on the surface of each cell is important to distinguish between the two. Adult CD90-positive fibroblasts exhibit stem cell characteristics and high immaturity potential. These cells are initially isolated and cultured and have the potential to differentiate into other cell types [104]. Furthermore, fibroblasts have anti-inflammatory, immunomodulatory, and regenerative properties similar to MSCs, and their application in regenerative medicine has been discussed [105,106]. It has been reported that MSCs and fibroblasts are deeply involved via macrophages and exosomes not only in wound healing and regenerative medicine but also in the field of cancer [16,17]. As the similarity in location and cell characteristics between MSCs and fibroblasts, the problem with conventional MSC isolation methods is that both types of cells can be confused. The conventional method was to isolate only whole cells from bone marrow, adipose tissue, dental pulp, and other tissues and then sow cells in flasks and culture cells to form colonies, which were then used as MSCs. However, this method cannot isolate pure MSCs, because fibroblasts also form colonies. Therefore, a more efficient MSC isolation method is required. Recently, exosomes have attracted attention in the interaction between MSCs and fibroblasts, and cell-free therapy using MSC-derived exosomes instead of MSCs has been investigated [107,108]. Several studies about MSCs’ function on fibroblasts are summarized in Table 1. Molecular biology elucidation of the interaction between MSCs and fibroblasts will provide more treatment options in the future.

3.5. MSC-Derived Exosome

Currently, MSC-derived exosomes are expected to have wound healing and anti-inflammatory effects. MSCs are thought to exert their physiological activity through paracrine action, and exosomes are the transporters responsible for this action. Extracellular vesicles are lipid-binding nanoparticles secreted from the cell into the extracellular space, and the exosome is one of the extracellular vesicles [115]. Exosomes are also released by cells other than MSCs and are responsible for intercellular communication. They are granule-like substances with a diameter of 50–150 nm secreted by cells. Its surface contains lipids and proteins derived from the cell membrane, whereas its interior contains nucleic acids (microRNA, messenger RNA, and DNA), proteins, and other intracellular substances. Vesicles with exosome characteristics can be isolated from fluids, such as semen [116], urine [117], saliva [118], blood [119], bile [120], milk [121], cerebrospinal fluid [122], and ascites [123]. Various tissue-derived MSCs secrete exosomes; however, the function of exosomes differs depending on the tissue of origin. Although only one case has been reported, bioinformatic analysis has revealed that exosomes from bone marrow-derived MSCs have a high regenerative capacity, those from adipose tissue-derived MSCs have a high immunomodulatory capacity, and those from umbilical cord-derived MSCs have high tissue damage repair capacity [124]. Bone marrow-derived MSCs exosomes can be used to treat kidney disease [125]. Exosomes from adipose tissue-derived MSCs are effective in healing skin injury, nerve regeneration, ischemia-reperfusion, parenchymal organ diseases, and obesity. In particular, adipose tissue-derived MSCs can increase type I and type III collagen production via the PI3K/Akt signaling pathway in fibroblasts. Furthermore, exosomes from adipose-derived MSCs have been found to prevent scarring, suggesting that cell-free therapy is an effective strategy [126]. Umbilical cord-derived MSCs exosomes can be used to treat liver fibrosis [127]. As the effect and mechanism of action of MSC exosomes vary depending on the tissue from which they are derived, the diseases and molecules targeted for treatment will naturally differ. For example, in an in vitro model of Alzheimer’s disease, adipose tissue-derived MSC exosomes appeared to be more effective in degrading Aβ than bone marrow-derived MSC exosomes [128]. When discussing the therapeutic effects of MSC-derived exosomes in the future, it is necessary to consider which tissues they are derived from and what mechanism of action is expected. An additional therapeutic approach that focuses on the benefits of other MSC-derived exosomes is cell-free therapy. Cell-free therapy using MSC-derived exosomes has attracted attention owing to its various advantages. In contrast to therapies that use whole cells, cell-free therapies that use MSC-derived exosomes are easier to manage and safer because they contain lower amounts of membrane-bound proteins, such as MHC molecules, and do not directly form tumors. Cell-free therapy using MSC-derived exosomes has been reported to be effective in wound healing, lung cancer, cardiovascular disease, and COVID-19 [129]. Thus, cell-free therapy is currently attracting attention as a regenerative therapy in which MSC-derived exosomes will play an important role.

3.6. Future Prospects of MSCs

Currently, research institutes and companies are developing methods for the mass culture of high-quality MSCs, but there are very few successful examples, and they cannot be applied in clinical settings. Therefore, researchers and physicians are carefully culturing flasks one by one and administering them to individual patients. The establishment of a simple and accurate mass culture method for MSCs is one of the issues that must be solved immediately. Cell sorting by flow cytometer needs not necessarily be used to harvest and isolate MSCs. However, we prefer to use a flow cytometer to collect highly pure MSCs efficiently. Crisan M et al. investigated the expression of MSC markers detected at the surface of native, non-cultured perivascular cells using a flow cytometer [130]. Although the use of a flow cytometer may have additional disadvantages in terms of cost and time, it should be useful in elucidating MSCs and further development. (Figure 2). Even when used for wound healing, MSCs will be able to be administered via intravenous infusion or topical injection without culture simultaneously [131]. A difficult administration method will cause confusion in the clinical setting and itself makes it difficult to further development. Ideally, MSCs should be administered in a manner that promotes rapid wound repair, similar to the administration of antimicrobial agents. Furthermore, it is hoped that the exosomes and wound-healing substances secreted by MSCs could be used intravenous injection instead of using MSCs in themselves, and these substances could provide to patients more conveniently due to the development of self-administration drugs.
To collect genuine MSCs, MSCs can be harvested and isolated from bone marrow and adipose tissue using a flow cytometer. Isolated MSCs can differentiate into osteocytes, adipocytes, and chondrocytes for forming the organization or self-renew to further increase the number of MSCs. MSCs themselves also secrete anti-inflammatory substances such as IL-10 and secrete exosomes to produce paracrine action.

4. Conclusions

Accelerated wound healing and proper collagen production by fibroblasts is a very important process as it prevents scar formation. MSCs have long been used in regenerative medicine owing to fewer ethical issues, relatively easy to harvest from organisms, a small risk of immune rejection if self-derived MSCs are used, smaller risk of tumor formation compared to induced pluripotent stem cells and ES cells. In recent years, MSC-derived exosomes have attracted attention for their inflammation-modulating properties, and their application in wound healing is expected. Since MSCs and fibroblasts are closely interrelated, fundamental elucidation of these mechanisms will greatly contribute to regenerative medicine, especially in plastic surgery and dermatology. However, some reports are skeptical about whether MSCs act on fibroblasts in the skin or affect wound healing [131,132,133,134]. The significance of using stem cells for chronic wound healing has been discussed, and there is a growing understanding of the mechanisms and pathophysiology.

Author Contributions

Conceptualization, K.T. and K.H.; writing—original draft preparation, K.T., R.O., S.Y., S.S. and K.H.; writing—review and editing, K.T., R.O. and K.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Birmingham, E.; Niebur, G.L.; McHugh, P.E.; Shaw, G.; Barry, F.P.; McNamara, L.M. Osteogenic differentiation of mesenchymal stem cells is regulated by osteocyte and osteoblast cells in a simplified bone niche. Eur. Cell Mater. 2012, 23, 13–27. [Google Scholar] [CrossRef] [PubMed]
  2. Han, L.; Wang, B.; Wang, R.; Gong, S.; Chen, G.; Xu, W. The shift in the balance between osteoblastogenesis and adipogenesis of mesenchymal stem cells mediated by glucocorticoid receptor. Stem Cell Res. Ther. 2019, 10, 377. [Google Scholar] [CrossRef] [PubMed]
  3. Li, X.; Wang, M.; Jing, X.; Guo, W.; Hao, C.; Zhang, Y.; Gao, S.; Chen, M.; Zhang, Z.; Zhang, X.; et al. Bone Marrow and Adipose Tissue-Derived Mesenchymal Stem Cells: Characterization, Differentiation, and Applications in Cartilage Tissue Engineering. Crit. Rev. Eukaryot. Gene Expr. 2018, 28, 285–310. [Google Scholar] [CrossRef] [PubMed]
  4. Gupta, P.K.; Das, A.K.; Chullikana, A.; Majumdar, A.S. Mesenchymal stem cells for cartilage repair in osteoarthritis. Stem Cell Res. Ther. 2012, 3, 25. [Google Scholar] [CrossRef] [PubMed]
  5. Li, S.N.; Wu, J.F. TGF-β/SMAD signaling regulation of mesenchymal stem cells in adipocyte commitment. Stem Cell Res. Ther. 2020, 11, 41. [Google Scholar] [CrossRef]
  6. Hu, L.; Yin, C.; Zhao, F.; Ali, A.; Ma, J.; Qian, A. Mesenchymal Stem Cells: Cell Fate Decision to Osteoblast or Adipocyte and Application in Osteoporosis Treatment. Int. J. Mol. Sci. 2018, 19, 360. [Google Scholar] [CrossRef]
  7. Mabuchi, Y.; Morikawa, S.; Harada, S.; Niibe, K.; Suzuki, S.; Renault-Mihara, F.; Houlihan, D.D.; Akazawa, C.; Okano, H.; Matsuzaki, Y. LNGFR(+)THY-1(+)VCAM-1(hi+) cells reveal functionally distinct subpopulations in mesenchymal stem cells. Stem Cell Rep. 2013, 1, 152–165. [Google Scholar] [CrossRef]
  8. Sacchetti, B.; Funari, A.; Michienzi, S.; Di Cesare, S.; Piersanti, S.; Saggio, I.; Tagliafico, E.; Ferrari, S.; Robey, P.G.; Riminucci, M.; et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 2007, 131, 324–336. [Google Scholar] [CrossRef]
  9. Liu, T.M.; Martina, M.; Hutmacher, D.W.; Hui, J.H.; Lee, E.H.; Lim, B. Identification of common pathways mediating differentiation of bone marrow and adipose tissue-derived human mesenchymal stem cells into three mesenchymal lineages. Stem Cells 2007, 25, 750–760. [Google Scholar] [CrossRef]
  10. Ong, W.K.; Tan, C.S.; Chan, K.L.; Goesantoso, G.G.; Chan, X.H.; Chan, E.; Yin, J.; Yeo, C.R.; Khoo, C.M.; So, J.B.; et al. Identification of specific cell-surface markers of adipose-derived stem cells from subcutaneous and visceral fat depots. Stem Cell Rep. 2014, 2, 171–179. [Google Scholar] [CrossRef]
  11. Battula, V.L.; Bareiss, P.M.; Treml, S.; Conrad, S.; Albert, I.; Hojak, S.; Abele, H.; Schewe, B.; Just, L.; Skutella, T.; et al. Human placenta and bone marrow derived MSC cultured in serum-free, b-FGF-containing medium express cell surface frizzled-9 and SSEA-4 and give rise to multilineage differentiation. Differentiation 2007, 75, 279–291. [Google Scholar] [CrossRef] [PubMed]
  12. Battula, V.L.; Treml, S.; Abele, H.; Bühring, H.J. Prospective isolation and characterization of mesenchymal stem cells from human placenta using a frizzled-9-specific monoclonal antibody. Differentiation 2008, 76, 326–336. [Google Scholar] [CrossRef] [PubMed]
  13. Gronthos, S.; Mankani, M.; Brahim, J.; Robey, P.G.; Shi, S. Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc. Natl. Acad. Sci. USA 2000, 97, 13625–13630. [Google Scholar] [CrossRef] [PubMed]
  14. Yasui, T.; Mabuchi, Y.; Toriumi, H.; Ebine, T.; Niibe, K.; Houlihan, D.D.; Morikawa, S.; Onizawa, K.; Kawana, H.; Akazawa, C.; et al. Purified Human Dental Pulp Stem Cells Promote Osteogenic Regeneration. J. Dent. Res. 2016, 95, 206–214. [Google Scholar] [CrossRef] [PubMed]
  15. Marofi, F.; Alexandrovna, K.I.; Margiana, R.; Bahramali, M.; Suksatan, W.; Abdelbasset, W.K.; Chupradit, S.; Nasimi, M.; Maashi, M.S. MSCs and their exosomes: A rapidly evolving approach in the context of cutaneous wounds therapy. Stem Cell Res. Ther. 2021, 12, 597. [Google Scholar] [CrossRef] [PubMed]
  16. Zhang, Q.; Chai, S.; Wang, W.; Wan, C.; Zhang, F.; Li, Y.; Wang, F. Macrophages activate mesenchymal stem cells to acquire cancer-associated fibroblast-like features resulting in gastric epithelial cell lesions and malignant transformation in vitro. Oncol. Lett. 2019, 17, 747–756. [Google Scholar] [CrossRef]
  17. Whiteside, T.L. Exosome and mesenchymal stem cell cross-talk in the tumor microenvironment. Semin. Immunol. 2018, 35, 69–79. [Google Scholar] [CrossRef]
  18. Rippa, A.L.; Kalabusheva, E.P.; Vorotelyak, E.A. Regeneration of Dermis: Scarring and Cells Involved. Cells 2019, 8, 607. [Google Scholar] [CrossRef]
  19. Broughton, G., 2nd; Janis, J.E.; Attinger, C.E. Wound healing: An overview. Plast. Reconstr. Surg. 2006, 117 (Suppl. 7), 1e-S–32e-S. [Google Scholar] [CrossRef]
  20. Eming, S.A.; Martin, P.; Tomic-Canic, M. Wound repair and regeneration: Mechanisms, signaling, and translation. Sci. Transl. Med. 2014, 6, 265sr6. [Google Scholar] [CrossRef]
  21. Jackson, W.M.; Nesti, L.J.; Tuan, R.S. Mesenchymal stem cell therapy for attenuation of scar formation during wound healing. Stem Cell Res. Ther. 2012, 3, 20. [Google Scholar] [CrossRef] [PubMed]
  22. Shirakami, E.; Yamakawa, S.; Hayashida, K. Strategies to prevent hypertrophic scar formation: A review of therapeutic interventions based on molecular evidence. Burns Trauma 2020, 8, tkz003. [Google Scholar] [CrossRef] [PubMed]
  23. Velnar, T.; Bailey, T.; Smrkolj, V. The wound healing process: An overview of the cellular and molecular mechanisms. J. Int. Med. Res. 2009, 37, 1528–1542. [Google Scholar] [CrossRef] [PubMed]
  24. Wilkinson, H.N.; Hardman, M.J. Wound healing: Cellular mechanisms and pathological outcomes. Open Biol. 2020, 10, 200223. [Google Scholar] [CrossRef] [PubMed]
  25. Diegelmann, R.F.; Evans, M.C. Wound healing: An overview of acute, fibrotic and delayed healing. Front. Biosci. 2004, 9, 283–289. [Google Scholar] [CrossRef] [PubMed]
  26. Krishnaswamy, S. The transition of prothrombin to thrombin. J. Thromb. Haemost. 2013, 11 (Suppl. 1), 265–276. [Google Scholar] [CrossRef]
  27. Whelihan, M.F.; Zachary, V.; Orfeo, T.; Mann, K.G. Prothrombin activation in blood coagulation: The erythrocyte contribution to thrombin generation. Blood 2012, 120, 3837–3845. [Google Scholar] [CrossRef]
  28. Weisel, J.W. Fibrinogen and fibrin. Adv. Protein Chem. 2005, 70, 247–299. [Google Scholar] [CrossRef]
  29. Werner, S.; Grose, R. Regulation of wound healing by growth factors and cytokines. Physiol. Rev. 2003, 83, 835–870. [Google Scholar] [CrossRef]
  30. Song, H.S.; Park, T.W.; Sohn, U.D.; Shin, Y.K.; Choi, B.C.; Kim, C.J.; Sim, S.S. The Effect of Caffeic Acid on Wound Healing in Skin-incised Mice. Korean J. Physiol. Pharmacol. 2008, 12, 343–347. [Google Scholar] [CrossRef]
  31. Kantapan, J.; Anukul, N.; Leetrakool, N.; Rolin, G.; Vergote, J.; Dechsupa, N. Iron-Quercetin Complex Preconditioning of Human Peripheral Blood Mononuclear Cells Accelerates Angiogenic and Fibroblast Migration: Implications for Wound Healing. Int. J. Mol. Sci. 2021, 22, 8851. [Google Scholar] [CrossRef]
  32. Werner, S.; Krieg, T.; Smola, H. Keratinocyte-fibroblast interactions in wound healing. J. Investig. Dermatol. 2007, 127, 998–1008. [Google Scholar] [CrossRef] [PubMed]
  33. Chu, H.; Wang, Y.; Wang, X.; Song, X.; Liu, H.; Li, X. Effects of transplanted adipose derived stem cells on the expressions of α-SMA and DCN in fibroblasts of hypertrophic scar tissues in rabbit ears. Exp. Ther. Med. 2018, 16, 1729–1734. [Google Scholar] [CrossRef] [PubMed]
  34. Rabello, F.B.; Souza, C.D.; Farina Júnior, J.A. Update on hypertrophic scar treatment. Clinics 2014, 69, 565–573. [Google Scholar] [CrossRef]
  35. Ogawa, R. Keloid and Hypertrophic Scars Are the Result of Chronic Inflammation in the Reticular Dermis. Int. J. Mol. Sci. 2017, 18, 606. [Google Scholar] [CrossRef]
  36. Steinstraesser, L.; Flak, E.; Witte, B.; Ring, A.; Tilkorn, D.; Hauser, J.; Langer, S.; Steinau, H.U.; Al-Benna, S. Pressure garment therapy alone and in combination with silicone for the prevention of hypertrophic scarring: Randomized controlled trial with intraindividual comparison. Plast. Reconstr. Surg. 2011, 128, 306e–313e. [Google Scholar] [CrossRef]
  37. Wang, M.; Xu, X.; Lei, X.; Tan, J.; Xie, H. Mesenchymal stem cell-based therapy for burn wound healing. Burns Trauma 2021, 9, tkab002. [Google Scholar] [CrossRef]
  38. Mushahary, D.; Spittler, A.; Kasper, C.; Weber, V.; Charwat, V. Isolation, cultivation, and characterization of human mesenchymal stem cells. Cytometry A 2018, 93, 19–31. [Google Scholar] [CrossRef]
  39. Piersma, A.H.; Brockbank, K.G.; Ploemacher, R.E.; van Vliet, E.; Brakel-van Peer, K.M.; Visser, P.J. Characterization of fibroblastic stromal cells from murine bone marrow. Exp. Hematol. 1985, 13, 237–243. [Google Scholar]
  40. Caplan, A.I. Mesenchymal stem cells. J. Orthop. Res. 1991, 9, 641–650. [Google Scholar] [CrossRef]
  41. Amiri, F.; Halabian, R.; Dehgan Harati, M.; Bahadori, M.; Mehdipour, A.; Mohammadi Roushandeh, A.; Habibi Roudkenar, M. Positive selection of Wharton’s jelly-derived CD105(+) cells by MACS technique and their subsequent cultivation under suspension culture condition: A simple, versatile culturing method to enhance the multipotentiality of mesenchymal stem cells. Hematology 2015, 20, 208–216. [Google Scholar] [CrossRef] [PubMed]
  42. Vieira, C.P.; McCarrel, T.M.; Grant, M.B. Novel Methods to Mobilize, Isolate, and Expand Mesenchymal Stem Cells. Int. J. Mol. Sci. 2021, 22, 5728. [Google Scholar] [CrossRef] [PubMed]
  43. Saleh, M.; Fotook Kiaei, S.Z.; Kavianpour, M. Application of Wharton jelly-derived mesenchymal stem cells in patients with pulmonary fibrosis. Stem Cell Res. Ther. 2022, 13, 71. [Google Scholar] [CrossRef]
  44. 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]
  45. Wang, X.; Wang, Y.; Gou, W.; Lu, Q.; Peng, J.; Lu, S. Role of mesenchymal stem cells in bone regeneration and fracture repair: A review. Int. Orthop. 2013, 37, 2491–2498. [Google Scholar] [CrossRef]
  46. Shekkeris, A.S.; Jaiswal, P.K.; Khan, W.S. Clinical applications of mesenchymal stem cells in the treatment of fracture non-union and bone defects. Curr. Stem Cell Res. Ther. 2012, 7, 127–133. [Google Scholar] [CrossRef] [PubMed]
  47. Lopez-Santalla, M.; Fernandez-Perez, R.; Garin, M.I. Mesenchymal Stem/Stromal Cells for Rheumatoid Arthritis Treatment: An Update on Clinical Applications. Cells 2020, 9, 1852. [Google Scholar] [CrossRef]
  48. Nifosì, G.; Nifosì, L.; Nifosì, A.F. Mesenchymal stem cells in the treatment of osteonecrosis of the jaw. J. Korean Assoc. Oral. Maxillofac. Surg. 2021, 47, 65–75. [Google Scholar] [CrossRef]
  49. Escobedo, M.F.; Junquera, S.; Gonzalez, C.; Vasatyuk, S.; Gallego, L.; Barbeito, E.; Junquera, L.M. Efficacy of complementary treatment with autologous platelet concentrates and/or mesenchymal stem cells in chemical osteonecrosis of the jaw. Systematic review of the literature. J. Stomatol. Oral Maxillofac. Surg. 2022, 123, 51–58. [Google Scholar] [CrossRef]
  50. Voss, P.J.; Matsumoto, A.; Alvarado, E.; Schmelzeisen, R.; Duttenhöfer, F.; Poxleitner, P. Treatment of stage II medication-related osteonecrosis of the jaw with necrosectomy and autologous bone marrow mesenchymal stem cells. Odontology 2017, 105, 484–493. [Google Scholar] [CrossRef]
  51. Zhao, L.; Chen, S.; Yang, P.; Cao, H.; Li, L. The role of mesenchymal stem cells in hematopoietic stem cell transplantation: Prevention and treatment of graft-versus-host disease. Stem Cell Res. Ther. 2019, 10, 182. [Google Scholar] [CrossRef] [PubMed]
  52. Włodarczyk, M.; Czerwińska, K.; Włodarczyk, J.; Fichna, J.; Dziki, A.; Dziki, Ł. Current Overview on the Use of Mesenchymal Stem Cells for Perianal Fistula Treatment in Patients with Crohn’s Disease. Life 2021, 11, 1133. [Google Scholar] [CrossRef] [PubMed]
  53. Holloman, J.P.; Ho, C.C.; Hukki, A.; Huntley, J.L.; Gallicano, G.I. The development of hematopoietic and mesenchymal stem cell transplantation as an effective treatment for multiple sclerosis. Am. J. Stem Cells 2013, 2, 95–107. [Google Scholar] [PubMed]
  54. Wang, F.; Tang, H.; Zhu, J.; Zhang, J.H. Transplanting Mesenchymal Stem Cells for Treatment of Ischemic Stroke. Cell Transplant. 2018, 27, 1825–1834. [Google Scholar] [CrossRef] [PubMed]
  55. Shafei, A.E.; Ali, M.A.; Ghanem, H.G.; Shehata, A.I.; Abdelgawad, A.A.; Handal, H.R.; Talaat, K.A.; Ashaal, A.E.; El-Shal, A.S. Mesenchymal stem cell therapy: A promising cell-based therapy for treatment of myocardial infarction. J. Gene Med. 2017, 19, e2995. [Google Scholar] [CrossRef] [PubMed]
  56. Cao, Y.; Ji, C.; Lu, L. Mesenchymal stem cell therapy for liver fibrosis/cirrhosis. Ann. Transl. Med. 2020, 8, 562. [Google Scholar] [CrossRef]
  57. Wecht, S.; Rojas, M. Mesenchymal stem cells in the treatment of chronic lung disease. Respirology 2016, 21, 1366–1375. [Google Scholar] [CrossRef]
  58. Misselwitz, B.; Juillerat, P.; Sulz, M.C.; Siegmund, B.; Brand, S. Swiss IBDnet, an official working group of the Swiss Society of Gastroenterology. Emerging Treatment Options in Inflammatory Bowel Disease: Janus Kinases, Stem Cells, and More. Digestion 2020, 101 (Suppl. 1), 69–82. [Google Scholar] [CrossRef]
  59. Chen, Y.; Yu, Q.; Hu, Y.; Shi, Y. Current Research and Use of Mesenchymal Stem Cells in the Therapy of Autoimmune Diseases. Curr. Stem Cell Res. Ther. 2019, 14, 579–582. [Google Scholar] [CrossRef]
  60. Samsonraj, R.M.; Raghunath, M.; Nurcombe, V.; Hui, J.H.; van Wijnen, A.J.; Cool, S.M. Concise Review: Multifaceted Characterization of Human Mesenchymal Stem Cells for Use in Regenerative Medicine. Stem. Cells Transl. Med. 2017, 6, 2173–2185. [Google Scholar] [CrossRef]
  61. Jung, J.W.; Kwon, M.; Choi, J.C.; Shin, J.W.; Park, I.W.; Choi, B.W.; Kim, J.Y. Familial occurrence of pulmonary embolism after intravenous, adipose tissue-derived stem cell therapy. Yonsei Med. J. 2013, 54, 1293–1296. [Google Scholar] [CrossRef] [PubMed]
  62. Peng, X.; Li, J.; Yu, X.; Tan, R.; Zhu, L.; Wang, J.; Wang, R.; Gu, G.; Liu, Q.; Ren, L.; et al. Therapeutic effectiveness of bone marrow-derived mesenchymal stem cell administration against acute pulmonary thromboembolism in a mouse model. Thromb. Res. 2015, 135, 990–999. [Google Scholar] [CrossRef] [PubMed]
  63. Lv, F.J.; Tuan, R.S.; Cheung, K.M.; Leung, V.Y. Concise review: The surface markers and identity of human mesenchymal stem cells. Stem Cells 2014, 32, 1408–1419. [Google Scholar] [CrossRef] [PubMed]
  64. Andrzejewska, A.; Dabrowska, S.; Lukomska, B.; Janowski, M. Mesenchymal Stem Cells for Neurological Disorders. Adv. Sci. 2021, 8, 2002944. [Google Scholar] [CrossRef] [PubMed]
  65. Liu, T.M. Application of mesenchymal stem cells derived from human pluripotent stem cells in regenerative medicine. World J. Stem Cells 2021, 13, 1826–1844. [Google Scholar] [CrossRef]
  66. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.J.; 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]
  67. Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal, R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti, D.W.; Craig, S.; Marshak, D.R. Multilineage potential of adult human mesenchymal stem cells. Science 1999, 284, 143–147. [Google Scholar] [CrossRef]
  68. Kfoury, Y.; Scadden, D.T. Mesenchymal cell contributions to the stem cell niche. Cell Stem Cell 2015, 16, 239–253. [Google Scholar] [CrossRef]
  69. Yu, V.W.; Scadden, D.T. Hematopoietic Stem Cell and Its Bone Marrow Niche. Curr. Top. Dev. Biol. 2016, 118, 21–44. [Google Scholar] [CrossRef]
  70. Morrison, S.J.; Scadden, D.T. The bone marrow niche for haematopoietic stem cells. Nature 2014, 505, 327–334. [Google Scholar] [CrossRef]
  71. Baker, N.; Boyette, L.B.; Tuan, R.S. Characterization of bone marrow-derived mesenchymal stem cells in aging. Bone 2015, 70, 37–47. [Google Scholar] [CrossRef] [PubMed]
  72. Houlihan, D.D.; Mabuchi, Y.; Morikawa, S.; Niibe, K.; Araki, D.; Suzuki, S.; Okano, H.; Matsuzaki, Y. Isolation of mouse mesenchymal stem cells on the basis of expression of Sca-1 and PDGFR-α. Nat. Protoc. 2012, 7, 2103–2111. [Google Scholar] [CrossRef] [PubMed]
  73. Sudo, K.; Kanno, M.; Miharada, K.; Ogawa, S.; Hiroyama, T.; Saijo, K.; Nakamura, Y. Mesenchymal progenitors able to differentiate into osteogenic, chondrogenic, and/or adipogenic cells in vitro are present in most primary fibroblast-like cell populations. Stem Cells 2007, 25, 1610–1617. [Google Scholar] [CrossRef] [PubMed]
  74. Denu, R.A.; Nemcek, S.; Bloom, D.D.; Goodrich, A.D.; Kim, J.; Mosher, D.F.; Hematti, P. Fibroblasts and Mesenchymal Stromal/Stem Cells Are Phenotypically Indistinguishable. Acta Haematol. 2016, 136, 85–97. [Google Scholar] [CrossRef]
  75. Alt, E.; Yan, Y.; Gehmert, S.; Song, Y.H.; Altman, A.; Gehmert, S.; Vykoukal, D.; Bai, X. Fibroblasts share mesenchymal phenotypes with stem cells, but lack their differentiation and colony-forming potential. Biol. Cell 2011, 103, 197–208. [Google Scholar] [CrossRef]
  76. Soundararajan, M.; Kannan, S. Fibroblasts and mesenchymal stem cells: Two sides of the same coin? J. Cell Physiol. 2018, 233, 9099–9109. [Google Scholar] [CrossRef]
  77. Wu, S.; Coombs, D.M.; Gurunian, R. Liposuction: Concepts, safety, and techniques in body-contouring surgery. Cleve. Clin. J. Med. 2020, 87, 367–375. [Google Scholar] [CrossRef]
  78. Ahmad, J.; Eaves, F.F., 3rd; Rohrich, R.J.; Kenkel, J.M. The American Society for Aesthetic Plastic Surgery (ASAPS) survey: Current trends in liposuction. Aesthet. Surg. J. 2011, 31, 214–224. [Google Scholar] [CrossRef]
  79. Nakagami, H.; Morishita, R.; Maeda, K.; Kikuchi, Y.; Ogihara, T.; Kaneda, Y. Adipose tissue-derived stromal cells as a novel option for regenerative cell therapy. J. Atheroscler. Thromb. 2006, 13, 77–81. [Google Scholar] [CrossRef]
  80. Konno, M.; Hamabe, A.; Hasegawa, S.; Ogawa, H.; Fukusumi, T.; Nishikawa, S.; Ohta, K.; Kano, Y.; Ozaki, M.; Noguchi, Y.; et al. Adipose-derived mesenchymal stem cells and regenerative medicine. Dev. Growth Differ. 2013, 55, 309–318. [Google Scholar] [CrossRef]
  81. Moghadam, F.H.; Alaie, H.; Karbalaie, K.; Tanhaei, S.; Nasr Esfahani, M.H.; Baharvand, H. Transplantation of primed or unprimed mouse embryonic stem cell-derived neural precursor cells improves cognitive function in Alzheimerian rats. Differentiation 2009, 78, 59–68. [Google Scholar] [CrossRef] [PubMed]
  82. Ikhapoh, I.A.; Pelham, C.J.; Agrawal, D.K. Sry-type HMG box 18 contributes to the differentiation of bone marrow-derived mesenchymal stem cells to endothelial cells. Differentiation 2015, 89, 87–96. [Google Scholar] [CrossRef] [PubMed]
  83. Lee, J.H.; Kosinski, P.A.; Kemp, D.M. Contribution of human bone marrow stem cells to individual skeletal myotubes followed by myogenic gene activation. Exp. Cell Res. 2005, 307, 174–182. [Google Scholar] [CrossRef]
  84. Banas, A.; Teratani, T.; Yamamoto, Y.; Tokuhara, M.; Takeshita, F.; Quinn, G.; Okochi, H.; Ochiya, T. Adipose tissue-derived mesenchymal stem cells as a source of human hepatocytes. Hepatology 2007, 46, 219–228. [Google Scholar] [CrossRef] [PubMed]
  85. Bunnell, B.A. Adipose Tissue-Derived Mesenchymal Stem Cells. Cells 2021, 10, 3433. [Google Scholar] [CrossRef] [PubMed]
  86. Strioga, M.; Viswanathan, S.; Darinskas, A.; Slaby, O.; Michalek, J. Same or not the same? Comparison of adipose tissue-derived versus bone marrow-derived mesenchymal stem and stromal cells. Stem Cells Dev. 2012, 21, 2724–2752. [Google Scholar] [CrossRef]
  87. Saad, A.; Dietz, A.B.; Herrmann, S.M.S.; Hickson, L.J.; Glockner, J.F.; McKusick, M.A.; Misra, S.; Bjarnason, H.; Armstrong, A.S.; Gastineau, D.A.; et al. Autologous Mesenchymal Stem Cells Increase Cortical Perfusion in Renovascular Disease. J. Am. Soc. Nephrol. 2017, 28, 2777–2785. [Google Scholar] [CrossRef]
  88. Mazini, L.; Rochette, L.; Admou, B.; Amal, S.; Malka, G. Hopes and Limits of Adipose-Derived Stem Cells (ADSCs) and Mesenchymal Stem Cells (MSCs) in Wound Healing. Int. J. Mol. Sci. 2020, 21, 1306. [Google Scholar] [CrossRef]
  89. He, X.; Dong, Z.; Cao, Y.; Wang, H.; Liu, S.; Liao, L.; Jin, Y.; Yuan, L.; Li, B. MSC-Derived Exosome Promotes M2 Polarization and Enhances Cutaneous Wound Healing. Stem Cells Int. 2019, 2019, 7132708. [Google Scholar] [CrossRef]
  90. Ha, D.H.; Kim, H.K.; Lee, J.; Kwon, H.H.; Park, G.H.; Yang, S.H.; Jung, J.Y.; Choi, H.; Lee, J.H.; Sung, S.; et al. Mesenchymal Stem/Stromal Cell-Derived Exosomes for Immunomodulatory Therapeutics and Skin Regeneration. Cells 2020, 9, 1157. [Google Scholar] [CrossRef]
  91. Arabpour, M.; Saghazadeh, A.; Rezaei, N. Anti-inflammatory and M2 macrophage polarization-promoting effect of mesenchymal stem cell-derived exosomes. Int. Immunopharmacol. 2021, 97, 107823. [Google Scholar] [CrossRef] [PubMed]
  92. Wu, P.; Zhang, B.; Shi, H.; Qian, H.; Xu, W. MSC-exosome: A novel cell-free therapy for cutaneous regeneration. Cytotherapy 2018, 20, 291–301. [Google Scholar] [CrossRef] [PubMed]
  93. Yu, B.; Zhang, X.; Li, X. Exosomes derived from mesenchymal stem cells. Int. J. Mol. Sci. 2014, 15, 4142–4157. [Google Scholar] [CrossRef] [PubMed]
  94. Hu, P.; Yang, Q.; Wang, Q.; Shi, C.; Wang, D.; Armato, U.; Prà, I.D.; Chiarini, A. Mesenchymal stromal cells-exosomes: A promising cell-free therapeutic tool for wound healing and cutaneous regeneration. Burns Trauma 2019, 7, 38. [Google Scholar] [CrossRef]
  95. Landén, N.X.; Li, D.; Ståhle, M. Transition from inflammation to proliferation: A critical step during wound healing. Cell Mol. Life Sci. 2016, 73, 3861–3885. [Google Scholar] [CrossRef]
  96. Nakao, Y.; Fukuda, T.; Zhang, Q.; Sanui, T.; Shinjo, T.; Kou, X.; Chen, C.; Liu, D.; Watanabe, Y.; Hayashi, C.; et al. Exosomes from TNF-α-treated human gingiva-derived MSCs enhance M2 macrophage polarization and inhibit periodontal bone loss. Acta Biomater. 2021, 122, 306–324. [Google Scholar] [CrossRef]
  97. Liu, W.; Yu, M.; Xie, D.; Wang, L.; Ye, C.; Zhu, Q.; Liu, F.; Yang, L. Melatonin-stimulated MSC-derived exosomes improve diabetic wound healing through regulating macrophage M1 and M2 polarization by targeting the PTEN/AKT pathway. Stem Cell Res. Ther. 2020, 11, 259. [Google Scholar] [CrossRef]
  98. Shapouri-Moghaddam, A.; Mohammadian, S.; Vazini, H.; Taghadosi, M.; Esmaeili, S.A.; Mardani, F.; Seifi, B.; Mohammadi, A.; Afshari, J.T.; Sahebkar, A. Macrophage plasticity, polarization, and function in health and disease. J. Cell Physiol. 2018, 233, 6425–6440. [Google Scholar] [CrossRef]
  99. Tutuianu, R.; Rosca, A.M.; Iacomi, D.M.; Simionescu, M.; Titorencu, I. Human Mesenchymal Stromal Cell-Derived Exosomes Promote In vitro Wound Healing by Modulating the Biological Properties of Skin Keratinocytes and Fibroblasts and Stimulating Angiogenesis. Int. J. Mol. Sci. 2021, 22, 6239. [Google Scholar] [CrossRef]
  100. Kim, S.; Lee, S.K.; Kim, H.; Kim, T.M. Exosomes Secreted from Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells Accelerate Skin Cell Proliferation. Int. J. Mol. Sci. 2018, 19, 3119. [Google Scholar] [CrossRef]
  101. Casado-Díaz, A.; Quesada-Gómez, J.M.; Dorado, G. Extracellular Vesicles Derived From Mesenchymal Stem Cells (MSC) in Regenerative Medicine: Applications in Skin Wound Healing. Front. Bioeng. Biotechnol. 2020, 8, 146. [Google Scholar] [CrossRef] [PubMed]
  102. Usunier, B.; Benderitter, M.; Tamarat, R.; Chapel, A. Management of fibrosis: The mesenchymal stromal cells breakthrough. Stem Cells Int. 2014, 2014, 340257. [Google Scholar] [CrossRef] [PubMed]
  103. Lynch, M.D.; Watt, F.M. Fibroblast heterogeneity: Implications for human disease. J. Clin. Investig. 2018, 128, 26–35. [Google Scholar] [CrossRef] [PubMed]
  104. Chang, Y.; Li, H.; Guo, Z. Mesenchymal stem cell-like properties in fibroblasts. Cell Physiol. Biochem. 2014, 34, 703–714. [Google Scholar] [CrossRef]
  105. Ichim, T.E.; O’Heeron, P.; Kesari, S. Fibroblasts as a practical alternative to mesenchymal stem cells. J. Transl. Med. 2018, 16, 212. [Google Scholar] [CrossRef]
  106. Weiss, R.A. Autologous cell therapy: Will it replace dermal fillers? Facial Plast. Surg. Clin. N. Am. 2013, 21, 299–304. [Google Scholar] [CrossRef]
  107. Huang, J.; Zhang, J.; Xiong, J.; Sun, S.; Xia, J.; Yang, L.; Liang, Y. Stem Cell-Derived Nanovesicles: A Novel Cell-Free Therapy for Wound Healing. Stem Cells Int. 2021, 2021, 1285087. [Google Scholar] [CrossRef]
  108. Roşca, A.M.; Ţuţuianu, R.; Titorencu, I.D. Mesenchymal stromal cells derived exosomes as tools for chronic wound healing therapy. Rom. J. Morphol. Embryol. 2018, 59, 655–662. [Google Scholar]
  109. Saheli, M.; Bayat, M.; Ganji, R.; Hendudari, F.; Kheirjou, R.; Pakzad, M.; Najar, B.; Piryaei, A. Human mesenchymal stem cells-conditioned medium improves diabetic wound healing mainly through modulating fibroblast behaviors. Arch. Dermatol. Res. 2020, 312, 325–336. [Google Scholar] [CrossRef]
  110. You, D.H.; Nam, M.J. Effects of human epidermal growth factor gene-transfected mesenchymal stem cells on fibroblast migration and proliferation. Cell Prolif. 2013, 46, 408–415. [Google Scholar] [CrossRef]
  111. Yuan, B.; Broadbent, J.A.; Huan, J.; Yang, H. The Effects of Adipose Stem Cell-Conditioned Media on Fibrogenesis of Dermal Fibroblasts Stimulated by Transforming Growth Factor-β1. J. Burn Care Res. 2018, 39, 129–140. [Google Scholar] [CrossRef]
  112. Lee, M.J.; Kim, J.; Lee, K.I.; Shin, J.M.; Chae, J.I.; Chung, H.M. Enhancement of wound healing by secretory factors of endothelial precursor cells derived from human embryonic stem cells. Cytotherapy 2011, 13, 165–178. [Google Scholar] [CrossRef] [PubMed]
  113. He, D.; Zhao, F.; Jiang, H.; Kang, Y.; Song, Y.; Lin, X.; Shi, P.; Zhang, T.; Pang, X. LOXL2 from human amniotic mesenchymal stem cells accelerates wound epithelialization by promoting differentiation and migration of keratinocytes. Aging 2020, 12, 12960–12986. [Google Scholar] [CrossRef] [PubMed]
  114. Fong, C.Y.; Tam, K.; Cheyyatraivendran, S.; Gan, S.U.; Gauthaman, K.; Armugam, A.; Jeyaseelan, K.; Choolani, M.; Biswas, A.; Bongso, A. Human Wharton’s jelly stem cells and its conditioned medium enhance healing of excisional and diabetic wounds. J. Cell Biochem. 2014, 115, 290–302. [Google Scholar] [CrossRef] [PubMed]
  115. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef] [PubMed]
  116. Ouattara, L.A.; Anderson, S.M.; Doncel, G.F. Seminal exosomes and HIV-1 transmission. Andrologia 2018, 50, e13220. [Google Scholar] [CrossRef] [PubMed]
  117. Yu, W.; Hurley, J.; Roberts, D.; Chakrabortty, S.K.; Enderle, D.; Noerholm, M.; Breakefield, X.O.; Skog, J.K. Exosome-based liquid biopsies in cancer: Opportunities and challenges. Ann. Oncol. 2021, 32, 466–477. [Google Scholar] [CrossRef] [PubMed]
  118. Kaczor-Urbanowicz, K.E.; Wei, F.; Rao, S.L.; Kim, J.; Shin, H.; Cheng, J.; Tu, M.; Wong, D.T.W.; Kim, Y. Clinical validity of saliva and novel technology for cancer detection. Biochim. Biophys. Acta Rev. Cancer 2019, 1872, 49–59. [Google Scholar] [CrossRef]
  119. Klingeborn, M.; Skiba, N.P.; Stamer, W.D.; Bowes Rickman, C. Isolation of Retinal Exosome Biomarkers from Blood by Targeted Immunocapture. Adv. Exp. Med. Biol. 2019, 1185, 21–25. [Google Scholar] [CrossRef]
  120. Yao, Y.; Jiao, D.; Li, Z.; Zhou, X.; Li, J.; Liu, Z.; Han, X. Roles of Bile-Derived Exosomes in Hepatobiliary Disease. Biomed. Res. Int. 2021, 2021, 8743409. [Google Scholar] [CrossRef]
  121. Kim, K.U.; Kim, W.H.; Jeong, C.H.; Yi, D.Y.; Min, H. More than Nutrition: Therapeutic Potential of Breast Milk-Derived Exosomes in Cancer. Int. J. Mol. Sci. 2020, 21, 7327. [Google Scholar] [CrossRef]
  122. Martinez, B.; Peplow, P.V. MicroRNAs in blood and cerebrospinal fluid as diagnostic biomarkers of multiple sclerosis and to monitor disease progression. Neural Regen. Res. 2020, 15, 606–619. [Google Scholar] [CrossRef] [PubMed]
  123. Andre, F.; Schartz, N.E.; Movassagh, M.; Flament, C.; Pautier, P.; Morice, P.; Pomel, C.; Lhomme, C.; Escudier, B.; Le Chevalier, T.; et al. Malignant effusions and immunogenic tumour-derived exosomes. Lancet 2002, 360, 295–305. [Google Scholar] [CrossRef]
  124. Wang, Z.G.; He, Z.Y.; Liang, S.; Yang, Q.; Cheng, P.; Chen, A.M. Comprehensive proteomic analysis of exosomes derived from human bone marrow, adipose tissue, and umbilical cord mesenchymal stem cells. Stem Cell Res. Ther. 2020, 11, 511. [Google Scholar] [CrossRef] [PubMed]
  125. Bruno, S.; Grange, C.; Deregibus, M.C.; Calogero, R.A.; Saviozzi, S.; Collino, F.; Morando, L.; Busca, A.; Falda, M.; Bussolati, B.; et al. Mesenchymal stem cell-derived microvesicles protect against acute tubular injury. J. Am. Soc. Nephrol. 2009, 20, 1053–1067. [Google Scholar] [CrossRef]
  126. Hong, P.; Yang, H.; Wu, Y.; Li, K.; Tang, Z. The functions and clinical application potential of exosomes derived from adipose mesenchymal stem cells: A comprehensive review. Stem Cell Res. Ther. 2019, 10, 242. [Google Scholar] [CrossRef] [PubMed]
  127. Li, T.; Yan, Y.; Wang, B.; Qian, H.; Zhang, X.; Shen, L.; Wang, M.; Zhou, Y.; Zhu, W.; Li, W.; et al. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate liver fibrosis. Stem Cells Dev. 2013, 22, 845–854. [Google Scholar] [CrossRef] [PubMed]
  128. Katsuda, T.; Tsuchiya, R.; Kosaka, N.; Yoshioka, Y.; Takagaki, K.; Oki, K.; Takeshita, F.; Sakai, Y.; Kuroda, M.; Ochiya, T. Human adipose tissue-derived mesenchymal stem cells secrete functional neprilysin-bound exosomes. Sci. Rep. 2013, 3, 1197. [Google Scholar] [CrossRef]
  129. Rezakhani, L.; Kelishadrokhi, A.F.; Soleimanizadeh, A.; Rahmati, S. Mesenchymal stem cell (MSC)-derived exosomes as a cell-free therapy for patients Infected with COVID-19: Real opportunities and range of promises. Chem. Phys. Lipids 2021, 234, 105009. [Google Scholar] [CrossRef]
  130. Crisan, M.; Yap, S.; Casteilla, L.; Chen, C.W.; Corselli, M.; Park, T.S.; Andriolo, G.; Sun, B.; Zheng, B.; Zhang, L.; et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 2008, 3, 301–313. [Google Scholar] [CrossRef]
  131. Akita, S.; Yoshimoto, H.; Akino, K.; Ohtsuru, A.; Hayashida, K.; Hirano, A.; Suzuki, K.; Yamashita, S. Early experiences with stem cells in treating chronic wounds. Clin. Plast. Surg. 2012, 39, 281–292. [Google Scholar] [CrossRef] [PubMed]
  132. Rafii, S.; Lyden, D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat. Med. 2003, 9, 702–712. [Google Scholar] [CrossRef] [PubMed]
  133. Rognoni, E.; Watt, F.M.M. Skin Cell Heterogeneity in Development, Wound Healing, and Cancer. Trends Cell Biol. 2018, 28, 709–722. [Google Scholar] [CrossRef] [PubMed]
  134. Sabbagh, F.; Kim, B.S. Recent advances in polymeric transdermal drug delivery systems. J. Control. Release 2022, 341, 132–146. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mesenchymal stem cells promote cutaneous wound healing.
Figure 1. Mesenchymal stem cells promote cutaneous wound healing.
Biomedicines 10 01391 g001
Figure 2. Isolation and differentiation of high-grade mesenchymal stem cells.
Figure 2. Isolation and differentiation of high-grade mesenchymal stem cells.
Biomedicines 10 01391 g002
Table 1. MSC functions on fibroblast under different conditions.
Table 1. MSC functions on fibroblast under different conditions.
Experiment SituationBiological SignalsEffectsRef.
Human bone marrow-derived MSC-CM in wound healing in DMEGF, bFGFIncrease proliferation, cell viability, and migration of fibroblast[109]
EGF transferred umbilical cord blood-derived MSCβ-catenin, N-cadherin, cofilin, ezrin,
phospho-MAPK/CDK substrate,
phospho-Arg-(Ser)-X-Tyr/Phe-X-pSer motif
Increase cell adhesion, dynamic effects, migration, and proliferation of fibroblast[110]
Adipose-derived MSC in wound healingTGF-βIncrease collagen production and inhibit fibroblast proliferation by avoiding excessive fibrogenesis[111]
Human ES cell-derived endothelial precursor cells in cutaneous excisional wound modelsEGF, bFGFIncrease proliferation and migration of fibroblast[112]
Human amniotic mesenchymal stem cellsLOXL2LOXL2 significantly enhanced in vitro keratinocyte migration and differentiation[113]
Human umbilical cord Wharton’s jelly-derived MSC in DMCytokeratin, involucrin, filaggrin,
ICAM-1, TIMP-1, and VEGF-A
Increase the number of invaded cells, cell viability, total collagen, elastin, and fibronectin levels[114]
Abbreviations: bFGF, basic fibroblast growth factor; CDK, cyclin-dependent kinase; CM, conditioned medium; DM, diabetes mellitus; EGF, epidermal growth factor; ES, embryonic stem; ICAM-1, intercellular adhesion molecule 1; LOXL2, lysyl oxidase-like 2; MAPK, mitogen-activated protein kinase; MSC, mesenchymal stem cell; TGF-β, transforming growth factor-β; TIMP-1, tissue inhibitors of metalloproteinases-1; VEGF-A, vascular endothelial growth factor-A.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tanaka, K.; Ogino, R.; Yamakawa, S.; Suda, S.; Hayashida, K. Role and Function of Mesenchymal Stem Cells on Fibroblast in Cutaneous Wound Healing. Biomedicines 2022, 10, 1391. https://doi.org/10.3390/biomedicines10061391

AMA Style

Tanaka K, Ogino R, Yamakawa S, Suda S, Hayashida K. Role and Function of Mesenchymal Stem Cells on Fibroblast in Cutaneous Wound Healing. Biomedicines. 2022; 10(6):1391. https://doi.org/10.3390/biomedicines10061391

Chicago/Turabian Style

Tanaka, Kotaro, Ryohei Ogino, Sho Yamakawa, Shota Suda, and Kenji Hayashida. 2022. "Role and Function of Mesenchymal Stem Cells on Fibroblast in Cutaneous Wound Healing" Biomedicines 10, no. 6: 1391. https://doi.org/10.3390/biomedicines10061391

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