Abstract
Mesenchymal stromal cells (MSCs) are multi-potent stromal-derived cells capable of self-renewal that possess several advantageous properties for wound healing, making them of interest to the field of dermatology. Research has focused on characterizing the unique properties of MSCs, which broadly revolve around their regenerative and more recently discovered immunomodulatory capacities. Because of ease of harvesting and expansion, differentiation potential and low immunogenicity, MSCs have been leading candidates for tissue engineering and regenerative medicine applications for wound healing, yet results from clinical studies have been variable, and promising pre-clinical work has been difficult to reproduce. Therefore, the specific mechanisms of how MSCs influence the local microenvironment in distinct wound etiologies warrant further research. Of specific interest in MSC-mediated healing is harnessing the secretome, which is composed of components known to positively influence wound healing. Molecules released by the MSC secretome can promote re-epithelialization and angiogenesis while inhibiting fibrosis and microbial invasion. This review focuses on the therapeutic interest in MSCs with regard to wound healing applications, including burns and diabetic ulcers, with specific attention to the genetic skin disease recessive dystrophic epidermolysis bullosa. This review also compares various delivery methods to support skin regeneration in the hopes of combating the poor engraftment of MSCs after delivery, which is one of the major pitfalls in clinical studies utilizing MSCs.
Keywords: epidermolysis bullosa, immunomodulation, MSCs, regenerative medicine, skin, wound healing
Introduction
Mesenchymal stromal cells (MSCs) are of therapeutic interest for chronic wound healing because of their extensive regenerative and immunomodulatory capacities. They demonstrate unique features, including facile culture expansion, multi-potency, low immunogenicity and positive impact on the wound healing processes of re-epithelialization and angiogenesis. Efforts to fully elucidate characteristics of MSC subtypes and capitalize on the promise of MSCs in clinical applications for chronic wound healing are ongoing.
Chronic or recurrent cutaneous wounds are the phenotypic hallmark of the heterogeneous inherited skin disease epidermolysis bullosa (EB). In a severe generalized form of genodermatosis, recessive dystrophic epidermolysis bullosa (RDEB), biallelic mutations in the COL7A1 gene result in absent or dysfunctional type VII collagen (C7). In healthy individuals, secreted C7 homotrimerizes to form anchoring fibrils in the basement membrane zone (BMZ) connecting the epidermal and dermal layers of the skin. Without these protein anchors to hold the two layers together, the skin easily separates into either blisters or wounds with mild mechanical trauma. Therapeutic options for wound management in RDEB are limited to palliative care with analgesia and protective bandaging. Based on their wound-healing properties, MSC-based therapies may represent a promising therapy in the treatment of the skin manifestations of RDEB.
This review aims to characterize the therapeutic advantages of MSCs in the context of wound healing with special attention to RDEB. RDEB is unique, in that it has characteristics of a burn yet is non-transient in nature. Treatment of burns and diabetic ulcers with MSCs will also be discussed, as this has been thoroughly described in the literature and the findings may be relevant to RDEB. The interplay of stromal and epithelial, mesenchymal and ectodermal elements in RDEB wounds makes this an opportunity to gain new knowledge about the regenerative response in genodermatoses, thermal/chemical burns and diabetic ulcers alike.
In this review, the authors first present a brief summary of MSC history and then outline the role of the MSC secretome in wound healing; provide background on recent MSC therapies for burns, diabetic wounds and EB; analyze the modes of cellular delivery; and discuss safety and optimization of MSCs for clinical use.
History and Definition of MSCs
In the 1960s and 1970s, Friedenstein et al. [1,2] discovered MSCs after finding that fibroblastic cells derived from mouse and guinea pig bone marrow were capable of producing clonal colonies of bone and reticular tissue following heterotopic transplantation. It was later discovered that these cells could also generate cartilage and adipose tissue [3]. In 1999, a subpopulation of stromal cells identified in human bone marrow was concluded to represent multi-potent stem cells with trilineage mesenchymal potential [4,5]. MSCs are generally characterized by their spindle shape and ability to adhere to tissue culture surfaces, but a specific MSC marker has yet to be defined, resulting in conflicting definitions of what constitutes an MSC. Consequently, the International Society for Cell & Gene Therapy MSC committee designated minimum criteria to define human MSCs as multi-potent mesenchymal stromal cells [6]. Importantly, the term “mesenchymal stem cells” is retained for the subpopulation of bone marrow cells that displays the two key properties of stem cells: self-renewal and multi-potency [6]. In 2019, the International Society for Cell & Gene Therapy MSC committee further clarified the nomenclature of MSCs and recommends that the acronym be supplemented with the tissue source origin of the cells to bring attention to tissue-specific properties [7].
Key properties of MSCs, such as their low levels of class 1 and class 2 HLA antigen expression, have made them an attractive clinical tool, allowing their use in transplantation without HLA matching. To date, the immunomodulatory properties of MSCs have been most prominently demonstrated as a therapy for graft-versus-host disease (GVHD). GVHD is a common complication after hematopoietic stem cell transplant in which the newly engrafted immune system (mainly CD4 and CD8 cells) attacks the host. A comprehensive review by Zhao et al. [8] outlines the clinical trials that have used MSCs for treatment of or prophylaxis against GVHD. In the context of GVHD, MSCs increase the prevalence of regulatory T cells that control GVHD by decreasing the number and pathological effects of CD4 and CD8 T cells, although there may be additional local immunomodulatory mechanisms at play [9].
MSCs have been successfully isolated from a wide range of human tissues, including bone marrow, adipose tissue and skin [4,10,11]. MSC isolation involves collection and digestion of tissue, expansion in culture and removal of non-adherent cells [12]. MSCs isolated from different tissue sources display heterogeneity in their ability to differentiate and proliferate, suggesting variable therapeutic relevance [13]. Such source-based variations in differentiation potential may be explained by different transcriptome, proteome, immunophenotype and immunomodulatory activities [14]. For example, proteomic analysis has shown that the MSC secretome is influenced by donor age and cellular niche, suggesting the therapeutic potential of MSCs could depend on tissue of origin [15]. Further research is needed to thoroughly assess the impact of cellular source and niche on MSC functions for clinical applications.
MSC Sources for Wound Healing
Although MSCs have been isolated from a variety of tissues, the most advantageous MSC source for wound healing has yet to be determined. The vast majority of evidence indicating that MSCs promote wound healing has been obtained using bone marrow-derived MSCs (BMSCs), mainly from data obtained in pre-clinical studies, with limited validation in clinical studies [16-19]. However, the use of BMSCs in a clinical capacity is not without its limitations. BMSCs must be obtained through aspiration, a generally safe but invasive procedure [20]. Furthermore, bone marrow cellularity declines with age, raising concern for the long-term differentiation potential of in vitro BMSCs [21,22]. Continued research into additional MSC sources that are more accessible than BMSCs and equally as efficacious is warranted.
Other examples of MSC sources that have been studied in the context of wound healing include adipose tissue, skin, umbilical cord and placenta [23-27]. Of these, adipose-derived MSCs (ADSCs) and dermal MSCs are both very accessible and do not have ethical controversies concerning their use. ADSCs have been shown to promote wound healing both in vitro and in vivo in the context of clinical trials for treatment of burns [28-30]. In terms of biological properties, BMSCs, ADSCs and dermal MSCs have similar immunogenicity and differentiation potentials [11,31-33]. However, these cells display differing paracrine expression profiles that can affect wound healing capacities [34]. The therapeutic potential of human BMSCs, ADSCs and amnion-derived MSCs in a murine model of cutaneous wounds was directly compared, and it was found that ADSCs had the most pronounced effect on wound closure [35]. Dermal MSCs have also demonstrated promising effects on wound healing in pre-clinical in vitro and animal studies and in several clinical trials [33,36,37].
In conclusion, the source of MSCs most likely has an impact on the ability of the cells to home to wounded tissue, engraft and exert anti-inflammatory wound healing effects [38-41]. These distinct differences, however, have not been well studied, and further pre-clinical and clinical studies are needed to determine the optimal MSC tissue source for wound healing.
Potential Role of MSCs in Cutaneous Wound Healing
MSCs have the capacity for self-renewal; can migrate to sites of injury; and provide key factors, such as cytokines and growth factors, via paracrine secretion to hasten healing [42]. Several studies have explored the immunomodulatory properties and proposed mechanisms of MSCs [43-47]. These attributes, including immunomodulation of the local microenvironment and ability to attract reparative cells and home to wounded tissue, have generated interest in the use of MSCs in cutaneous wound healing in the past decade because of lack of curative treatments for chronic wounds [42]. MSC therapies for skin wounds are based on repairing and replacing cellular substrates, attenuating inflammation, increasing angiogenesis and enhancing migration of reparative cells, although the mechanisms of distinct differentiation, mobilization and homing of MSCs are highly complex and require further study [42,48,49]. The following sections will focus on the roles of MSCs and their paracrine secretion, known as the secretome, in each stage of cutaneous wound healing. Although it has been shown that MSCs can home to wounded skin, the homing mechanism and induction of wound healing by MSC-secreted factors are complex and not fully understood. Therefore, the authors will focus on the known mechanisms of MSCs, with the goal of supplementing these cells exogenously to hasten and improve wound healing.
The skin is the body’s first line of defense against pathogens and the external environment. Epidermal keratinocytes maintain skin homeostasis by sensing tissue damage and foreign pathogens via innate signaling receptors, which enable the interface of a dynamic mechanical environment with an expansive skin-resident immunological network. Upon injury, such as in wounds, burns and dermatological diseases like EB, keratinocytes, dermal fibroblasts and immune cells coordinate wound healing in three stages: (i) inflammation, (ii) proliferation and (iii) remodeling (Figure 1).
Fig. 1.
MSC recruitment to wounded skin and the inflammatory phase, and known and potential roles of MSCs in each phase of wound healing. (A) Skin injury and hemostasis. (B) Inflammation. (C) Proliferation. (D) Remodeling. ADM, adrenomedullin; KGF, keratinocyte growth factor; MMPs, matrix metalloproteinases; TIMPs, tissue inhibitors of metalloproteinases.
When skin is injured, MSCs can infiltrate the wound in response to local inflammatory mediators such as TNF-α and IL-1β, secreted by neutrophils, which are the first to infiltrate the skin wound [48,50]. It may be advantageous to exogenously supplement these cells when they are dysfunctional or missing. When endogenous or exogenous MSCs reach the wound, they can secrete pro-inflammatory cytokines such as CSF2, IL-6, IL-8, CCL2 and CCL3, recruiting neutrophils and pro-inflammatory macrophages (Figure 1A,B) [51]. The neutrophils and macrophages then activate the adaptive immune system, distinguish commensal skin microbiota from foreign pathogens and begin to degrade damaged tissue. MSCs can also recruit plasmacytoid dendritic cells and limit the number of activated T cells and other innate immune cells to mediate the transition from the inflammatory phase to the proliferative phase [48].
MSCs in the proliferative phase of wound healing
In the proliferative phase of wound healing, MSCs secrete proteins to promote re-epithelialization (epidermal growth factor [EGF], keratinocyte growth factor and hepatocyte growth factor [HGF]) and angiogenesis (vascular endothelial growth factor [VEGF] A, ANGPT1 and platelet-derived growth factor B [PDGF-B]) (Figure 1C) [52]. They also prime fibroblasts to produce matrix proteins via fibroblast growth factor (FGF) [53].
MSCs can skew tissue-resident macrophages from an inflammatory phenotype (M1) toward a wound healing (M2) phenotype [54]. M2 macrophages are characterized by expression of IL-1RA, IL-4, IL-13, IL-10 and TGF-β. These macrophages promote a pro-reparative environment and help clear cellular debris. Local injection of MSCs has been shown to hasten wound healing and decrease infiltration of inflammatory M1 macrophages in pre-clinical models [54].
MSCs in the remodeling phase
In the final stage of wound healing, MSCs contribute to matrix remodeling through secretion of matrix metalloproteinases to induce matrix deposition and tissue inhibitors of metalloproteinases to abrogate deposition of extracellular matrix (ECM) proteins, which is vital for wounds to heal without scarring (Figure 1D) [45]. MSCs have also been thought to prevent hypertrophic scarring through secretion of HGF, FGF, adrenomedullin and TGF-β3 [55].
MSC secretome in wound healing
The secretome is defined as the set of molecules a cell secretes into the extracellular space. The MSC secretome has been shown to be involved in many different processes, such as paracrine signaling, stem cell differentiation, chemoattraction, inhibition of fibrosis and apoptosis, secretion of pro-angiogenic factors and immunomodulation [15,56-58]. This is an area of active research aimed at improving clinical outcomes in patients with chronic wounds and dermatological conditions that result in delayed or aberrant wound healing.
As mentioned previously, the MSC secretome most likely varies based on the tissue of origin but contains a core group of cytokines, including CCL2, CCL5, EGF, FGF, IL-6, IL-8, IL-10, TGF-β and VEGF-A [56]. The MSC secretome in human skin during wound healing lacks characterization, but recent work has provided some important insights. Components of the MSC secretome that have been shown to play a role in wound healing are described in Table 1.
Table 1.
Known components of the MSC secretome and their roles in wound healing.
| Wound healing phase affected |
|||||
|---|---|---|---|---|---|
| Component of MSC secretome | Inflammatory | Proliferation | Remodeling | Chronic wounds | Known or potential role in wound healing |
| CCL2, also known as MCP-1 and CCL3 | x | Chemokines, monocyte chemoattractants. Recruits and promotes essential macrophage functions in initial stages of skin wounding [59]. | |||
| CCL5 | x | Released by neutrophils and macrophages to recruit inflammatory cells [60]. Recruits bone marrow-derived epithelial progenitor cells. | |||
| GM-CSF, also known as CSF2 | x | x | Increases inflammatory cytokine IL-1 and MCP-1 to improve wound healing in diabetic ulcers via increased neovascularization and macrophage/neutrophil infiltration [61]. | ||
| EGF | x | Stimulates epidermal and dermal regeneration [48]. | |||
| FGF-1, FGF-2, FGF-5 and FGF-7 | x | Stimulate proliferation of various cells of mesodermal, ectodermal and endodermal origin [62]. Can stimulate angiogenesis. FGF-7, also known as KGF, is specific for epithelial cells. | |||
| Inflammatory cytokines IL-6 and IL-1 | x | x | Mobilize immune cells to wound. Keratinocyte, fibroblast proliferation and migration [58]. Can cause damage at high concentrations. | ||
| IL-8 | x | x | Cell migration and chemotaxis of neutrophils to wound. Potential role in keratinocyte migration in skin wounds [63]. | ||
| TGF-β1 and TGF-β3 | x | Promote ECM synthesis and wound contraction; anti-inflammatory in some contexts [58]. | |||
| VEGF | x | x | Collagen deposition, angiogenesis and epithelialization. Potential mitogenic, chemotactic and permeability benefits in non-healing wounds [64]. | ||
| ANGPT1 | x | x | x | Enhanced angiogenesis [65], lymphogenesis and blood flow in treated diabetic mice [66]. | |
| PDGF-BB | x | x | Stimulates chemotaxis of fibroblasts and proliferation and influences gene expression in macrophages and fibroblasts for tissue repair [67, 68]. | ||
| HGF | x | Secreted by mesenchymal cells and acts as a multi-functional cytokine on cells of mainly epithelial origin; plays a role in angiogenesis, tumorigenesis and tissue regeneration [69]. Potentially prevents fibrosis [70]. | |||
| MMPs and TIMPs | x | Promote ECM degradation (MMPs) and deposition (TIMPs) during wound healing, present in acute and chronic wounds, correct regulation necessary for wound resolution [71]. | |||
| ADM | x | Accelerates wound healing by promoting angiogenesis, collagen deposition and remodeling. Treatment shortened wound closure in rat model [72]. | |||
ADM, adrenomedullin; GM-CSF, granulocyte-macrophage colony-stimulating factor; KGF, keratinocyte growth factor; MMPs, matrix metalloproteinases; TIMPs, tissue inhibitors of metalloproteinases.
Another avenue of exploration in MSC-based therapies for wound healing is the use of MSC-derived exosomes. Exosomes are small extracellular vesicles (30–100 nm in size) derived from endosomal compartment cells, which contain biomolecules such as proteins, messenger RNA and microRNA. Exosomes are of specific interest in transplantation and wound healing treatments because they contain bioactive molecules similar to those found in MSCs but are “cell-free” and therefore not limited by the potential host rejection seen with “whole-cell” treatments [73-77]. Exosomes can be harvested from conditioned MSC medium under standard cell culture conditions. Hu et al. [78] recently published a comprehensive review of MSC exosomes for wound healing applications.
To summarize, MSC exosomes contain many components found in the MSC secretome and are modulated by the specific microenvironment. A recent study found that MSC exosomes derived from bone marrow, adipose tissue and umbilical cord contained wound healing-mediated growth factors (VEGF-A, FGF-2, HGF and PDGF-B), whereas TGF-β was found only in exosomes from umbilical cord MSCs [62]. This result stresses the importance of characterizing exosomes by MSC source before clinical application to ensure the presence and quality of exosomes can be replicated with the end application in mind (burns, diabetic ulcers or genetic diseases) [79].
In conclusion, exosomes provide a promising way to harness some of the potential of MSCs without the need for whole-cell treatments. The drawbacks to exosomes are their heterogeneity based on culture conditions and lack of complete mechanism of action of whole MSCs, including paracrine secretion of growth factors and the ability to persist and differentiate at the site of transplantation.
Wound Etiopathogenesis and Clinical Applications of MSCs
MSCs have gained traction in clinical applications because of their role in tissue regeneration and inflammation modulation. In the dermatological field specifically, MSCs have been used to improve resolution of diabetic wounds [80]; burns [81]; GVHD [82-86]; and inflammatory skin diseases [87-89] such as atopic dermatitis [90], psoriasis [91], lupus erythematosus [92] and genodermatosis, including EB [93,94]. Importantly, these cutaneous afflictions showcase a broad variety of wound etiologies, which vary in the depth of the wound and the host’s ability to heal. They can be broadly broken up into two categories, extrinsic (burns, pressure, injury) and intrinsic (nutrition status, diabetes, genetic disease), which will be discussed in this section [95]. When designing MSC therapies, the wound etiopathology should be carefully considered to dictate the best strategy for repair.
A concise review by Golchin et al. [89] outlines many MSC clinical trials for skin diseases. Despite a plethora of studies currently underway, many have not shown tangible improvements for patients. This furthers the need for pre-clinical studies to focus on individual mechanisms of immunomodulation and interaction with other cell types in the skin. A similar MSC review by Lee et al. reported that although multiple clinical studies have shown MSCs to enhance wound healing by decreasing time to wound closure, the mechanism of this benefit in vivo is unclear, with potentially multiple factors at play [97]. It is hypothesized that MSCs travel to wounded sites, differentiate into epithelial progenitors and secrete paracrine factors depending on their microenvironment [96]. To date, in vitro studies have found that MSCs can enhance migration of fibroblasts and keratinocytes either with direct co-culture or with the use of MSC-conditioned media [97-100]. The authors will now discuss previous clinical and pre-clinical studies of MSC treatments on specific wound etiologies with specific attention to RDEB and how targeted MSC therapy could best be used or adjusted to fit the needs of each type of wound.
Genetic Skin Wounds: EB
Genetic skin diseases such as the heterogenous EB group are characterized by skin fragility and caused by mutations in one of 19 different skin proteins [101,102]. These patients have missing or dysfunctional skin structural proteins in the epidermis (EB simplex) or BMZ between the epidermis and dermis, as seen in junctional EB and RDEB. Wounds caused by these genetic diseases have difficulty healing correctly because of the disrupted architecture and loss of skin structural proteins necessary for skin repair. RDEB, which is caused by mutations in the ECM protein C7, is the most severe of this group, with even slight touch causing blistering of the skin. Many patients have sites of recurrent skin separation that turn into chronic wounds.
Alterations in wound repair in RDEB include increased and persistent inflammation; reduced proliferation and migration of dermal fibroblasts and epidermal keratinocytes; and, potentially, pathological remodeling, resulting in wounds healing by scarring [102]. Specifically, chronic wounds in RDEB allow for bacterial colonization and infection, in turn aggravating the immune response and further delaying wound healing. IL-4R; IL-13RA1; CXCL13; MCP-1; regulated on activation, normal T-cell expressed and secreted; and IFN-α are all overexpressed in RDEB skin, revealing a dysfunctional immune response [102]. Additionally, a pre-clinical study in Col7a1-deficient mice found that loss of C7 alters keratinocyte migration by disrupting laminin-332 deposition and impairs fibroblast migration and granulation tissue formation after cutaneous insult [103]. These studies exemplify a profound perturbation of normal wound healing in RDEB and the need for targeted therapies to address the specific needs of this patient population. As MSCs have the capability of producing growth factors needed for proper wound healing, improving angiogenesis and modulating inflammation, they could be a viable treatment option for genodermatological diseases such as RDEB, especially if non-autologous MSCs are used, and could provide the missing or dysfunctional protein to the skin.
Pre-Clinical and Clinical Use of MSCs in RDEB
RDEB is a systemic disorder. In addition to the readily observable skin wounds in RDEB patients, there are other sites of basement membrane disruption. Blistering in the mouth or fusion of the tongue to the floor of the mouth can make eating almost impossible. Esophageal erosions can lead to webs and strictures that can cause severe dysphagia. Consequently, malnutrition and vitamin and mineral deficiencies may lead to growth restriction in young RDEB patients. Finally, it is thought that repeated cycles of inflammation and fibrosis lead to a lifetime risk of aggressive squamous cell carcinoma that is higher than 90%, with a 5-year survival rate near zero [104,105].
There is currently no cure for RDEB, but strides to improve treatment have been made with bone marrow transplant and use of MSCs to dampen aberrant inflammatory responses and improve wound healing [106-111]. A drawback to these therapies is that the beneficial effects have shown to be only transient. Repeated injections throughout a patient’s life would likely be necessary for MSC-based treatments. Additionally, the mechanism of improved patient phenotype and decreased blistering is unclear. It is hypothesized that MSCs migrate to wounded tissue and then exert anti-inflammatory effects, providing benefit via the MSC secretome. Therefore, scientific focus should be on and research efforts should be geared toward pre-clinical studies to dissect out the concrete mechanisms that are at play when MSCs are delivered systemically or locally. Pre-clinical studies are limited and have focused on in vitro MSC interactions with skin cells (keratinocytes and fibroblasts) and using MSCs in wound healing models of humanized mice or mice without competent immune systems.
An in vitro study using BMSC exosomes on RDEB fibroblasts showed that exosomes can influence RDEB fibroblasts by transporting C7 to the extracellular space directly and providing fibroblasts with messenger RNA coding for C7, allowing for the fibroblasts to translate and synthesize C7 themselves [112]. Another group generated MSC-like cells from induced pluripotent stem cells derived from keratinocytes [113]. The researchers detected C7 at the dermal–epidermal junction after subcutaneous and intravenous injection of these cells into mice.
An in vivo murine study demonstrated that MSCs can be engineered to overproduce C7 [114]. These MSCs were incorporated into human bioengineered skin (using RDEB keratinocytes and fibroblasts) and grafted onto a NOD-scid IL2Rgammanu11 mouse. Engineered MSCs were locally retained, deposited C7 at the dermal–epidermal junction and restored anchoring fibril density. An additional study reported similar results in a tamoxifen-inducible Col7a1 knock-out mouse, showing that local injection of congenic MSCs restored the anchoring fibrils at the dermal–epidermal junction and partially reversed the RDEB phenotype [115]. Pre-conditioning of mice with cytokines TGF-β or TNF-α for 24–72 h before harvesting BMSCs showed increased Col7a1 expression, providing additional support for MSC pre-conditioning as a mechanism to increase C7 production in MSCs before clinical application [116]. Another in vivo study found bone marrow transplant to restore C7 in the cutaneous BMZ, specifically with PDGFRa+ bone marrow-derived cells migrating to skin grafts from Col7a1null mice that had been grafted on to green fluorescent protein (GFP) bone marrow-transplanted C57BL6/N wild-type mice [117].
A small percentage (1–2%) of dermal cells in the skin express the marker ABCB5 and have been shown to display protein expression and anti-inflammatory benefits similar to those of BMSCs [54,118]. The use of dermal-derived ABCB5+ MSCs provided systemically to neonatal Col7a1−/−RDEB mice significantly prolonged survival compared with phosphate-buffered saline-injected control animals, with ABCB5+ dermal MSC-treated mice showing decreased M1 macrophage skin infiltration [119]. Additionally, recent work in a wounded mouse model has shown that ABCB5+ MSCs exhibit superior homing to skin wounds compared with BMSCs after peripheral injection [5]. RNA sequencing of ABCB5+ MSCs compared with BMSCs showed expression of major histocompatibility complex class II and homeobox (Hox) genes, specifically HOXA3, in ABCB5+ MSCs. Critical to inducing migration of endothelial and epithelial cells for wound repair, HOXA3 is a promising gene candidate for investigating mechanisms of wound homing and local wound healing [5]. These cells are being evaluated in early-phase clinical trials, including local therapy for chronic venous ulcers and systemic administration for treatment of RDEB (NCT03529877, EudraCT 2018-001009-98). Results from the first human trial in which 12 wounds from nine patients with chronic venous ulcers were treated with local administration of ABCB5+ dermal MSCs showed safety, tolerability, median wound size reduction of 63% and perceived decrease in pain [120].
MSCs have shown promise in clinical trials for improving wound healing and quality-of-life outcomes in dermatological diseases such as RDEB, although the mechanism of action of MSCs in the disease process remains unclear. It is possible that BMSCs are recruited to wounded skin by the release of HMGB1 by apoptotic keratinocytes. This has been shown in a pre-clinical murine model of skin grafting, and HMGB1 is the only known biomarker of RDEB, with serum levels correlating with severity of disease [67,121]. A comprehensive list of MSC-based therapies injected either locally or systemically and their outcomes in RDEB is shown in Table 2, excluding case reports.
Table 2.
Clinical RDEB studies and their outcomes using MSCs as therapy.
| Authors | Study type and ID number | RDEB patients, n | Intervention | Findings |
|---|---|---|---|---|
| El-Darouti et al. [122] | Double-blind | 7 BMSCs + cyclosporine, 7 BMSCs no cyclosporine | One single injection of BMSCs ± cyclosporine | • Number of new blisters decreased significantly in both groups (P = 0.003 and 0.004). • Rate of healing of new blisters significantly faster in both groups (P < 0.001). |
| Petrof et al. [123] | Unblinded, prospective phase 1/2 study; EudraCT 2012-001394-87, ISRCTN46615946 | 10 | Three intravenous infusions of non-HLA-matched BMSCs (day 0, day 7 and day 28) | • No increase in C7 and no new anchoring fibrils at day 60. • Patients reported decrease in disease severity. • Improved wound healing (increased suction blister time and decreased blister counts) for up to 6 months. |
| Rashidghamat et al. [121] | Phase 1/2 clinical trial; EudraCT 2014-004500-30 | 10 | Two intravenous infusions of BMSCs (day 0 and day 14) | • Reduction in disease activity scores (eight of 10 subjects). • Significant reduction in pruritus. • Decrease in biomarker HMGB1 at day 28 and day 60. |
| Conget et al. [124] | Prospective study of two patients, unblinded | 2 | Intradermal allogeneic BMSC injection | • C7 protein detected along BMZ day 7 after injection. C7 present for 4 months after injection of BMSCs. |
ID, identifier. BMZ, basement membrane zone
In conclusion, although in vivo studies have shown promise for restoring C7 in the BMZ, clinical studies have shown variable and transient improvement in patients with RDEB. Additional investigation of the specific immunomodulatory properties of MSCs and their impact on the skin microenvironment demands further study in vitro.
Burns
Burns are the fourth leading cause of injury in the world and can leave permanent functional damage [125]. Treatment of deep (second-degree) or full-thickness (third-degree) burns relies on excision of necrotic tissue followed by replenishment of healthy tissue, often with the use of autologous skin grafts. Burns differ from other types of skin wounds, in that they can progress and deepen with time and, similar to RDEB, can heal with scarring. A recent review by Rangatchew et al. [126] highlights the use of MSCs for burn therapy, summarizing three human and a multitude of murine and porcine studies. The three human studies included two single case reports and a larger study with 60 patients randomized to one of three groups (n = 20 per group): (i) excision of burn with autologous skin graft, (ii) excision with BMSC injections 2 days and 10 days post-burn and (iii) excision and injection or topical spraying of umbilical cord MSCs 2 days and 10 days post-burn [127-129]. Results from the human studies found increased vascularization of granulation tissue, as indicated by visual assessment and decreased percentage of burn area after MSC treatment. Importantly, aside from infection at the burn site in 25% of patients (not likely attributed to MSC application), no additional adverse events were reported [126]. Although these studies are initially promising, further studies with more rigorous, objective measures of wound healing, such as total body surface area of wound, skin biopsies to show granulation tissue formation, analysis of inflammatory cytokines and infections in wound beds or time to complete wound closure, are needed.
Animal studies using MSCs to treat burns are more numerous, showing that local treatment improves body weight and activity level of mice while significantly reducing local and systemic inflammation, apoptosis and burn wound progression. The limitations of these studies include great heterogeneity in animals and burn models as well as the type of MSCs used. Because of the progressive nature of burns, treatment with MSCs should focus on prompt delivery to combat progression of the wound and induce the formation of vascularized granulation tissue to hasten wound healing.
Diabetic Wounds
In diabetes, skin ulcerations are common on the lower extremities, affecting 15% of patients, 14–24% of whom require amputation as a result of inadequate wound healing and infection [130]. Diabetic skin ulcers result from poor vascular perfusion and are characterized by disintegration of the dermis and epidermis. Diabetic wounds are similar to RDEB, in that they are remarkably resistant to healing and frequently become colonized with pathogens. Additionally, treatment for diabetic wounds focuses on non-adherent dressings, wound gel and prevention of additional trauma, with minimal targeted therapies to address the disrupted microenvironment [95]. The specific impact of MSCs on the improvement of angiogenesis could be valuable in the treatment of diabetic wounds, as outlined in the review by Maranda et al. [81]. In chronic diabetic wounds, angiogenic factors such as VEGF-A, IL-6, FGF and TGF-β1 are decreased, impeding wound healing [131]. Additionally, decreased vascular flow can cause a hypoxic wound bed. Interestingly, MSCs cultured in hypoxic conditions demonstrate increased production of anti-inflammatory cytokines and growth factors such as TGF-β and VEGF-A and expression of ECM molecules (type III collagen, fibronectin and elastin) and improve the viability and migration of dermal fibroblasts, potentially increasing their therapeutic efficacy in this type of wound [132-135].
Pre-clinical studies in rats have found that BMSCs promote the production of MMP-2, EGF and IGF-1 by human keratinocytes and enhance the migration and proliferation of keratinocytes by downregulating TIMP1 and TIMP2 and initiating Erk signaling [136]. A review of the trials using MSCs as therapy in diabetic wounds is outlined by Cao et al. [137]. These studies ranged from 15 to 96 patients, and the outcomes showed positive results, including increased pain-free walking distance, improvement in leg perfusion and ankle-brachial index and increase in the vascularity of the dermis. Interestingly, a study of 96 patients with diabetic foot ulcers in whom autologous BMSCs were injected into the tibial artery showed 79% limb salvage [138]. Overall, these results are promising and allow for objective, quantitative measurements of efficacy; however, the mechanism of MSC migration and local contribution to wound healing is still uncertain.
Chronic Wounds
If resolution of the inflammatory phase of wound healing does not occur because of infection, disrupted skin architecture or poor circulation, a chronic wound can ensue. This is common in both RDEB and diabetic ulcers [102]. This state of chronic inflammation can cause increased levels of metalloproteinases that disrupt normal remodeling of the skin after injury by destroying components of the ECM and abrogating the secretion of growth factors needed for resolution of wound healing [48]. The traditional treatment for chronic wounds relies on debridement (removing foreign debris and contaminated/necrotic tissues), compression, pressure relief, hyperbaric oxygen, antimicrobials and moist dressings to hasten wound healing. None of these treatments target the aberrant inflammatory environment of a chronic wound [139]. MSCs have been found to not only support wound healing, as described earlier, but also combat harmful infections. Direct antimicrobial activity was demonstrated by BMSCs against Escherichia coliand Staphylococcus aureus, both common wound pathogens, in in vitro and in vivomodels [140]. A more recent study found that ADSCs effectively decreased the growth of E coli in vitro, demonstrating significant antimicrobial activity [141]. Additionally, ADSCs along with umbilical cord-derived MSCs have been shown to inhibit the growth of Pseudomonas aeruginosa, another type of bacteria common to skin wounds [142,143]. The antimicrobial effects observed by MSCs further justify their therapeutic use in wound healing.
In conclusion, wounds vary widely depending on the origin of insult. This should be noted when developing therapeutic strategies to target the disruption by focusing on attenuating inflammation, increasing angiogenesis, genetically modifying cells to increase expression of missing/dysfunctional proteins (genetic diseases), pre-conditioning them to increase expression of immunomodulatory factors or using specific culture conditions to achieve desired MSC efficacy. The downfalls of using MSCs in clinical trials may result from poor engraftment of MSCs after delivery. Therefore, the mode of delivery must be carefully considered and will be discussed in the following section.
Mechanisms of MSC Delivery to Skin
Topical delivery
Topical delivery can include both intradermal injection and cutaneous application. Local injection of MSCs to the wound site allows for a greater number of cells to be introduced to the area, but results in past studies have been transient, lasting up to 4 months [124]. BMSCs have also been delivered to human diabetic wounds in a fibrin spray [17]. This study found the application of the cells was safe, but a large number of cells were needed (>1 × 106 per cm2 of wound area) to decrease wound size (P = 0.0058). The same delivery method using GFP+ BMSCs was tested in a diabetic mouse model, and GFP+ cells were found near blood vessels, suggesting that these cells may persist, although the time point of analysis was 5 days after treatment.
Systemic delivery
Systemic injection of MSCs has the advantage that the cells can travel to multiple sites of disruption, although the mechanism involved and the ability of MSCs to home to wounded skin are still being studied. MSCs express several chemokine receptors, principally CCR7, which is a receptor of CCL21. Previous work has shown that this plays a role in MSC migration, and CCL21 expression is increased in irritated skin [144]. MSC migration to the skin was increased after intradermal injection of SLC/CCL21 and also resulted in accelerated wound repair [59]. Drawbacks to systemic injection of MSCs include infusion reactions such as fever and death of cells in the small vasculature, specifically of the lungs, without ever reaching a cutaneous destination. This is termed the “pulmonary first-pass effect,” which is a major barrier in the ability of MSCs to home to cutaneous wounded tissue. Animal studies in rats have estimated that approximately 4% of systemically injected MSCs are likely to survive passage through the pulmonary vasculature, although studies in humans are lacking [145,146].
Scaffolds and Three-Dimensional Bioprinting
Incorporating biomaterials into MSC delivery has been shown to be advantageous in wound healing. Seeding of MSCs on a collagen sponge prior to application to human skin wounds demonstrated therapeutic efficacy, as determined by healing of the wounds and increased epithelialization of surrounding tissue [18]. When applied to wounds of nude mice, the collagen sponges containing MSCs facilitated marked tissue regeneration after 2 weeks, whereas this was not detected when MSCs were injected without the collagen scaffold. This use of biomaterials to aid in cell delivery is an example of the “biological wound dressing concept” [147]. Though only a limited number of biological wound dressings have demonstrated significantly superior healing outcomes, the delivery of MSCs within a scaffold composed of biomaterials is a promising avenue for MSC therapies.
MSCs can be seeded within a scaffold of biomaterials, achieving both physical and biomechanical integrity. Yannas et al. [148] determined specific structural features, including highly cross-linked collagen glycosaminoglycan matrices, to be the foundation of a “template” for skin regeneration following injury. This acellular scaffold was shown to delay the onset of wound contraction by about 10 days in an in vivo experiment on adult guinea pigs [149]. Inclusion of cells such as MSCs in a bioengineered scaffold is hypothesized to be superior to the use of either component independently. In an in vivo rat wound model, human MSCs were seeded on a medical-grade polycaprolactone dressing. Compared with control groups, this combined MSC and medical-grade polycaprolactone dressing demonstrated significantly improved skin regeneration and decreased wound contraction [150]. Theoretically, bioactive materials secreted from MSCs could be delivered to the wound site through the porous structure of the scaffold to promote wound healing. Additionally, MSC scaffolds may contain multiple cell types and ECM components that produce skin-like structures and promote skin regeneration. MSCs have been shown to differentiate into an epidermal lineage when co-culturing MSCs and fibroblasts on a collagen gel and adding EGF and vitamin D3 [151].
Although scaffolds allow for the delivery of MSCs as well as possibly advantageous biomaterials and cell types to accelerate wound healing, they are laborious to produce and lack precise cell placement. Three-dimensional (3D) bioprinting, or printing of biomaterials into a specific shape, can be theoretically fully standardized and automated, resulting in control over cell and biomaterial organization. The route of 3D delivery of MSCs is thought to more closely replicate the natural in vivo environment of MSCs and thus foster a natural cellular response. Culturing MSCs within a pelleted cell spheroid, for instance, has been shown to improve MSC differentiation into both the typical lineages and hepatocytes [152]. MSCs have been bioprinted in structures replicating tissues such as cartilage and tumors [153,154]. Furthermore, with their ability to differentiate into endothelial-like cells, MSCs are being investigated for their use in vasculature bioprinting [155]. Replicating complex structures such as blood vessels is important when considering bioprinting for skin regeneration. Although 3D bioprinting has potential as a cell and tissue delivery system, this technology is still very much in its infancy and requires incredible optimization. At the current stage, functional skin cannot be bioprinted; rather, cells can be bioprinted to mimic skin structure. Importantly, the biological process that follows the actual bioprinting is crucial for the development of skin-like structures and the overall success of this method.
A comparison of the advantages and disadvantages of each aforementioned MSC delivery method is shown in Table 3. The favored mechanism of MSC delivery to skin wounds requires additional investigation, with the safety and efficacy of systemic versus topical administration of MSCs as critical variables in MSC therapy for chronic wounds in patients with RDEB-associated or other chronic wounds.
Table 3.
A comparison of various methods of MSC delivery to skin wounds.
| Delivery method | Advantage | Disadvantage |
|---|---|---|
| Topical spray/local injection | Delivery directly to affected areas; avoids injection reactions. | Transient improvement in phenotype. |
| Systemic injection | Delivery to all affected areas, potential for long-term persistence. | Potential risk of infusion reactions, cell death in small vasculature; efficacy is dependent on homing ability of cells. |
| Scaffold | Addition of biomaterials and other cell types to promote healing, template for MSCs to expand and secrete wound healing biomaterials. | Laborious to produce and customize; lacks precision of cell placement. |
| 3D bioprinting | Standardized and automated process; allows for precise cell and biomaterial placement | Technology is in very early phases; bioprinted skin-like structures lack vasculature and complex multi-layered skin structure. |
Genetic Modifications of MSCs
Genetic modification of MSCs has been used to promote skin regeneration and increase MSC-mediated immunomodulatory effects [156]. Developing MSC-based therapies for genodermatological skin diseases like RDEB is a continuing challenge. Although MSCs demonstrate innate wound healing properties, genetic modification of these cells is an exciting technique for enhancing their therapeutic potential. MSCs can be genetically modified to overexpress or inhibit gene expression or modify certain pathways [157,158]. Additionally, modification of MSCs with CRISPR/Cas-based non-viral gene editing has been shown to increase the viability of transplanted MSCs by increasing HIF1a gene levels [159]. In RDEB, genetically modified MSCs have the potential to not only promote wound healing but also act as a vehicle for gene, protein or drug delivery. Over a decade ago, gene therapy was one of the most obvious avenues to investigate in the treatment of RDEB and other forms of EB. In a recent study, human umbilical cord-derived MSCs were transduced with a self-inactivating lentiviral vector encoding COL7A1 transgene to express C7 [114]. The engineered MSCs were delivered intradermally into human RDEB grafts pre-established on immunodeficient mice. These MSCs persisted, deposited C7 and restored anchoring fibril density. Alternative approaches using retroviral gene-corrected keratinocytes have been successful in individual cases of EB [160-162]. However, all gene therapy approaches for EB remain experimental [163]. The use of genetic modification to improve the natural properties of MSCs or target diseases like RDEB specifically greatly increases the possible utility of MSCs.
Approaches to Improving the Potential of MSCs for Wound Treatment
Because of their broad potential and impact on immune modulation, MSCs are one of the most studied cellular applications for the treatment of human disease. Clinical studies have fallen short of promising pre-clinical results, and several studies have examined this history [164,165]. In 2019, biomedical research as a whole was criticized for having a “replication crisis” in pre-clinical studies [166]. MSC research similarly demonstrates inconsistent outcomes. Poor reproducibility in the field may result from differences in MSC tissue source, culture conditions or MSC handling before their use in clinical applications.
Typically, human subjects enrolled in clinical trials receive one or multiple doses of allogeneic MSCs that have been previously cryobanked under Good Manufacturing Practice (GMP) conditions or following cell culture with or without pre-conditioning the cells with cytokine stimulation, serum deprivation, 3D culture conditions, hypoxic conditions or another form of cellular manipulation. For previously cryobanked MSCs that are thawed directly prior to clinical use, there have been observations of molecular signatures of cell injury, such as a decrease in suppressive functions and shortened persistence in vivo, which could be related to freezing-induced senescence and could account for the varied efficacy of MSCs used in clinical trials. This is outlined further in the review by Moll et al. [167].
There have been multiple attempts to improve the ability of MSCs to exert clinical benefit in patients. These have included augmenting MSC survival with ECM proteins or hypoxic conditioning [132]. For example, ECM proteins of interest include tenascin C and decorin, which have been shown to decrease inflammation, improve tissue repair during wound healing and abrogate skin scarring [132].
One of the most studied mechanisms by which inflammation triggers MSC activity involves IFN-γ. This cytokine is typically produced during Th1 immune responses, which are associated with autoimmunity mediated by cellular means, such as CD8 T cells and natural killer cells. Exposure of MSCs to INF-γ has been demonstrated by numerous groups to increase immune suppressive activity via stimulation of the enzyme indoleamine-2,3-dioxygenase [168]. MSC exposure to indoleamine-2,3-dioxygenase induces the production of other inhibitors of inflammation by MSCs, including the complement inhibitor factor H and the immunomodulatory molecules TGF-β and HGF. TNF-α pre-treatment of MSCs increases angiogenic activity in vitro, as assessed by expression of VEGF, and serum deprivation in MSC cultures enhances expression of VEGF, angiopoietins, IGF-1 and HGF, which are markers of angiogenesis [169].
Finally, another approach could be culturing MSCs in 3D scaffolds, which upregulates the production of anti-inflammatory molecules compared with monolayer culture [168]. Although there are many possible ways to enhance the in vitro suppressive and favorable wound healing activity of MSCs, a systematic study of these approaches needs to be tested in GMP-cultured MSCs with clinical applications in mind. Most importantly, criteria for clinical MSC preparations should be matched to the hypothesized mechanism of action and tested pre-clinically, and clinical handling of the MSCs prior to use should depend on the tissue source, type of disease and desired recipients [165].
Safety and Manufacturing of MSCs for Wound Healing Applications
The use of MSC-related therapies is not without risk, and the safety of these cells must be considered fully when developing clinical applications. Primary safety concerns regarding MSC administration include tumorigenicity, fibrosis and pro-inflammation. MSCs have the capacity to develop into tumors and have been shown to actually promote tumor development by releasing certain chemokines and growth factors [14]. Additionally, it has been reported that the immunosuppressive qualities of MSCs support tumorigenicity and metastasis of tumor cells [170,171]. In certain animal models, MSCs have been reported to home to tumors and promote tumor development [172,173]. To safely administer MSCs, the potential for malignant transformation must be ruled out with strict cell quality control measures. Furthermore, it has been found in ex vivo expansion that MSCs develop genomic instability during later passages [174,175]. This finding stresses the importance of determining appropriate cell passage numbers when generating clinical therapies. Although these findings are very concerning, at the time of this review, current in vivo clinical trials involving MSC therapies have not reported adverse cases of tumorigenicity [176].
MSC-based therapies also pose a risk of fibrosis [177]. Though MSCs do have immunosuppressive effects, this has been shown only following exposure to very high levels of pro-inflammatory cytokines. In the presence of low levels of TNF-α and IFN-γ, MSCs have actually been shown to have an inflammatory response [178]. The environment of MSCs is crucial to their effect on the immune system, and this must be considered when developing MSC therapies. Importantly, thrombogenic events have been reported in several clinical trials following MSC infusion [179-182]. In addition to mechanical vessel obstruction by MSCs (reported to be as large as 30 μm in diameter), in vitro and in vivo studies have shown that MSCs display pro-coagulant activity [183-185]. The use of anti-coagulant drugs like heparin or bivalirudin may counter the hypercoagulable milieu induced by MSC infusion [186]. Continued research into assessing and limiting coagulation following MSC therapies is vital.
Although MSCs are thought to be generally safe, caution and protective measures are vital and also warrant further investigation into the use of MSC-conditioned medium therapeutically as opposed to administering MSCs as a cellular product. As explained previously, MSCs promote wound healing mainly through paracrine action of the secretome. MSC-conditioned medium contains the secreted factors that are thought to cause the advantageous effects, such as re-epithelialization, angiogenesis and anti-fibrosis. Furthermore, Yagi et al. [141] demonstrated that MSC-conditioned medium alone was effective at significantly suppressing S aureus bacterial growth. Clinical therapies utilizing conditioned medium rather than MSCs may be an advantageous option and may possibly reduce the risk of tumorigenicity. Animal studies implementing this strategy to promote wound healing have verified it as a promising technique [64,70,187,188].
Finally, it is vital that MSCs for clinical application in wound healing be cultured and expanded in reproducible conditions for consistent characterization–such as retention of MSC markers or functional analyses–prior to administration. Therefore, much effort has focused on creating GMP standards to allow for clinical use of MSCs. Factors that need to be considered are standardization of culture conditions (incubator temperature, oxygen and carbon dioxide levels); cell seeding density; length of culture; and culture medium, specifically the use of xenogeneic additives such as fetal bovine serum (FBS) [189,190]. FBS use in the clinical context brings a variety of risks, such as prion and viral transmission or adverse immunological reactions to xenogeneic components as well as extreme variability of FBS batches [191]. A potential alternative is the use of human serum or human platelet lysate. Comparison of human serum with FBS at 10% v/v has shown higher proliferation capacity of MSCs in human serum. Mushahary et al. [192] discuss the impact of using human components on MSC differentiation and expansion in further detail in their review.
In conclusion, comprehensive studies directly comparing human components with FBS in MSC expansion and differentiation from various tissue sources are lacking. This leaves many questions unanswered regarding the best way to prepare MSCs for optimal clinical results. Therefore, further studies and research in this area are urgently needed.
Discussion
MSC-based therapies have shown promise in aiding wound healing. The advantageous effects of the MSC secretome make them a strong candidate for a novel wound therapy, although to create a treatment targeted to the specific wound microenvironment, the wound etiology must be taken into account. Furthermore, although MSCs have been shown to play a beneficial role in the inflammation, proliferation and remodeling phases of wound healing, the logistics of catering to each of the different wound healing phases must be contemplated. The immunomodulatory and antimicrobial effects of MSCs make them an especially promising therapy for genetic skin wounds, such as those seen in RDEB, burns and diabetic ulcers. Several critical challenges need to be addressed when developing MSC-based therapies, including choice of cell source; delivery method; in vitro pre-conditioning; and, depending on the application, overall safety profile. Continued research–specifically pre-clinical studies focusing on the mechanisms of immunomodulation of the surrounding microenvironment and ability of MSCs to engraft and persist in wounds–is recommended to further elucidate the current limitations and unknown mechanisms involved in MSC orchestration of wound healing, especially their differing effects depending on specific niche and environmental stimuli. Current data, including pre-clinical and clinical trials, however, do support the future realities of MSC-based therapies for wound healing, although the mode of therapy and pre-conditioning regimen will need to be standardized for each application.
Funding
No funding was received
Footnotes
Declaration of Competing Interest
The authors have no commercial, proprietary or financial interest in the products or companies described in this article.
References
- [1].Friedenstein AJ, Piatetzky IIS, Petrakova KV. Osteogenesis in transplants of bone marrow cells. J Embryol Exp Morphol 1966;16:381–90. [PubMed] [Google Scholar]
- [2].Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet 1970;3:393–403. [DOI] [PubMed] [Google Scholar]
- [3].Caplan AI. Mesenchymal stem cells. J Orthop Res 1991;9:641–50. [DOI] [PubMed] [Google Scholar]
- [4].Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284:143–7. [DOI] [PubMed] [Google Scholar]
- [5].Riedl J, Pickett-Leonard M, Eide C, Kluth MA, Ganss C, Frank NY, et al. ABCB5+ dermal mesenchymal stromal cells with favorable skin homing and local immunomodulation for recessive dystrophic epidermolysis bullosa treatment Stem Cells 2021;39:897–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement Cytotherapy 2006;8:315–7. [DOI] [PubMed] [Google Scholar]
- [7].Viswanathan S, Shi Y, Galipeau J, Krampera M, Leblanc K, Martin I, et al. Mesenchymal stem versus stromal cells: International Society for Cell & Gene Therapy (ISCT(R)) Mesenchymal Stromal Cell committee position statement on nomenclature. Cytotherapy 2019;21:1019–24. [DOI] [PubMed] [Google Scholar]
- [8].Zhao Q, Ren H, Han Z. Mesenchymal stem cells: immunomodulatory capability and clinical potential in immune diseases. J Cell Immunother 2016;2:3–20. [Google Scholar]
- [9].Zhao K, Lou R, Huang F, Peng Y, Jiang Z, Huang K, et al. Immunomodulation effects of mesenchymal stromal cells on acute graft-versus-host disease after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2015;21:97–104. [DOI] [PubMed] [Google Scholar]
- [10].Dicker A, Le Blanc K, Astrom G, van Harmelen V, Gotherstrom C, Blomqvist L, et al. Functional studies of mesenchymal stem cells derived from adult human adipose tissue. Exp Cell Res 2005;308:283–90. [DOI] [PubMed] [Google Scholar]
- [11].Richardson GD, Arnott EC, Whitehouse CJ, Lawrence CM, Hole N, Jahoda CA. Cultured cells from the adult human hair follicle dermis can be directed toward adipogenic and osteogenic differentiation. J Invest Dermatol 2005;124:1090–1. [DOI] [PubMed] [Google Scholar]
- [12].Soleimani M, Nadri S. A protocol for isolation and culture of mesenchymal stem cells from mouse bone marrow. Nat Protoc 2009;4:102–6. [DOI] [PubMed] [Google Scholar]
- [13].Andrzejewska A, Lukomska B, Janowski M. Concise Review: Mesenchymal Stem Cells: From Roots to Boost Stem Cells 2019;37:855–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Han Y, Li X, Zhang Y, Han Y, Chang F, Ding J. Mesenchymal Stem Cells for Regenerative Medicine. Cells 2019;8:886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Gaetani M, Chinnici CM, Carreca AP, Di Pasquale C, Amico G, Conaldi PG. Unbiased and quantitative proteomics reveals highly increased angiogenesis induction by the secretome of mesenchymal stromal cells isolated from fetal rather than adult skin. J Tissue Eng Regen Med 2018;12:e949–61. [DOI] [PubMed] [Google Scholar]
- [16].Badiavas EV, Falanga V. Treatment of chronic wounds with bone marrow-derived cells. Arch Dermatol 2003;139:510–6. [DOI] [PubMed] [Google Scholar]
- [17].Falanga V, Iwamoto S, Chartier M, Yufit T, Butmarc J, Kouttab N, et al. Autologous bone marrow-derived cultured mesenchymal stem cells delivered in a fibrin spray accelerate healing in murine and human cutaneous wounds. Tissue Eng 2007;13:1299–312. [DOI] [PubMed] [Google Scholar]
- [18].Yoshikawa T, Mitsuno H, Nonaka I, Sen Y, Kawanishi K, Inada Y, et al. Wound therapy by marrow mesenchymal cell transplantation. Plast Reconstr Surg 2008;121:860–77. [DOI] [PubMed] [Google Scholar]
- [19].Hu MS, Borrelli MR, Lorenz HP, Longaker MT, Wan DC. Mesenchymal Stromal Cells and Cutaneous Wound Healing: A Comprehensive Review of the Background, Role, and Therapeutic Potential. Stem Cells Int 2018;2018:6901983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Kita K, Gauglitz GG, Phan TT, Herndon DN, Jeschke MG. Isolation and characterization of mesenchymal stem cells from the sub-amniotic human umbilical cord lining membrane. Stem Cells Dev 2010;19:491–502. [DOI] [PubMed] [Google Scholar]
- [21].Rao MS, Mattson MP. Stem cells and aging: expanding the possibilities. Mech Ageing Dev 2001;122:713–34. [DOI] [PubMed] [Google Scholar]
- [22].Barry FP, Murphy JM. Mesenchymal stem cells: clinical applications and biological characterization. Int J Biochem Cell Biol 2004;36:568–84. [DOI] [PubMed] [Google Scholar]
- [23].Natesan S, Wrice NL, Baer DG, Christy RJ. Debrided skin as a source of autologous stem cells for wound repair. Stem Cells 2011;29:1219–30. [DOI] [PubMed] [Google Scholar]
- [24].Roubelakis MG, Trohatou O, Anagnou NP. Amniotic fluid and amniotic membrane stem cells: marker discovery. Stem Cells Int 2012;2012:107836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Kong P, Xie X, Li F, Liu Y, Lu Y. Placenta mesenchymal stem cell accelerates wound healing by enhancing angiogenesis in diabetic Goto-Kakizaki (GK) rats. Biochem Biophys Res Commun 2013;438:410–9. [DOI] [PubMed] [Google Scholar]
- [26].Arno AI, Amini-Nik S, Blit PH, Al-Shehab M, Belo C, Herer E, et al. Human Wharton’s jelly mesenchymal stem cells promote skin wound healing through paracrine signaling. Stem Cell Res Ther 2014;5:28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Bottcher-Haberzeth S, Biedermann T, Klar AS, Pontiggia L, Rac J, Nadal D, et al. Tissue engineering of skin: human tonsil-derived mesenchymal cells can function as dermal fibroblasts. Pediatr Surg Int 2014;30:213–22. [DOI] [PubMed] [Google Scholar]
- [28].Nie C, Yang D, Xu J, Si Z, Jin X, Zhang J. Locally administered adipose-derived stem cells accelerate wound healing through differentiation and vasculogenesis. Cell Transplant 2011;20:205–16. [DOI] [PubMed] [Google Scholar]
- [29].Lee SH, Jin SY, Song JS, Seo KK, Cho KH. Paracrine effects of adipose-derived stem cells on keratinocytes and dermal fibroblasts. Ann Dermatol 2012;24:136–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Kato Y, Iwata T, Washio K, Yoshida T, Kuroda H, Morikawa S, et al. Creation and Transplantation of an Adipose-derived Stem Cell (ASC) Sheet in a Diabetic Wound-healing Model. J Vis Exp 2017;2017:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng 2001;7:211–28. [DOI] [PubMed] [Google Scholar]
- [32].Lu F, Li J, Gao J, Ogawa R, Ou C, Yang B, et al. Improvement of the survival of human autologous fat transplantation by using VEGF-transfected adipose-derived stem cells. Plast Reconstr Surg 2009;124:1437–46. [DOI] [PubMed] [Google Scholar]
- [33].Ma D, Kua JE, Lim WK, Lee ST, Chua AW. In vitro characterization of human hair follicle dermal sheath mesenchymal stromal cells and their potential in enhancing diabetic wound healing. Cytotherapy 2015;17:1036–51. [DOI] [PubMed] [Google Scholar]
- [34].Hsiao ST, Asgari A, Lokmic Z, Sinclair R, Dusting GJ, Lim SY, et al. Comparative analysis of paracrine factor expression in human adult mesenchymal stem cells derived from bone marrow, adipose, and dermal tissue. Stem Cells Dev 2012;21:2189–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Liu X, Wang Z, Wang R, Zhao F, Shi P, Jiang Y, et al. Direct comparison of the potency of human mesenchymal stem cells derived from amnion tissue, bone marrow and adipose tissue at inducing dermal fibroblast responses to cutaneous wounds. Int J Mol Med 2013;31:407–15. [DOI] [PubMed] [Google Scholar]
- [36].Boink MA, van den Broek LJ, Roffel S, Nazmi K, Bolscher JG, Gefen A, et al. Different wound healing properties of dermis, adipose, and gingiva mesenchymal stromal cells. Wound Repair Regen 2016;24:100–9. [DOI] [PubMed] [Google Scholar]
- [37].Martinez ML, Escario E, Poblet E, Sanchez D, Buchon FF, Izeta A, et al. Hair follicle-containing punch grafts accelerate chronic ulcer healing: a randomized controlled trial. J Am Acad Dermatol 2016;75:1007–14. [DOI] [PubMed] [Google Scholar]
- [38].Hass R, Kasper C, Bohm S, Jacobs R. Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Commun Signal 2011;9:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Heo JS, Choi Y, Kim HS, Kim HO. Comparison of molecular profiles of human mesenchymal stem cells derived from bone marrow, umbilical cord blood, placenta and adipose tissue. Int J Mol Med 2016;37:115–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Li CY, Wu XY, Tong JB, Yang XX, Zhao JL, Zheng QF, et al. Comparative analysis of human mesenchymal stem cells from bone marrow and adipose tissue under xeno-free conditions for cell therapy. Stem Cell Res Ther 2015;6:55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Kern S, Eichler H, Stoeve J, Kluter H, Bieback K. Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells 2006;24:1294–301. [DOI] [PubMed] [Google Scholar]
- [42].Ding DC, Shyu WC, Lin SZ. Mesenchymal stem cells. Cell Transplant 2011;20:5–14. [DOI] [PubMed] [Google Scholar]
- [43].Pittenger MF, Discher DE, Peault BM, Phinney DG, Hare JM, Caplan AI. Mesenchymal stem cell perspective: cell biology to clinical progress. NPJ Regen Med 2019;4:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Bernardo ME, Fibbe WE. Mesenchymal stromal cells: sensors and switchers of inflammation. Cell Stem Cell 2013;13:392–402. [DOI] [PubMed] [Google Scholar]
- [45].De Miguel MP, Fuentes-Julian S, Blazquez-Martinez A, Pascual CY, Aller MA, rias J, et al. Immunosuppressive properties of mesenchymal stem cells: advances and applications. Curr Mol Med 2012;12:574–91. [DOI] [PubMed] [Google Scholar]
- [46].Fan XL, Zhang Y, Li X, Fu QL. Mechanisms underlying the protective effects of mesenchymal stem cell-based therapy. Cell Mol Life Sci 2020;77:2771–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Mishra VK, Shih HH, Parveen F, Lenzen D, Ito E, Chan TF, et al. Identifying the Therapeutic Significance of Mesenchymal Stem Cells. Cells 2020;9:1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Park SR, Kim JW, Jun HS, Roh JY, Lee HY, Hong IS. Stem Cell Secretome and Its Effect on Cellular Mechanisms Relevant to Wound Healing. Mol Ther 2018;26:606–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Motegi SI, Ishikawa O. Mesenchymal stem cells: the roles and functions in cutaneous wound healing and tumor growth. J Dermatol Sci 2017;86:83–9. [DOI] [PubMed] [Google Scholar]
- [50].Wilgus TA, Roy S, McDaniel JC. Neutrophils and Wound Repair: Positive Actions and Negative Reactions. Adv Wound Care (New Rochelle) 2013;2:379–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Maxson S, Lopez EA, Yoo D, Danilkovitch-Miagkova A, Leroux MA. Concise review: role of mesenchymal stem cells in wound repair. Stem Cells Transl Med 2012;1:142–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Wu Y, Chen L, Scott PG, Tredget EE. Mesenchymal stem cells enhance wound healing through differentiation and angiogenesis. Stem Cells 2007;25:2648–59. [DOI] [PubMed] [Google Scholar]
- [53].Ma S, Xie N, Li W, Yuan B, Shi Y, Wang Y. Immunobiology of mesenchymal stem cells. Cell Death Differ 2014;21:216–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Vander Beken S, de Vries JC, Meier-Schiesser B, Meyer P, Jiang D, Sindrilaru A, et al. Newly Defined ATP-Binding Cassette Subfamily B Member 5 Positive Dermal Mesenchymal Stem Cells Promote Healing of Chronic Iron-Overload Wounds via Secretion of Interleukin-1 Receptor Antagonist. Stem Cells 2019;37:1057–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Fang F, Huang RL, Zheng Y, Liu M, Huo R. Bone marrow derived mesenchymal stem cells inhibit the proliferative and profibrotic phenotype of hypertrophic scar fibroblasts and keloid fibroblasts through paracrine signaling. J Dermatol Sci 2016;83:95–105. [DOI] [PubMed] [Google Scholar]
- [56].Zimmerlin L, Park TS, Zambidis ET, Donnenberg VS, Donnenberg AD. Mesenchymal stem cell secretome and regenerative therapy after cancer. Biochimie 2013;95:2235–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Wood S, Jayaraman V, Huelsmann EJ, Bonish B, Burgad D, Sivaramakrishnan G, et al. Pro-inflammatory chemokine CCL2 (MCP-1) promotes healing in diabetic wounds by restoring the macrophage response. PLoS One 2014;9:e91574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Ferreira JR, Teixeira GQ, Santos SG, Barbosa MA, Almeida-Porada G, Goncalves RM. Mesenchymal Stromal Cell Secretome: Influencing Therapeutic Potential by Cellular Pre-conditioning. Front Immunol 2018;9:2837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Sasaki M, Abe R, Fujita Y, Ando S, Inokuma D, Shimizu H. Mesenchymal stem cells are recruited into wounded skin and contribute to wound repair by trans-differentiation into multiple skin cell type. J Immunol 2008;180:2581–7. [DOI] [PubMed] [Google Scholar]
- [60].Ishida Y, Kimura A, Kuninaka Y, Inui M, Matsushima K, Mukaida N, et al. Pivotal role of the CCL5/CCR5 interaction for recruitment of endothelial progenitor cells in mouse wound healing. J Clin Invest 2012;122:711–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Fang Y, Shen J, Yao M, Beagley KW, Hambly BD, Bao S. Granulocyte-macrophage colony-stimulating factor enhances wound healing in diabetes via upregulation of proinflammatory cytokines. Br J Dermatol 2010;162:478–86. [DOI] [PubMed] [Google Scholar]
- [62].Hoang DH, Nguyen TD, Nguyen HP, Nguyen XH, Do PTX, Dang VD, et al. Differential Wound Healing Capacity of Mesenchymal Stem Cell-Derived Exosomes Originated From Bone Marrow, Adipose Tissue and Umbilical Cord Under Serum- and Xeno-Free Condition. Front Mol Biosci 2020;7:119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Jiang WG, Sanders AJ, Ruge F, Harding KG. Influence of interleukin-8 (IL-8) and IL-8 receptors on the migration of human keratinocytes, the role of PLC-gamma and potential clinical implications. Exp Ther Med 2012;3:231–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Chen L, Tredget EE, Wu PY, Wu Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One 2008;3:e1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Balaji S, Han N, Moles C, Shaaban AF, Bollyky PL, Crombleholme TM, et al. Angiopoietin-1 improves endothelial progenitor cell-dependent neovascularization in diabetic wounds. Surgery 2015;158:846–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Cho CH, Sung HK, Kim KT, Cheon HG, Oh GT, Hong HJ, et al. COMP-angiopoietin-1 promotes wound healing through enhanced angiogenesis, lymphangiogenesis, and blood flow in a diabetic mouse model. Proc Natl Acad Sci U S A 2006;103:4946–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Tamai K, Yamazaki T, Chino T, Ishii M, Otsuru S, Kikuchi Y, et al. PDGFRalpha-positive cells in bone marrow are mobilized by high mobility group box 1 (HMGB1) to regenerate injured epithelia. Proc Natl Acad Sci U S A 2011;108:6609–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Seppa H, Grotendorst G, Seppa S, Schiffmann E, Martin GR. Platelet-derived growth factor in chemotactic for fibroblasts. J Cell Biol 1982;92:584–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Li JF, Duan HF, Wu CT, Zhang DJ, Deng Y, Yin HL, et al. HGF accelerates wound healing by promoting the dedifferentiation of epidermal cells through beta1-integrin/ILK pathway. Biomed Res Int 2013;2013:470418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Shen C, Lie P, Miao T, Yu M, Lu Q Feng T, et al. Conditioned medium from umbilical cord mesenchymal stem cells induces migration and angiogenesis. Mol Med Rep 2015;12:20–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Caley MP, Martins VL, O’Toole EA. Metalloproteinases and Wound Healing. Adv Wound Care (New Rochelle) 2015;4:225–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Idrovo JP, Yang WL, Jacob A, Ajakaiye MA, Cheyuo C, Wang Z, et al. Combination of adrenomedullin with its binding protein accelerates cutaneous wound healing. PLoS One 2015;10:e0120225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Phinney DG, Pittenger MF. Concise Review: MSC-Derived Exosomes for Cell-Free Therapy. Stem Cells 2017;35:851–8. [DOI] [PubMed] [Google Scholar]
- [74].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. [DOI] [PubMed] [Google Scholar]
- [75].Keshtkar S, Azarpira N, Ghahremani MH. Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Res Ther 2018;9:63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Tsiapalis D, O'Driscoll L. Mesenchymal Stem Cell Derived Extracellular Vesicles for Tissue Engineering and Regenerative Medicine Applications. Cells 2020;9:991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Gowen A, Shahjin F, Chand S, Odegaard KE, Yelamanchili SV. Mesenchymal Stem Cell-Derived Extracellular Vesicles: Challenges in Clinical Applications. Front Cell Dev Biol 2020;8:149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Hu P, Yang Q, Wang Q, Shi C, Wang D, Armato U, et al. Mesenchymal stromal cells-exosomes: a promising cell-free therapeutic tool for wound healing and cutaneous regeneration. Burns Trauma 2019;7:38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Kusuma GD, Carthew J, Lim R, Frith JE. Effect of the Microenvironment on Mesenchymal Stem Cell Paracrine Signaling: Opportunities to Engineer the Therapeutic Effect. Stem Cells Dev 2017;26:617–31. [DOI] [PubMed] [Google Scholar]
- [80].Fui LW, Lok MPW, Govindasamy V, Yong TK, Lek TK, Das AK. Understanding the multifaceted mechanisms of diabetic wound healing and therapeutic application of stem cells conditioned medium in the healing process. J Tissue Eng Regen Med 2019;13:2218–33. [DOI] [PubMed] [Google Scholar]
- [81].Maranda EL, Rodriguez-Menocal L, Badiavas EV. Role of Mesenchymal Stem Cells in Dermal Repair in Burns and Diabetic Wounds. Curr Stem Cell Res Ther 2017;12:61–70. [DOI] [PubMed] [Google Scholar]
- [82].Le Blanc K, Rasmusson I, Sundberg B, Gotherstrom C, Hassan M, Uzunel M, et al. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 2004;363:1439–41. [DOI] [PubMed] [Google Scholar]
- [83].Fang B, Song Y, Liao L, Zhang Y, Zhao RC. Favorable response to human adipose tissue-derived mesenchymal stem cells in steroid-refractory acute graft-versus-host disease. Transplant Proc 2007;39:3358–62. [DOI] [PubMed] [Google Scholar]
- [84].Ball LM, Bernardo ME, Roelofs H, van Tol MJ, Contoli B, Zwaginga JJ, et al. Multiple infusions of mesenchymal stromal cells induce sustained remission in children with steroid-refractory, grade III-IV acute graft-versus-host disease. Br J Haematol 2013;163:501–9. [DOI] [PubMed] [Google Scholar]
- [85].Introna M, Lucchini G, Dander E, Galimberti S, Rovelli A, Balduzzi A, et al. Treatment of graft versus host disease with mesenchymal stromal cells: a phase I study on 40 adult and pediatric patients. Biol Blood Marrow Transplant 2014;20:375–81. [DOI] [PubMed] [Google Scholar]
- [86].Kurtzberg J, Prockop S, Teira P, Bittencourt H, Lewis V, Chan KW, et al. Allogeneic human mesenchymal stem cell therapy (remestemcel-L, Prochymal) as a rescue agent for severe refractory acute graft-versus-host disease in pediatric patients. Biol Blood Marrow Transplant 2014;20:229–35. [DOI] [PubMed] [Google Scholar]
- [87].Kim KH, Blasco-Morente G, Cuende N, Arias-Santiago S. Mesenchymal stromal cells: properties and role in management of cutaneous diseases. J Eur Acad Dermatol Venereol 2017;31:414–23. [DOI] [PubMed] [Google Scholar]
- [88].Shin TH, Kim HS, Choi SW, Kang KS. Mesenchymal Stem Cell Therapy for Inflammatory Skin Diseases: Clinical Potential and Mode of Action. Int J Mol Sci 2017;18:244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Golchin A, Farahany TZ, Khojasteh A, Soleimanifar F, Ardeshirylajimi A. The Clinical Trials of Mesenchymal Stem Cell Therapy in Skin Diseases: An Update and Concise Review. Curr Stem Cell Res Ther 2019;14:22–33. [DOI] [PubMed] [Google Scholar]
- [90].Daltro SRT, Meira CS, Santos IP, Ribeiro Dos Santos R, Soares MBP. Mesenchymal Stem Cells and Atopic Dermatitis: A Review. Front Cell Dev Biol 2020;8:326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Paganelli A, Tarentini E, Benassi L, Kaleci S, Magnoni C. Mesenchymal stem cells for the treatment of psoriasis: a comprehensive review. Clin Exp Dermatol 2020;45:824–30. [DOI] [PubMed] [Google Scholar]
- [92].Khosrotehrani K. Mesenchymal stem cell therapy in skin: why and what for? Exp Dermatol 2013;22:307–10. [DOI] [PubMed] [Google Scholar]
- [93].Hsu CK, Wang SP, Lee JY, McGrath JA. Treatment of hereditary epidermolysis bullosa: updates and future prospects. Am J Clin Dermatol 2014;15:1–6. [DOI] [PubMed] [Google Scholar]
- [94].Keith AR, Twaroski K, Ebens CL, Tolar J. Leading edge: emerging drug, cell, and gene therapies for junctional epidermolysis bullosa. Expert Opin Biol Ther 2020;20:911–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Murphree RW. Impairments in Skin Integrity. Nurs Clin North Am 2017;52:405–17. [DOI] [PubMed] [Google Scholar]
- [96].Liubaviciute A, Ivaskiene T, Biziuleviciene G. Modulated mesenchymal stromal cells improve skin wound healing. Biologicals 2020;67:1–8. [DOI] [PubMed] [Google Scholar]
- [97].Lee DE, Ayoub N, Agrawal DK. Mesenchymal stem cells and cutaneous wound healing: novel methods to increase cell delivery and therapeutic efficacy. Stem Cell Res Ther 2016;7:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Walter MN, Wright KT, Fuller HR, MacNeil S, Johnson WE. Mesenchymal stem cell-conditioned medium accelerates skin wound healing: an in vitro study of fibroblast and keratinocyte scratch assays. Exp Cell Res 2010;316:1271–81. [DOI] [PubMed] [Google Scholar]
- [99].Smith AN, Willis E, Chan VT, Muffley LA, Isik FF, Gibran NS, et al. Mesenchymal stem cells induce dermal fibroblast responses to injury. Exp Cell Res 2010;316:48–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].Jeon YK, Jang YH, Yoo DR, Kim SN, Lee SK, Nam MJ. Mesenchymal stem cells' interaction with skin: wound-healing effect on fibroblast cells and skin tissue. Wound Repair Regen 2010;18:655–61. [DOI] [PubMed] [Google Scholar]
- [101].Has C, Fischer J. Inherited epidermolysis bullosa: New diagnostics and new clinical phenotypes. Exp Dermatol 2019;28:1146–52. [DOI] [PubMed] [Google Scholar]
- [102].Cianfarani F, Zambruno G, Castiglia D, Odorisio T. Pathomechanisms of Altered Wound Healing in Recessive Dystrophic Epidermolysis Bullosa. Am J Pathol 2017;187:1445–53. [DOI] [PubMed] [Google Scholar]
- [103].Nystrom A, Velati D, Mittapalli VR, Fritsch A, Kern JS, Bruckner-Tuderman L. Collagen VII plays a dual role in wound healing. J Clin Invest 2013;123:3498–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Varki R, Sadowski S, Uitto J, Pfendner E. Epidermolysis bullosa. II. Type VII collagen mutations and phenotype-genotype correlations in the dystrophic subtypes. J Med Genet 2007;44:181−92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Nystrom A, Bruckner-Tuderman L. Injury- and inflammation-driven skin fibrosis: The paradigm of epidermolysis bullosa. Matrix Biol 2018;68-69:547–60. [DOI] [PubMed] [Google Scholar]
- [106].Ebens CL, McGrath JA, Tamai K, Hovnanian A, Wagner JE, Riddle MJ, et al. Bone marrow transplant with post-transplant cyclophosphamide for recessive dystrophic epidermolysis bullosa expands the related donor pool and permits tolerance of nonhaematopoietic cellular grafts. Br J Dermatol 2019;181:1238–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Tolar J, Ishida-Yamamoto A, Riddle M, McElmurry RT, Osborn M, Xia L, et al. Amelioration of epidermolysis bullosa by transfer of wild-type bone marrow cells. Blood 2009;113:1167–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Tolar J, Wagner JE. Allogeneic blood and bone marrow cells for the treatment of severe epidermolysis bullosa: repair of the extracellular matrix. Lancet 2013;382:1214–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Wagner JE, Ishida-Yamamoto A, McGrath JA, Hordinsky M, Keene DR, Woodley DT, et al. Bone marrow transplantation for recessive dystrophic epidermolysis bullosa. N Engl J Med 2010;363:629–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Webber BR, Tolar J. From marrow to matrix: novel gene and cell therapies for epidermolysis bullosa. Mol Ther 2015;23:987–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Nevala-Plagemann C, Lee C, Tolar J. Placenta-based therapies for the treatment of epidermolysis bullosa. Cytotherapy 2015;17:786–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].McBride JD, Rodriguez-Menocal L, Candanedo A, Guzman W, Garcia-Contreras M, Badiavas EV. Dual mechanism of type VII collagen transfer by bone marrow mesenchymal stem cell extracellular vesicles to recessive dystrophic epidermolysis bullosa fibroblasts. Biochimie 2018;155:50–8. [DOI] [PubMed] [Google Scholar]
- [113].Nakayama C, Fujita Y, Matsumura W, Ujiie I, Takashima S, Shinkuma S, et al. The development of induced pluripotent stem cell-derived mesenchymal stem/stromal cells from normal human and RDEB epidermal keratinocytes. J Dermatol Sci 2018;91:301–10. [DOI] [PubMed] [Google Scholar]
- [114].Petrova A, Georgiadis C, Fleck RA, Allison L, McGrath JA, Dazzi F, et al. Human Mesenchymal Stromal Cells Engineered to Express Collagen VII Can Restore Anchoring Fibrils in Recessive Dystrophic Epidermolysis Bullosa Skin Graft Chimeras. J Invest Dermatol 2020;140:121–31. e6. [DOI] [PubMed] [Google Scholar]
- [115].Alexeev V, Uitto J, Igoucheva O. Gene expression signatures of mouse bone marrow-derived mesenchymal stem cells in the cutaneous environment and therapeutic implications for blistering skin disorder. Cytotherapy 2011;13:30–45. [DOI] [PubMed] [Google Scholar]
- [116].Perdoni C, McGrath JA, Tolar J. Preconditioning of mesenchymal stem cells for improved transplantation efficacy in recessive dystrophic epidermolysis bullosa. Stem Cell Res Ther 2014;5:121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Iinuma S, Aikawa E, Tamai K, Fujita R, Kikuchi Y, Chino T, et al. Transplanted bone marrow-derived circulating PDGFRalpha+ cells restore type VII collagen in recessive dystrophic epidermolysis bullosa mouse skin graft. J Immunol 2015;194:1996–2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Schatton T, Yang J, Kleffel S, Uehara M, Barthel SR, Schlapbach C, et al. ABCB5 Identifies Immunoregulatory Dermal Cells. Cell Rep 2015;12:1564–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Webber BR, O'Connor KT, McElmurry RT, Durgin EN, Eide CR, Lees CJ, et al. Rapid generation of Col7a1(−/−) mouse model of recessive dystrophic epidermolysis bullosa and partial rescue via immunosuppressive dermal mesenchymal stem cells. Lab Invest 2017;97:1218–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Kerstan A, Niebergall-Roth E, Esterlechner J, Schroder HM, Gasser M, Waaga-Gasser AM, et al. Ex vivo-expanded highly pure ABCB5(+) mesenchymal stromal cells as Good Manufacturing Practice-compliant autologous advanced therapy medicinal product for clinical use: process validation and first in-human data. Cytotherapy 2021;23:165–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Rashidghamat E, Kadiyirire T, Ayis S, Petrof G, Liu L, Pullabhatla V, et al. Phase I/II open-label trial of intravenous allogeneic mesenchymal stromal cell therapy in adults with recessive dystrophic epidermolysis bullosa. J Am Acad Dermatol 2020;83:447–54. [DOI] [PubMed] [Google Scholar]
- [122].El-Darouti M, Fawzy M, Amin I, Abdel Hay R, Hegazy R, Gabr H, et al. Treatment of dystrophic epidermolysis bullosa with bone marrow non-hematopoeitic stem cells: a randomized controlled trial. Dermatol Ther 2016;29:96–100. [DOI] [PubMed] [Google Scholar]
- [123].Petrof G, Lwin SM, Martinez-Queipo M, Abdul-Wahab A, Tso S, Mellerio JE, et al. Potential of Systemic Allogeneic Mesenchymal Stromal Cell Therapy for Children with Recessive Dystrophic Epidermolysis Bullosa. J Invest Dermatol 2015;135:2319–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Conget P, Rodriguez F, Kramer S, Allers C, Simon V, Palisson F, et al. Replenishment of type VII collagen and re-epithelialization of chronically ulcerated skin after intradermal administration of allogeneic mesenchymal stromal cells in two patients with recessive dystrophic epidermolysis bullosa. Cytotherapy 2010;12:429–31. [DOI] [PubMed] [Google Scholar]
- [125].Peck MD. Epidemiology of burns throughout the world. Part I: Distribution and risk factors. Burns 2011;37:1087–100. [DOI] [PubMed] [Google Scholar]
- [126].Rangatchew F, Vester-Glowinski P, Rasmussen BS, Haastrup E, Munthe-Fog L, Talman ML, et al. Mesenchymal stem cell therapy of acute thermal burns: a systematic review of the effect on inflammation and wound healing. Burns 2021;47:270–94. [DOI] [PubMed] [Google Scholar]
- [127].Abo-Elkheir W, Hamza F, Elmofty AM, Emam A, Abdl-Moktader M, Elsherefy S, et al. Role of cord blood and bone marrow mesenchymal stem cells in recent deep burn: a case-control prospective study. Am J Stem Cells 2017;6:23–35. [PMC free article] [PubMed] [Google Scholar]
- [128].Rasulov MF, Vasilchenkov AV, Onishchenko NA, Krasheninnikov ME, Kravchenko VI, Gorshenin TL, et al. First experience of the use bone marrow mesenchymal stem cells for the treatment of a patient with deep skin burns. Bull Exp Biol Med 2005;139:141–4. [DOI] [PubMed] [Google Scholar]
- [129].Mansilla E, Marin GH, Berges M, Scafatti S, Rivas J, Nunez A, et al. Cadaveric bone marrow mesenchymal stem cells: first experience treating a patient with large severe burns. Burns Trauma 2015;3:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Okonkwo UA, DiPietro LA. Diabetes and Wound Angiogenesis. Int J Mol Sci 2017;18:1419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [131].Lam T,M. Mesenchymal Stem Cell Therapies for Skin Repair and Regeneration. J Dermat Cosmetol 2017;1:62–4. [Google Scholar]
- [132].Sylakowski K, Bradshaw A, Wells A. Mesenchymal Stem Cell/Multipotent Stromal Cell Augmentation of Wound Healing: Lessons from the Physiology of Matrix and Hypoxia Support. Am J Pathol 2020;190:1370–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Lee EY, Xia Y, Kim WS, Kim MH, Kim TH, Kim KJ, et al. Hypoxia-enhanced wound-healing function of adipose-derived stem cells: increase in stem cell proliferation and up-regulation of VEGF and bFGF. Wound Repair Regen 2009;17:540–7. [DOI] [PubMed] [Google Scholar]
- [134].Chen L, Xu Y, Zhao J, Zhang Z, Yang R, Xie J, et al. Conditioned medium from hypoxic bone marrow-derived mesenchymal stem cells enhances wound healing in mice. PLoS One 2014;9:e96161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Jun EK, Zhang Q, Yoon BS, Moon JH, Lee G, Park G, et al. Hypoxic conditioned medium from human amniotic fluid-derived mesenchymal stem cells accelerates skin wound healing through TGF-beta/SMAD2 and PI3K/Akt pathways. Int J Mol Sci 2014;15:605–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [136].Li M, Zhao Y, Hao H, Dai H, Han Q, Tong C, et al. Mesenchymal stem cell-conditioned medium improves the proliferation and migration of keratinocytes in a diabetes-like microenvironment. Int J Low Extrem Wounds 2015;14:73–86. [DOI] [PubMed] [Google Scholar]
- [137].Cao Y, Gang X, Sun C, Wang G. Mesenchymal Stem Cells Improve Healing of Diabetic Foot Ulcer. J Diabetes Res 2017;2017:9328347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Prochazka V, Gumulec J, Jaluvka F, Salounova D, Jonszta T, Czerny D, et al. Cell therapy, a new standard in management of chronic critical limb ischemia and foot ulcer. Cell Transplant 2010;19:1413–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Otero-Vinas M, Falanga V. Mesenchymal Stem Cells in Chronic Wounds: The Spectrum from Basic to Advanced Therapy. Adv Wound Care (New Rochelle) 2016;5:149–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Krasnodembskaya A, Song Y, Fang X, Gupta N, Serikov V, Lee JW, et al. Antibacterial effect of human mesenchymal stem cells is mediated in part from secretion of the antimicrobial peptide LL-37. Stem Cells 2010;28:2229–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Yagi H, Chen AF, Hirsch D, Rothenberg AC, Tan J, Alexander PG, et al. Antimicrobial activity of mesenchymal stem cells against Staphylococcus aureus. Stem Cell Res Ther 2020;11:293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Wood CR, Al Dhahri D, Al Delfi I, Pickles NA, Sammons RL, Worthington T, et al. Human adipose tissue-derived mesenchymal stem/stromal cells adhere to and inhibit the growth of Staphylococcus aureus and Pseudomonas aeruginosa. J Med Microbiol 2018;67:1789–95. [DOI] [PubMed] [Google Scholar]
- [143].Ren Z, Zheng X, Yang H, Zhang Q, Liu X, Zhang X, et al. Human umbilical-cord mesenchymal stem cells inhibit bacterial growth and alleviate antibiotic resistance in neonatal imipenem-resistant Pseudomonas aeruginosa infection. Innate Immun 2020;26:215–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [144].Eberhard Y, Ortiz S, Ruiz Lascano A, Kuznitzky R, Serra HM. Up-regulation of the chemokine CCL21 in the skin of subjects exposed to irritants. BMC Immunol 2004;5:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [145].Harting MT, Jimenez F, Xue H, Fischer UM, Baumgartner J, Dash PK, et al. Intravenous mesenchymal stem cell therapy for traumatic brain injury. J Neurosurg 2009;110:1189–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [146].Fischer UM, Harting MT, Jimenez F, Monzon-Posadas WO, Xue H, Savitz SI, et al. Pulmonary passage is a major obstacle for intravenous stem cell delivery: the pulmonary first-pass effect. Stem Cells Dev 2009;18:683–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [147].Rizzi SC, Upton Z, Bott K, Dargaville TR. Recent advances in dermal wound healing: biomedical device approaches. Expert Rev Med Devices 2010;7:143–54. [DOI] [PubMed] [Google Scholar]
- [148].Yannas IV, Burke JF, Orgill DP, Skrabut EM. Wound tissue can utilize a polymeric template to synthesize a functional extension of skin. Science 1982;215:174–6. [DOI] [PubMed] [Google Scholar]
- [149].Yannas IV, Lee E, Orgill DP, Skrabut EM, Murphy GF. Synthesis and characterization of a model extracellular matrix that induces partial regeneration of adult mammalian skin. Proc Natl Acad Sci U S A 1989;86:933–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Shafiee A, Cavalcanti AS, Saidy NT, Schneidereit D, Friedrich O, Ravichandran A, et al. Convergence of 3D printed biomimetic wound dressings and adult stem cell therapy. Biomaterials 2021;268:120558. [DOI] [PubMed] [Google Scholar]
- [151].Ma K, Laco F, Ramakrishna S, Liao S, Chan CK. Differentiation of bone marrow-derived mesenchymal stem cells into multi-layered epidermis-like cells in 3D organotypic coculture. Biomaterials 2009;30:3251–8. [DOI] [PubMed] [Google Scholar]
- [152].Ong SY, Dai H, Leong KW. Inducing hepatic differentiation of human mesenchymal stem cells in pellet culture. Biomaterials 2006;27:4087–97. [DOI] [PubMed] [Google Scholar]
- [153].Gurkan UA, El Assal R, Yildiz SE, Sung Y, Trachtenberg AJ, Kuo WP, et al. Engineering anisotropic biomimetic fibrocartilage microenvironment by bioprinting mesenchymal stem cells in nanoliter gel droplets. Mol Pharm 2014;11:2151–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [154].Moore CA, Shah NN, Smith CP, Rameshwar P. 3D Bioprinting and Stem Cells. Methods Mol Biol 2018;1842:93–103. [DOI] [PubMed] [Google Scholar]
- [155].Jang EH, Kim JH, Lee JH, Kim DH, Youn YN. Enhanced Biocompatibility of Multi-Layered, 3D Bio-Printed Artificial Vessels Composed of Autologous Mesenchymal Stem Cells. Polymers (Basel) 2020;12:538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Park JS, Suryaprakash S, Lao YH, Leong KW. Engineering mesenchymal stem cells for regenerative medicine and drug delivery. Methods 2015;84:3–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [157].Hodgkinson CP, Gomez JA, Mirotsou M, Dzau VJ. Genetic engineering of mesenchymal stem cells and its application in human disease therapy. Hum Gene Ther 2010;21:1513–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [158].Nowakowski A, Walczak P, Lukomska B, Janowski M. Genetic Engineering of Mesenchymal Stem Cells to Induce Their Migration and Survival. Stem Cells Int 2016;2016:4956063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [159].Lee K, Lee N, Shin E, Lee N, Chang J, Na D, et al. Enhancing the therapeutic potential of mesenchymal stem cell-based therapy via CRISPR/Cas9-based genome editing. Cytotherapy 2020;20:S15. [Google Scholar]
- [160].Mavilio F, Pellegrini G, Ferrari S, Di Nunzio F, Di Iorio E, Recchia A, et al. Correction of junctional epidermolysis bullosa by transplantation of genetically modified epidermal stem cells. Nat Med 2006;12:1397–402. [DOI] [PubMed] [Google Scholar]
- [161].Bauer JW, Koller J, Murauer EM, De Rosa L, Enzo E, Carulli S, et al. Closure of a Large Chronic Wound through Transplantation of Gene-Corrected Epidermal Stem Cells. J Invest Dermatol 2017;137:778–81. [DOI] [PubMed] [Google Scholar]
- [162].Hirsch T, Rothoeft T, Teig N, Bauer JW, Pellegrini G, De Rosa L, et al. Regeneration of the entire human epidermis using transgenic stem cells. Nature 2017;551:327–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [163].Bruckner-Tuderman L. Newer Treatment Modalities in Epidermolysis Bullosa. Indian Dermatol Online J 2019;10:244–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Galipeau J, Sensebe L. Mesenchymal Stromal Cells: Clinical Challenges and Therapeutic Opportunities. Cell Stem Cell 2018;22:824–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [165].Martin I, Galipeau J, Kessler C, Le Blanc K, Dazzi F. Challenges for mesenchymal stromal cell therapies. Sci Transl Med 2019;11:1–4. [DOI] [PubMed] [Google Scholar]
- [166].Stupple A, Singerman D, Cell LA. The reproducibility crisis in the age of digital medicine. NPJ Digit Med 2019;2:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [167].Moll G, Hoogduijn MJ, Ankrum JA. Editorial: Safety, Efficacy and Mechanisms of Action of Mesenchymal Stem Cell Therapies. Front Immunol 2020;11:243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [168].Madrigal M, Rao KS, Riordan NH. A review of therapeutic effects of mesenchymal stem cell secretions and induction of secretory modification by different culture methods. J Transl Med 2014;12:260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [169].L PK, Kandoi S, Misra R, S V, K R, Verma RS. The mesenchymal stem cell secretome: a new paradigm towards cell-free therapeutic mode in regenerative medicine. Cytokine Growth Factor Rev 2019;46:1–9. [DOI] [PubMed] [Google Scholar]
- [170].Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 2007;449:557–63. [DOI] [PubMed] [Google Scholar]
- [171].Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol 2008;8:726–36. [DOI] [PubMed] [Google Scholar]
- [172].Suzuki K, Sun R, Origuchi M, Kanehira M, Takahata T, Itoh J, et al. Mesenchymal stromal cells promote tumor growth through the enhancement of neovascularization. Mol Med 2011;17:579–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [173].Xie C, Yang Z, Suo Y, Chen Q, Wei D, Weng X, et al. Systemically Infused Mesenchymal Stem Cells Show Different Homing Profiles in Healthy and Tumor Mouse Models. Stem Cells Transl Med 2017;6:1120–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [174].Kim M, Rhee JK, Choi H, Kwon A, Kim J, Lee GD, et al. Passage-dependent accumulation of somatic mutations in mesenchymal stromal cells during in vitro culture revealed by whole genome sequencing. Sci Rep 2017;7:14508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [175].Nikitina V, Astrelina T, Nugis V, Ostashkin A, Karaseva T, Dobrovolskaya E, et al. Clonal chromosomal and genomic instability during human multipotent mesenchymal stromal cells long-term culture. PLoS One 2018;13:e0192445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [176].Huang YZ, Gou M, Da LC, Zhang WQ, Xie HQ, Mesenchymal Stem Cells for Chronic Wound Healing: Current Status of Preclinical and Clinical Studies. Tissue Eng Part B Rev 2020;26:555–70. [DOI] [PubMed] [Google Scholar]
- [177].Popova AP, Bozyk PD, Goldsmith AM, Linn MJ, Lei J, Bentley JK, et al. Autocrine production of TGF-beta1 promotes myofibroblastic differentiation of neonatal lung mesenchymal stem cells. Am J Physiol Lung Cell Mol Physiol 2010;298:L735–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [178].Li W, Ren G, Huang Y, Su J, Han Y, Li J, et al. Mesenchymal stem cells: a double-edged sword in regulating immune responses. Cell Death Differ 2012;19:1505–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [179].Wu Z, Zhang S, Zhou L, Cai J, Tan J, Gao X, et al. Thromboembolism Induced by Umbilical Cord Mesenchymal Stem Cell Infusion: A Report of Two Cases and Literature Review. Transplant Proc 2017;49:1656–8. [DOI] [PubMed] [Google Scholar]
- [180].Wang H, Strange C, Nietert PJ, Wang J, Turnbull TL, Cloud C, et al. Autologous Mesenchymal Stem Cell and Islet Cotransplantation: Safety and Efficacy. Stem Cells Transl Med 2018;7:11–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [181].Melmed GY, Pandak WM, Casey K, Abraham B, Valentine J, Schwartz D, et al. Human Placenta-derived Cells (PDA-001) for the Treatment of Moderate-to-severe Crohn's Disease: A Phase 1b/2a Study. Inflamm Bowel Dis 2015;21:1809–16. [DOI] [PubMed] [Google Scholar]
- [182].Trujillo-Rodriguez M, Viciana P, Rivas-Jeremias I, Alvarez-Rios AI, Ruiz-Garcia A, Espinosa-Ibanez O, et al. Mesenchymal stromal cells in human immunodeficiency virus-infected patients with discordant immune response: Early results of a phase I/II clinical trial. Stem Cells Transl Med 2021;10:534–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [183].Stephenne X, Nicastro E, Eeckhoudt S, Hermans C, Nyabi O, Lombard C, et al. Bivalirudin in combination with heparin to control mesenchymal cell procoagulant activity. PLoS One 2012;7:e42819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [184].Liao L, Shi B, Chang H, Su X, Zhang L, Bi C, et al. Heparin improves BMSC cell therapy: Anticoagulant treatment by heparin improves the safety and therapeutic effect of bone marrow-derived mesenchymal stem cell cytotherapy. Theranostics 2017;7:106–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [185].Ge J, Guo L, Wang S, Zhang Y, Cai T, Zhao RC, et al. The size of mesenchymal stem cells is a significant cause of vascular obstructions and stroke. Stem Cell Rev Rep 2014;10:295–303. [DOI] [PubMed] [Google Scholar]
- [186].Coppin LCF, Smets F, Ambroise J, Sokal EEM, Stephenne X. Infusion-related thrombogenesis by liver-derived mesenchymal stem cells controlled by anticoagulant drugs in 11 patients with liver-based metabolic disorders. Stem Cell Res Ther 2020;11:51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [187].Fong CY, Tam K, Cheyyatraivendran S, Gan SU, Gauthaman K, Armugam A, et al. Human Wharton’s jelly stem cells and its conditioned medium enhance healing of excisional and diabetic wounds. J Cell Biochem 2014;115:290–302. [DOI] [PubMed] [Google Scholar]
- [188].Sun J, Zhang Y, Song X, Zhu J, Zhu Q, The Healing Effects of Conditioned Medium Derived from Mesenchymal Stem Cells on Radiation-Induced Skin Wounds in Rats. Cell Transplant 2019;28:105–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [189].Grisendi G, Anneren C, Cafarelli L, Sternieri R, Veronesi E, Cervo GL, et al. GMP-manufactured density gradient media for optimized mesenchymal stromal/stem cell isolation and expansion. Cytotherapy 2010;12:466–77. [DOI] [PubMed] [Google Scholar]
- [190].Wuchter P, Vetter M, Saffrich R, Diehlmann A, Bieback K, Ho AD, et al. Evaluation of GMP-compliant culture media for in vitro expansion of human bone marrow mesenchymal stromal cells. Exp Hematol 2016;44:508–18. [DOI] [PubMed] [Google Scholar]
- [191].Fekete N, Rojewski MT, Furst D, Kreja L, Ignatius A, Dausend J, et al. GMP-compliant isolation and large-scale expansion of bone marrow-derived MSC. PLoS One 2012;7:e43255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [192].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. [DOI] [PubMed] [Google Scholar]

