Skip to main content
Stem Cells Translational Medicine logoLink to Stem Cells Translational Medicine
. 2023 Jul 24;12(9):576–587. doi: 10.1093/stcltm/szad043

Orchestration of Mesenchymal Stem/Stromal Cells and Inflammation During Wound Healing

Mengting Zhu 1,2, Lijuan Cao 3,4, Sonia Melino 5, Eleonora Candi 6, Ying Wang 7, Changshun Shao 8, Gerry Melino 9, Yufang Shi 10,, Xiaodong Chen 11,
PMCID: PMC10502569  PMID: 37487541

Abstract

Wound healing is a complex process and encompasses a number of overlapping phases, during which coordinated inflammatory responses following tissue injury play dominant roles in triggering evolutionarily highly conserved principals governing tissue repair and regeneration. Among all nonimmune cells involved in the process, mesenchymal stem/stromal cells (MSCs) are most intensely investigated and have been shown to play fundamental roles in orchestrating wound healing and regeneration through interaction with the ordered inflammatory processes. Despite recent progress and encouraging results, an informed view of the scope of this evolutionarily conserved biological process requires a clear understanding of the dynamic interplay between MSCs and the immune systems in the process of wound healing. In this review, we outline current insights into the ways in which MSCs sense and modulate inflammation undergoing the process of wound healing, highlighting the central role of neutrophils, macrophages, and T cells during the interaction. We also draw attention to the specific effects of MSC-based therapy on different pathological wound healing. Finally, we discuss how ongoing scientific advances in MSCs could be efficiently translated into clinical strategies, focusing on the current limitations and gaps that remain to be overcome for achieving preferred functional tissue regeneration.

Keywords: wound healing, MSCs, inflammation

Graphical Abstract

Graphical Abstract.

Graphical Abstract


Significance Statement.

In this review, we outline current insights into the ways in which MSCs sense and modulate inflammation in the process of wound healing, highlighting the central role of neutrophils, macrophages, and T cells during the interaction. We also draw attention to the specific effects of MSC-based therapy on different pathological wound healing. Finally, we discuss how ongoing scientific advances in MSCs could be efficiently translated into clinical strategies, focusing on the current limitations and gaps that remain to be overcome for achieving desired tissue regeneration.

Introduction

Wound healing is dynamic but a fine-tuned biological process in which damaged tissue acquires the ability to regenerate itself following impairment. Under a desirable condition of skin wound, this process undergoes distinct but overlapping well-staged phases of inflammation, cell proliferation, and remodeling,1 which involves a complex interplay between many cellular players of the skin, comprising primarily keratinocytes, fibroblasts, endothelial cells of vessels, and recruited immune cells, and their associated extracellular matrix (ECM).2,3 During the early phase of wound healing, various types of leukocytes, including neutrophils, macrophages, and T cells, are attracted to the site of injury by chemotactic signals. Neutrophils play a crucial role in eliminating bacteria and debris from the wound, while macrophages are responsible for removing dead cells and other debris and producing growth factors that promote healing. T cells become activated in response to cytokines and chemokines released by other cells such as macrophages and dendritic cells. Once activated, T cells can produce cytokines and growth factors that promote the proliferation of other cells involved in wound healing, such as fibroblasts and keratinocytes. During the proliferative phase of wound healing, fibroblasts are activated and differentiate into myofibroblasts to reconstruct the wound. ECM deposition, especially collagen triggers the re-epithelialization process.4 Keratinocytes are the primary cells involved in re-epithelialization and are stimulated to migrate and proliferate during this phase. Once they arrive at the wound site, keratinocytes divide and migrate over the wound bed to close the wound. Endothelial cells are also involved in the proliferative phase, as they aid in the formation of new blood vessels through a process called angiogenesis which is critical for providing the new tissue with essential nutrients and oxygen. Overall, these cell types work together during the proliferative phase to repair the damaged tissue and restore normal tissue function. Finally, the collagen fibers reorganize from collagen types III to I, and the tissue remodels, slowly gaining strength and flexibility by promoting epithelialization and neovascularization.5-7 However, the complex healing process of tissue repair and regeneration after the damage is still far from fully understood. Many risk factors, limitations of effective treatment, and associated pathological processes still cause deficient wound closure, and even the failure of the conserved wound healing cascade often leads to pathological fibrosis, which could lead to malignant transformation.

The application of MSC-based cell therapy in the treatment of wounds is currently an active area of investigation, providing a promising opportunity for those patients with refractory wounds. MSCs, identified as mesenchymal stem/stromal cells, have multi-lineage differentiation potential and immune-modulatory properties. They exist in almost all tissues and play an essential role in tissue regeneration and closely interact with cells of the immune system in the tissue microenvironment during the repair of damaged tissues. Technically, MSCs can be derived and amplified from several sources, including the umbilical cord, bone marrow, fat pad, and other tissues. These impressive characteristics make them a promising cell type for cell therapy.

A series of studies have shown that MSCs are able to migrate into injured tissue sites and contribute to wound repair by promoting the generation and differentiation of multiple skin cell types.1,8,9 Cell tracking and fate tracing of transplanted exogenously expanded MSCs have demonstrated that those cells can differentiate into fibroblasts, endothelial cells, keratinocytes, and skin appendages, and even form glandular structures and hair follicles, subsequently participating in cutaneous wound healing and hair regeneration.8,10 However, the proportion of transplanted MSCs differentiated is very small and the functionality of the differentiated cells remains unknown, especially when allogeneic MSCs are used. The biggest question is what is the role of the short-lived infused MSCs in promoting tissue regeneration during wound healing.

Of note, inflammation is the first step in wound healing, and a moderate inflammatory response is beneficial for wound repair. Some pro-inflammatory factors, such as interleukin-1, 6, and 8, can promote vascularization and accelerate wound healing by inhibiting endothelial cell apoptosis and activating the endothelial cells to form blood vessels.11,12 However, in the experimental injury models, hyper-inflammation has been shown to delay wound healing and result in increased scarring. Furthermore, chronic inflammation, a hallmark of the non-healing wound, predisposes to a protracted course of wound healing, unhealed wound, or even pathological transformation. Therefore, inflammation at the damaged tissue sites should be well-staged and precisely orchestrated along with the wound healing cascade. It has been shown that inflammation is critical for attracting MSCs to wound sites and increasing evidence strongly indicates that MSCs can accelerate wound healing by modulating inflammation, improving the motility of various cell types such as fibroblasts and endothelial cells, and promoting angiogenesis and extracellular matrix deposition and remodeling.2,13-15 Therefore, a meticulous understanding of the orchestration of MSCs and inflammation during the entire process of wound healing should provide key information for better treatment of wounds, as well as the acquired knowledge about tissue microenvironment during pathological processes such as fibrosis and tumor development.

MSCs Modulate the Inflammatory Responses During Wound Healing

Once an acute wound occurs, the exposure of collagen initiates the intrinsic and extrinsic clotting cascades. Acute inflammation follows immediately, causing an increase in blood vessel leaking and local swelling. It is an inevitable consequence of injury, aiming for controlling bleeding, preventing infection, and triggering the repair process. But excessive inflammation will cause chronic wounds even the failure of wound healing. Indeed, altered inflammation in the early stage has been deemed essential to affect subsequent steps of the repair process that could influence cell proliferation, proper wound healing, and remodeling. In a bone marrow transplantation mouse model, green fluorescent protein (GFP) labeled endogenous MSCs can be found preferentially in areas of cutaneous injury, which are probably activated by early signals of pathological and inflammatory changes to initiate a cascade of wound healing.9 Available evidence shows that MSCs-based therapy is likely to reduce inflammation but not replace damaged cells. For instance, in treating acute and chronic liver injury, the administration of MSCs is a strategy widely used in the early stage to regulate organ inflammation in different tissue sites. Even for SARS-CoV-2 infection, MSCs intravenous infusion was suggested as a treatment for COVID-19 pneumonia to suppress pulmonary inflammatory storm.16

Major Effector Molecules Released by MSCs

Numerous studies have shed light on the immunomodulatory properties of MSCs in treating inflammatory diseases and found several factors and molecules derived from MSCs linked to this function. These include vascular endothelial growth factor C (VEGFC), transforming growth factor-β1 (TGF-β1), IL-6, nitric oxide (NO), indoleamine 2, 3-dioxygenase (IDO), tumor necrosis factor stimulated gene-6 (TSG6), prostaglandin E2, IL-1 receptor antagonist, IL-10, hepatocyte growth factor (HGF), platelet-derived growth factor (PDGF), insulin growth factor (IGF), stromal cell-derived factor-1 (SDF-1), and an antagonistic variant of the chemokine CCL2.13,17-19 In addition, MSCs can also secrete a large amount of MMPs, which could regulate the inflammation by regulating the activity of chemokines and mediate the phagocytic functions and immune responses of inflammatory cells through matricryptins.20,21 At least in principle, some of these regulations could be exerted by p63, an epithelial regulator able to impinge on several interleukins.22-24 The ability of MSCs to modulate the inflammatory response in wounds supports their favorable effect on the healing response. A severe burning rat model, which adopted the human umbilical cord-derived MSCs treatment, demonstrated a significantly decreased number of inflammatory cells and pro-inflammatory cytokines such as IL-1, IL-6, and TNF-α. Importantly, these wounds also exhibited a shorter recovery time and faster healing rate.25 MSCs can also lower the level of the pro-inflammatory cytokine and accelerate wound healing in the cornea undergoing chemical injury.26 TSG-6 is a crucial immunosuppressive factor conferring MSCs with prominent anti-inflammatory properties for treating myocardial infarction,27 peritonitis,28 acute lung injury,29 and corneal injury.30 The TSG-6 released from MSCs infused into wound margins contributed to the suppression of the release of TNF-α from activated macrophages, accelerating wound healing, and reducing tissue fibrosis in murine full-thickness skin wounds.31 Interestingly, TSG-6-modified MSCs effectively ameliorated pathological scarring, decreased inflammatory molecular secretion such as monocyte chemoattractant protein-1 (MCP-1), TNF-α, IL1β, and IL6, and attenuated collagen deposition in a mouse skin wound model. Notably, the pro-inflammatory cytokines were decreased.32

Plasticity of MSCs in Immunomodulation

It is important to note that MSCs can be “licensed” to exert their immunomodulatory effects after stimulation with interferon-γ (IFN-γ) in the presence of one (or more) other cytokines (s), including TNF, IL-1α, or IL-1β. But this exclusive feature also depends upon the kinds and concentrations of inflammatory mediators present in the tissue microenvironment. Low concentrations of cytokines were sufficient to upregulate chemokine secretion but were not enough to induce substantial expression of iNOS or IDO and ultimately led to greater inflammation.33 In addition, the types of cytokines present in the environment can further enhance or weaken the immunosuppressive effects of MSCs induced by TNF and IFN. For example, IL-17 present in the environment further strengthens the immunosuppressive ability of MSCs.34 Reversely, TGF-β and IL-10 result in the MSCs less immunosuppressive.35,36 Another factor that affects the immunoregulatory ability of MSCs is the recognition of microbial molecular patterns through Toll-like receptors (TLRs). For instance, TLR-4 activation induces a pro-inflammatory response, while TLR-3 activation results in an anti-inflammatory signal. Interestingly, TLRs have also been reported to play an important role in skin injury repair.37,38 Therefore, specific stimuli are crucial in provoking the immunomodulatory capacities of MSCs during MSC-based therapy for wound healing.

A growing number of studies on the crosstalk between inflammation and MSC-mediated immunosuppression strongly support the notion that the immunoregulatory function of MSCs is highly plastic. An impressive study in the mouse model of liver fibrosis has demonstrated the therapeutic roles of MSC treatment for the initial and progressive stages of liver fibrotic injury, indicating distinct effects of variable inflammatory stages of diseases with various kinds of cytokines.39 Also, concurrent administration of steroids, a widely used clinical immunosuppressor, abolished the therapeutic effects of MSCs on liver cirrhosis by reversing the immunosuppressive property of MSCs.40 Likewise, a similar outcome found in a GvHD mouse model, which was attributed to the effects of VEGFC, a novel immunomodulatory factor derived from MSCs.41 Those findings suggest that the appropriate inflammatory state of the disease for intervention is one of the most critical conditions for MSC therapy to be effective in wound healing.

The Interaction Between MSCs and Various Immune Cells

Upon injury, several inflammatory cell types,42 which bridge innate and adaptive immunity, have been found to play a critical role in wound healing.43-45 Especially during the inflammation phase, the infiltrated immune cells and inflammatory cytokines are in a dynamic change, which significantly affects the immunoregulation of MSCs. Correspondingly, several studies have demonstrated that MSCs also can exert immunomodulatory function by regulating the activation of various immune cells, including macrophages, neutrophils, natural killer cells, dendritic cells, T lymphocytes, and B lymphocytes46,47 (Fig. 1).

Figure 1.

Figure 1.

The immunoregulation of MSCs during wound healing. The inflammatory phase is one of the main steps for deciding on an ordinary or impaired wound healing course; this phase is necessary to clean bacteria, tissue debris, apoptotic cells, and clots from the wound. MSCs can exert immunomodulatory function by regulating various cells’ activation and operation of the innate and adaptive immune systems, including mast cells, neutrophils, macrophages, and T cells.

Neutrophils

Neutrophils, which are usually not observed in the normal skin, are produced in the bone marrow from promyelocytes and are recruited as “first responders” from blood in response to “find me” signals, including DAMPs, hydrogen peroxide, lipid mediators, and chemokines released from regions of injury or infection.48 Neutrophils reach the injury site immediately after injury and peak after 24 h. These sites combat infectious threats by releasing toxic granules, producing an oxidative burst, initiating phagocytosis, and generating neutrophil extracellular traps (NETs).42 Increasing evidence suggests that the injected MSCs promote wound healing by regulating neutrophil activation and NETs formation, which is associated with protection from infection.49,50

However, neutrophils are not just occurred in response to infection. It has been reported that they also penetrate the injury site and perform essential repair functions in a fully repairing sterile thermal hepatic injury.51 We also reported that the lung-resident MSCs promote neutrophil recruitment and the formation of NETs, which facilitate cancer cell metastasis to the lungs.52 The above studies indicate that neutrophils have alternative functions in addition to their classic bactericidal functions. Interestingly, apoptotic neutrophils significantly induce a repair macrophage phenotype that involves the induced production of TGF-β and IL-10.53 Moreover, it is reported that MSCs-conditioned medium is responsible for tissue repair by inducing neutrophil apoptosis in the lipopolysaccharide (LPS)-induced acute lung injury (ALI) mouse model.54 However, it does not always occur the same way. A growing number of studies have demonstrated excessive neutrophil infiltration during this phase may increase inflammation and impede wound healing. NETs could destroy wound structures, impair angiogenesis in the wound area, and also affect the number or functions of cells participating in wound repair, eventually leading to delayed wound healing.55 In the ALI mouse model, MSCs or MSCs conditioned medium treatment significantly reduced the number of infiltrated neutrophils and alleviated the progression.56,57 Moreover, we found that human adipose-derived MSCs (ADSCs) could effectively promote the clearance of neutrophils, reduce the NETs formation during the granulation stage, inhibit neovascularization, and reduce the opacification of the ethanol-injured cornea.58 The importance of neutrophils in wound healing was clearly demonstrated in mice genetically deficient for CXCR2- a chemokine receptor crucial for neutrophils recruitment to wound sites. These mice exhibited delayed re-epithelialization of skin wounds compared to the WT mice.59 However, in another study, the author found that the re-epithelialization was accelerated when the neutrophils were depleted after the mice were injected with anti-Gr1 antibody.60

Macrophages

Macrophages, which arrive at the injured site followed by neutrophils, are essential to routine wound healing and tissue regeneration. The wound-accumulated macrophages originated from monocytes-derived macrophages or expanded tissue-resident macrophages.42 Significantly, in a murine wound model, depletion of macrophages shows delayed wound closure.61,62 Conversely, increasing the number of monocytes or macrophages in the wound can significantly accelerate both normal and diabetic murine wound healing.63 In the early stages of wound healing, macrophages are microbicidal and pro-inflammatory, expressing TNF-α and IL-6 and IL-1, referred to as M1. Macrophage depletion in this phase has demonstrated roles in the formation of vascularized granulation tissue, epithelialization, and diminishment of scar formation.64,65 With the resolution of inflammation, the transition of macrophages from M1 to anti-inflammatory phenotype (M2) is critical for the subsequent proliferation stage and, eventually, tissue regeneration. Depletion of macrophage during the proliferation stage led to hemorrhage and a failure of wound closure.64

The functionally converted macrophages further enhance the migration and proliferation abilities of fibroblasts to facilitate the wound-healing process.66 Several early studies have demonstrated that apoptotic bodies or exosomes derived from MSCs could also promote cutaneous wound healing by triggering macrophages to polarize towards the M2 phenotype. Exosomes derived from MSCs are also reported to alter the M2 polarization and enhance wound healing.67 The same phenomenon was observed in a full-thickness wound experiment. It was found that miRNAs Let-7b in exosomes from LPS-pretreated MSCs regulate macrophage M2 polarization via the TLR4/NF-κB/STAT3/AKT signaling cascade.68 In addition, exosomes derived from melatonin-stimulated MSCs have been applied to promote diabetic wound healing by regulating macrophage M2 polarization and increasing the expression of IL-10 and Arg-1 via targeting the PTEN/AKT pathway.69 Moreover, IL-6 and GM-CSF produced by MSCs are reported to significantly elicit the M2 polarization process of macrophages and contribute to a marked acceleration of wound healing.70 To develop technologies that can be used for clinical treatment, CCR2-engineered MSCs were created to induce a regenerative immune microenvironment for tissue repair by modulating the inflammatory properties of macrophages.60 Our studies have shown that hypoxia-conditioned MSCs allow macrophages to acquire an anti-inflammatory phenotype by producing IGF-2, thereby alleviating experimental autoimmune encephalomyelitis (EAE).71 Mechanistically, nuclear translocation of ligated IGF2R complexed with GSK3 can induce proton rechanneling and promote oxidative phosphorylation (OXPHOS) in maturing macrophages.72 These studies provide novel strategies for the clinical use of MSCs in wound healing by regulating macrophage polarization.

Mast Cells

The mast cell is a type of granulocyte derived from the myeloid stem cell, which is widely distributed around the microvessels under the skin.73 These cells represent up to 8% of the total nucleated cells within the dermis and their cutaneous versions are localized adjacent to the epidermis and the subdermal vasculature and nerves. At the time of injury, mast cells become activated and secret a large number of pre-existed mediators in the granules, including vasoactive amines (eg, histamine, serotonin), lipid mediators (eg, LTB4, PGE2), cytokines (eg, IL-1β, IL4, IL6, TNF-α), and growth factors (eg, FGF-2, PDGF, VEGF). These mediators have been investigated during various stages of wound healing, including inflammation, proliferation, and remodeling, with a number of contradictory findings in the wound healing outcomes in mast cell-deficient mice using different skin injury mouse models (WBB6F1-KitW/KitW-v VS Mcpt5Cre/DTR mice).74-76 It has been proved that mast cells are mechanoresponsive and able to modulate the recruitment of neutrophils into injury sites using mast cell-deficient mice.76 In addition to mediating the early inflammatory response, the mast cells also directly contribute to skin fibrosis and scar formation-based wound contraction by activating fibroblast proliferation and collagen synthesis.77,78 On the other hand, it was observed that these cells could stimulate keratinocyte proliferation and re-epithelialization to functionally facilitate wound contraction and skin regeneration by generating histamine,79 and even inhibit fibrosis by suppressing the differentiation of endogenous mesenchymal stem cells to myofibroblasts.80 A balance between 2 distinct patterns of repair engaged by mast cells is likely to determine the fate of wound repair. Accordingly, increasing studies investigated the potential of using MSCs to regulate mast cells for wound repair based on the biological properties of these cells. In a murine atopic dermatitis model, MSC administration significantly inhibited mast cell degranulation and alleviated inflammation by producing PGE2 and TGF-β1.81 Of note, numerous studies are focused on the effects of MSC exosome components on mast cells. MSC exosomes have been shown to inhibit the increase in the number of dermal mast cells in TLR7 agonist-treated mice.82 Likewise, the culture medium from TNF-α-stimulated MSCs could reduce mast cell activation and histamine release through a COX2-dependent mechanism and attenuate experimental allergic conjunctivitis.83 Although regulatory effects have been identified in animal studies, further research is needed to focus on the key molecular mechanisms by which human MSCs regulate mast cell function for clinical precision therapy.

T cells

T cells are one of the important types of the immune system and play a central role in the adaptive immune response, drawn by chemokines produced in the wound, such as CCL9 and CCL10, are the final immune cells to infiltrate into the wound.84 Since the immune system is intimately involved in wound healing, extensive work with innovative tools has examined the impact of T cells and their cytokines on the final wound outcome. T cell-mediated immune responses are dynamic in wound healing. They are recruited in smaller numbers at the late stages with a Th1 response in the acute inflammatory phase, which is primarily driven by interferon-gamma (IFN-γ). As a proinflammatory cytokine, IFN-γ is able to aid in the polarization of macrophages to the M1 phenotype. When the inflammatory phase progresses, it is shifted into a Th2 response as T cells are progressively polarized. These cells produce IL-4, IL-5, IL-10, and IL-13, which, although anti-inflammatory, also have significant profibrotic effects. Murine studies support this observation and demonstrate that the interleukins, IL-4, IL-5, and IL-13, secreted from the Th2 cells resulted in scar formation.85,86 Consistently, impaired wound healing, characterized by excessive inflammation, is associated with increases in Th2 cell-derived TNF-α level. Overlapping with the Th1 and Th2 immune responses, the immune responses driven by Th17 cells result in re-epithelialization of the wound as well as hair follicle neogenesis through neutrophil recruitment. In addition, regulatory T cells (Treg cells) were found to promote tissue homeostasis and regeneration following tissue damage via restraining excessive inflammation. Strikingly, MSCs have been acknowledged for their ability to regulate T-cell immune responses and are therefore widely used for the treatment of inflammatory diseases, including refractory wounds. MSCs broadly suppress T-cell activation and proliferation in vitro via a plethora of soluble and cell contact-dependent mediators. These mediators may act directly upon CD4+T cells and CD8+T cells or indirectly via modulation of antigen-presenting cells and other accessory cells. The multiple effects of MSCs on cellular immunity may reflect their diverse influences on the different T-cell effector subpopulations and their capacity to specifically protect or induce Treg populations. For instance, the CCR2 over-expressed MSCs were reported could promote the accumulation of Treg cells in diabetic wounds and accelerate wound closure.87 MSCs also reduce collagen deposition13 and attenuate scar formation.32,88 It’s demonstrated that MSCs usually perform their therapeutic function by balancing pro-inflammatory and anti-inflammatory responses and directly or indirectly regulating disease-associated T-cell subsets (Th1, Th2, Th17, Treg).89 Although the inflammatory pathological process of wounds and the immunomodulatory properties of MSCs are clearly characterized, the specific mechanisms of interaction and regulatory targets between MSCs and T-cell subsets remain to be investigated for the clinical use of MSCs in the treatment of wounds in order to optimize the period and route of cell delivery.

In skin, one specific type of T cell that is important in skin wound healing is γδ T cells. They are known to recruit immune cells, stimulate angiogenesis, and activate keratinocytes.90 In addition, skin TRMs (tissue-resident memory T cells) are another unique subset of T cells that reside in the skin and provide long-lasting immune protection against re-infection by a pathogen. They are critical for local immunity in the skin and have been shown to play a key role in wound healing by modulating the immune response and promoting tissue repair. However, few articles focus on the impact of MSCs-based therapy on them in wound healing treatment.

Cancer and Wound Healing

Cancer, regarded as “non-healing wounds,”91 is believed to take advantage of the pro-regenerative ability of host cells to facilitate local cancer growth, resistance to therapy, and metastases to remote organs.92-94 It shares the same process with routine wound healing, including inflammation, tissue proliferation, and remodeling. However, the wound healing response of tumorigenesis possesses one or multiple prolonged, uncompleted phases, and tumors can therefore be considered as an “over-healing wound.”95 MSCs have recently been studied for their role in the tumor microenvironment. First, the tumor-associated MSCs (TA-MSCs) are involved in the pathological inflammatory process of tumors. It has been found that TA-MSCs are derived from normal resident MSCs altered by the tumor microenvironment. These cells are able to recruit monocytes and neutrophils to the tumor by secreting chemokines and inhibit the function of T cells and NK cells, thus maintaining tumor growth.96-99 Secondly, increasing evidence demonstrated that MSCs could promote an immunosuppressive environment that ultimately favors tumor progression and metastasis by inducing differentiation of leukocytes into immunosuppressive myeloid-derived suppressor cells (MDSCs)100 and increasing the proportion of Treg cells.101 Of note, we recently reported that the C3 secreted by lung resident MSCs could lead to pulmonarymetastasis of tumors by inducing NETs formation,52 which has been proved to act as a “trap” for circulatingtumorcells in previous studies.102 These results shed light on the mechanisms involved in the promotion of tumor progression by MSCs from an immunological perspective.

The Application of MSC Therapy on Different Pathological Wounds

Skin covers the surface of the human body, serving as a protective barrier against pathogens, this largest organ of the human body plays a vital role in maintaining the homeostasis of the body. In addition to trauma and burns, vascular insufficiency, diabetes mellitus, local pressure effects and systemic factors such as poor nutritional condition, infection as well as altered immunological status can lead to refractory wounds. Many studies have been performed on the therapeutic potential of MSCs for different chronic refractory wounds, including burn wounds, diabetic ulcers, and pressure sores (Table 1).

Table 1.

Outcome assessment parameters from clinical trials.

Study Wound healing Patients Cell type Cell number Cell source Cell administration Outcome
Rasulov103 Thermal burn Female, 45 years, 1 BM-MSC 20-30 × 103cells/cm2 Allogenic BM-MSC suspension was applied to burn wounds and the first skin graft transplantation was carried out in 4 days. Additional transplantations of MSCs onto wounds and immediately made. Proved safety, improved new vessel, and granulation formation and reduced coarse cicatrices.
Eduardo Mansilla104 Thermal burn Male, 26 years, 1 BM-MSC 106/cm2 Allogenic Spraying the MSCs onto the burn wound surface after deep escharotomy using 2 sterile conical tubes. Then the surface was dressed with a sterile polymeric transparent film and keep changing the film. Good blood supply without infection, better granulation tissue formation, and better re-epithelialization.
Xu105 Thermal burn Male, 19 years, 1 BM-MSC 1 × 104 cells/cm2 Autologous The autologous split-skin graft was spread over the dermal matrix sheet to cover the wound. No pain, infection or other side effects during wound healing and reduced skin contraction after 2 years.
Nikolaos Arkoulis106 Thermal burn Female, 42, 55 years, 2 Adipose stem MSCs 46 400 cells/cm2 Autologous Surface of burn + skin-graft Positive
Wael Abo-Elkheir107 Thermal burn All, 15 to 50 years, 60 BM-MSC/UC-MSC Not mentioned Allogenic Two days after the surgical excision of deeply burned tissue, stem cells were injected into the burned area and the application was repeated in 10 days. Improved rate of healing and reduced hospitalization period in both BM-MSC and UC-MSC groups.
Vincent Falanga108 Lower extremity wounds 7 BM-MSC 2 × 106/cm2 Autologous Delivered topically in a fibrin spray, using the same methodology with respect to fibrin (no more than 2 mL). Positive
Nihar Ranjan Dash109 Lower extremity wounds 6 BM-MSC >1 × 106/cm2 Allogenic Along the edges of nonhealing ulcers Improved recovery of the ulcer area after transplantation.
Lafosse Aurore110 Lower extremity wounds Two male, 46 and 21 years, 1 female, 41 years Adipose stem MSCs 5.8 × 105 cells/cm2 Allogenic Covered with human acellular collagen matrix surface Positive
Qin111 Lower extremity wounds Not mentioned UC-MSC Between 5 and 10 × 106 cells/cm2 Allogenic Multiple points Positive
Xiangxia Zeng112 Lower extremity wounds Female, 57 years, 1 Placenta-derived Mesenchymal Stem Cells 1  ×  106 cells/cm2 Allogenic On topical MSCs hydrogel After 3 weeks, the wound had completely healed.
JJ Lataillade113 Radiation burn Male, 27 years, 3 BM-MSC 200 × 103 cells/cm2 Autologous Excision plus skin autograft and a local MSC therapy the healing was complete without any functional impairment.
Marc Benderitter114 Radiation burn 3 BM-MSC Totally 76 × 106 cells Autologous Surface of burn Perfect healing persist 3 years after the implantation.
Portas115 Radiation Burn Male, 66 years, 1 BM-MSC Not mentioned Allogenic Surface of burn Ulcer dimensions were reduced with local recovery and remission of signs and symptoms.
Damián García-Olmo116 Perianafistula 5 Adipose Mesenchymal Stem Cells 10-15 × 103 cells/cm2 Allogenic Injected into the tract walls Positive
Garcia-Olmo117 Perianal fistula 35 Adipose Mesenchymal Stem Cells 103 cells/cm2 Allogenic Injected into the tract walls Quality of life scores were higher in patients who received ASCs than in those who received fibrin glue alone.
Takafumi Yoshikawa118 Burn, lower extremity wounds, pressure ulcers All, average 64.8 years, 2 BM-MSC Not mentioned Allogenic Artificial dermis was cut to match the shape of skin wounds and the cultured marrow mesenchymal cells were combined in the artificial dermis. Confirmed skin regeneration and epithelialization in 2 to 4 weeks. One year after graft, the scarring degree was milder.
González Sarasúa119 Pressure ulcers 19 male, 3 female BM-MSC 60 million cells Autologous Injected into the ulcers Positive

Radiation Burns

Severe radiation burns continue to be a significant challenge for wound care therapies, as they are often insensitive or resistant to conventional treatments, which are very lengthy and difficult.120 Several studies have explored the therapeutic potential of MSCs for radiation burns. Alain Chapel et al investigated the potential of a combined infusion of autologous ex vivo expanded hematopoietic cells with MSCs for the treatment of multi-organ failure syndrome following irradiation in a non-human primate model in 2003. And then they reported remarkable outcome of a clinical trial in which bone marrow-derived MSCs were first used to treat radiation-induced proctitis in patients over-irradiated at Epinal Hospital. It was demonstrated that MSCs reduced pain and the number of episodes of diarrhea in three patients who were resistant to symptomatic treatments.121,122 In another trial, a decline of CD4+CD127+CD25+ T lymphocytes and an increase of CD4+CD25+ (potentially regulatory) T lymphocytes were observed in patients after receiving MSC treatments.122 Besides autologous BM-MSCs, allogenic BM-MSCs have also been used to treat a radiation burn, showing significant improvement in vasculature and skin quality with inflammatory remission. The activation of T and B cells was suppressed along with the decreased levels of C-reactive protein (CRP)115 after MSCs administration. These promising clinical findings also provide a wealth of prospective information for the use of MSCs in the treatment of severe radiation burns.

Lower Extremity Wounds

Lower extremity wounds are a prevalent and reoccurring type of complicated wound, becoming a big public healthcare issue with critical social and economic concern. They can be caused by one or a combination of problems including poor circulation, critical limb ischemia, diabetes, and other medical problems. Currently, nearly 1% of adults and 3.6% of older patients are reported to suffer from this chronic condition.123 Several investigators have reported their studies regarding the wound-healing potential therapeutic effects of MSCs on these refractory wounds.

Lafosse et al reported that implantation of adipose-derived MSCs seeded onto human acellular collagen matrix (biological dressing) represents a promising therapy for nonhealing wounds, offering improvement in dermal angiogenesis and remodeling. Significant macrophagic recruitment was also found in injured tissues after treatment without modification of lymphocyte infiltration.110

Another clinical study also showed a tremendous therapeutic effect of hUC-MSC on diabetic foot, and it demonstrated that the ratios of Treg/Th17 and Treg/Th1 cells were significantly increased after hUC-MSC transplantation. In contrast, the ratio of Th17/Th1 cells remained unchanged. CRP and TNF-α levels were also considerably reduced after hUC-MSC treatment.124 It indicated that hUC-MSC exerted an anti-inflammatory effect on diabetic foot ulcers, offering great potential for clinical application as an alternative to amputation.

Pressure Sores

Pressure sores are a significant cause of morbidity resulting from unrelieved pressure against the skin. Although many preventative and treatment modalities have been developed in recent years, the treatment of pressure ulcers remains frustrating and time-consuming. Considering that MSCs can promote skin wound healing, several clinical studies have been performed to determine the therapeutic potential of MSCs for pressure sores.

In 2008, Yoshikawa et al treated 11 patients suffering from pressure sores that had persisted for more than 3 months with autologous BM-MSCs impregnated on a collagen sponge. In 9 of the 11 patients, the ulcers almost healed; in the remaining 2 patients, the ulcers became smaller after treatment. Long-term follow-up of those surviving patients was favorable. Except for 2 patients with worsened nutritional states, the decubitus ulcers in the other 7 patients did not recur for at least 1-year post-treatment.118 We hypothesize that transplantation of BM-MSCs at a wounded site could reconstitute and modulate the dermal fibroblast population to facilitate the local cutaneous angiogenic formation, accommodate collagen deposition, and manipulate inflammation through anti-inflammatory effects or by suppressing the immune response.

Conclusion and Prospects

Though increasing investigations have shown the therapeutic effects of MSCs on wound healing, the underlying mechanisms remain largely undefined. The ability of MSCs to modulate the inflammatory response in wounds supports their favorable impact on the healing response. However, the inflammation also alters the MSC’s secretome, including a source of growth factors that promote the proliferation and differentiation of tissue cells. Therefore, a more in-depth understanding of the complicated link between MSCs and inflammatory conditions should be investigated in the future, which is essential for translation into clinical application.

In addition, many challenges need to be addressed before MSCs fulfill their therapeutic potential for clinical treatment. Allogeneic MSCs have the advantage of enabling complete cell characterization prior to treatment and increasing the possibility of rapidly applying cells to patients with low immunogenicity. However, some studies have described the generation of antibodies against and immune rejection of allogeneic donor MSCs.125 In addition, the MSCs immunoregulation capability is affected by the status of the disease and the age of donors.126,127 Moreover, the dosage of MSCs application is another issue that needs to be concerned, as chronic inflammation delay wound healing, however, a moderate inflammatory response is beneficial for wound repair. Therefore, the application of MSCs should control inflammation at an appropriate level to aid the wound healing transition from the prolonged inflammation phase to the proliferative phase. In order to maximize the effect of MSCs application in clinical treatment, all of these factors should be considered. In addition, the delivery route, and administration period should also be taken into consideration.

Moreover, one of the most significant strategies for optimizing MSCs-based therapy is to license MSCs and make them produce specific factors for treating various diseases. It has been reported that human umbilical cord-derived MSCs treated with TNF-α and IFN-γ could secret a large amount of VEGFC, which promotes angiogenesis, thus accelerating skin wound healing. Therefore, empowerment with inflammatory cytokines may have tremendous therapeutic potential in terms of enhancing the ability of MSCs to achieve an appropriate therapeutic effect in wound healing.

Contributor Information

Mengting Zhu, The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, People’s Republic of China; Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata,” Rome, Italy.

Lijuan Cao, The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, People’s Republic of China; Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata,” Rome, Italy.

Sonia Melino, Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata,” Rome, Italy.

Eleonora Candi, Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata,” Rome, Italy.

Ying Wang, CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Shanghai, People’s Republic of China.

Changshun Shao, The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, People’s Republic of China.

Gerry Melino, Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata,” Rome, Italy.

Yufang Shi, The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, People’s Republic of China.

Xiaodong Chen, Wuxi Sinotide New Drug Discovery Institutes, Huishan Economic and Technological Development Zone, Wuxi, Jiangsu, People’s Republic of China.

Funding

This study was supported by grants from the National Key R&D Program of China (2022YFA0807300), National Natural Science Foundation of China (81930085), Jiangsu Province International Joint Laboratory for Regenerative Medicine Fund, Suzhou Foreign Academician Workstation Fund (SWY202202). Wuxi Taihu Tanlent Top Talent Team, Jiangsu Province, China (2021).

Conflict of Interest

The authors declared no potential conflicts of interest.

Author Contributions

M.Z.: conception and design, writing—original draft preparation. L.C.: conception and design, writing—original draft preparation. S.M., E.C., Y.W., C.S.: conception and design. G.M.: conception and design, writing—review and editing. Y.S., X.C.: conception, design, supervision of study conduct and operations, writing—review and editing, final approval of manuscript.

Data Availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  • 1. Martin P, Nunan R.. Cellular and molecular mechanisms of repair in acute and chronic wound healing. Br J Dermatol. 2015;173(2):370-378. 10.1111/bjd.13954 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. 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. 10.1186/s13287-016-0303-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. 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(2):136-143. 10.5021/ad.2012.24.2.136 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Mathew-Steiner SS, Roy S, Sen CK.. Collagen in wound healing. Bioengineering (Basel). 2021;8(5):63. 10.3390/bioengineering8050063 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Leavitt T, Hu MS, Marshall CD, et al. Scarless wound healing: finding the right cells and signals. Cell Tissue Res. 2016;365(3):483-493. 10.1007/s00441-016-2424-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Velnar T, Bailey T, Smrkolj V.. The wound healing process: an overview of the cellular and molecular mechanisms. J Int Med Res. 2009;37(5):1528-1542. 10.1177/147323000903700531 [DOI] [PubMed] [Google Scholar]
  • 7. Baron JM, Glatz M, Proksch E.. Optimal support of wound healing: new insights. Dermatology. 2020;236(6):593-600. 10.1159/000505291 [DOI] [PubMed] [Google Scholar]
  • 8. Sasaki M, Abe R, Fujita Y, et al. Mesenchymal stem cells are recruited into wounded skin and contribute to wound repair by transdifferentiation into multiple skin cell type. J Immunol. 2008;180(4):2581-2587. 10.4049/jimmunol.180.4.2581 [DOI] [PubMed] [Google Scholar]
  • 9. Ishii G, Sangai T, Sugiyama K, et al. In vivo characterization of bone marrow-derived fibroblasts recruited into fibrotic lesions. Stem Cells. 2005;23(5):699-706. 10.1634/stemcells.2004-0183 [DOI] [PubMed] [Google Scholar]
  • 10. Deng W, Han Q, Liao L, et al. Engrafted bone marrow-derived flk-(1+) mesenchymal stem cells regenerate skin tissue. Tissue Eng. 2005;11(1-2):110-119. 10.1089/ten.2005.11.110 [DOI] [PubMed] [Google Scholar]
  • 11. Mantovani A, Barajon I, Garlanda C.. IL-1 and IL-1 regulatory pathways in cancer progression and therapy. Immunol Rev. 2018;281(1):57-61. 10.1111/imr.12614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Botto S, Streblow DN, DeFilippis V, et al. IL-6 in human cytomegalovirus secretome promotes angiogenesis and survival of endothelial cells through the stimulation of survivin. Blood. 2011;117(1):352-361. 10.1182/blood-2010-06-291245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Zhu M, Chu Y, Shang Q, et al. Mesenchymal stromal cells pretreated with pro-inflammatory cytokines promote skin wound healing through VEGFC-mediated angiogenesis. Stem Cells Transl Med. 2020;9(10):1218-1232. 10.1002/sctm.19-0241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Schweizer R, Kamat P, Schweizer D, et al. Bone marrow-derived mesenchymal stromal cells improve vascular regeneration and reduce leukocyte-endothelium activation in critical ischemic murine skin in a dose-dependent manner. Cytotherapy. 2014;16(10):1345-1360. 10.1016/j.jcyt.2014.05.008 [DOI] [PubMed] [Google Scholar]
  • 15. Schlosser S, Dennler C, Schweizer R, et al. Paracrine effects of mesenchymal stem cells enhance vascular regeneration in ischemic murine skin. Microvasc Res. 2012;83(3):267-275. 10.1016/j.mvr.2012.02.011 [DOI] [PubMed] [Google Scholar]
  • 16. Verkhratsky A, Li Q, Melino S, Melino G, Shi Y.. Can COVID-19 pandemic boost the epidemic of neurodegenerative diseases? Biol Direct. 2020;15(1):28. 10.1186/s13062-020-00282-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Shi Y, Hu G, Su J, et al. Mesenchymal stem cells: a new strategy for immunosuppression and tissue repair. Cell Res. 2010;20(5):510-518. 10.1038/cr.2010.44 [DOI] [PubMed] [Google Scholar]
  • 18. Qiu X, Liu J, Zheng C, et al. Exosomes released from educated mesenchymal stem cells accelerate cutaneous wound healing via promoting angiogenesis. Cell Prolif. 2020;53(8):e12830. 10.1111/cpr.12830 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Uccelli A, Moretta L, Pistoia V.. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008;8(9):726-736. 10.1038/nri2395 [DOI] [PubMed] [Google Scholar]
  • 20. Struyf S, Proost P, Vandercappellen J, et al. Synergistic up-regulation of MCP-2/CCL8 activity is counteracted by chemokine cleavage, limiting its inflammatory and anti-tumoral effects. Eur J Immunol. 2009;39(3):843-857. 10.1002/eji.200838660 [DOI] [PubMed] [Google Scholar]
  • 21. Adair-Kirk TL, Senior RM.. Fragments of extracellular matrix as mediators of inflammation. Int J Biochem Cell Biol. 2008;40(6-7):1101-1110. 10.1016/j.biocel.2007.12.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Pecorari R, Bernassola F, Melino G, Candi E.. Distinct interactors define the p63 transcriptional signature in epithelial development or cancer. Biochem J. 2022;479(12):1375-1392. 10.1042/BCJ20210737 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Brauweiler AM, Leung DYM, Goleva E.. The transcription factor p63 is a direct effector of IL-4- and IL-13-mediated repression of keratinocyte differentiation. J Invest Dermatol. 2021;141(4):770-778. 10.1016/j.jid.2020.09.009 [DOI] [PubMed] [Google Scholar]
  • 24. Rizzo JM, Oyelakin A, Min S, et al. DeltaNp63 regulates IL-33 and IL-31 signaling in atopic dermatitis. Cell Death Differ. 2016;23(6):1073-1085. 10.1038/cdd.2015.162 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Liu L, Yu Y, Hou Y, et al. Human umbilical cord mesenchymal stem cells transplantation promotes cutaneous wound healing of severe burned rats. PLoS One. 2014;9(2):e88348. 10.1371/journal.pone.0088348 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Oh JY, Kim MK, Shin MS, et al. The anti-inflammatory and anti-angiogenic role of mesenchymal stem cells in corneal wound healing following chemical injury. Stem Cells. 2008;26(4):1047-1055. 10.1634/stemcells.2007-0737 [DOI] [PubMed] [Google Scholar]
  • 27. Lee RH, Pulin AA, Seo MJ, et al. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell. 2009;5(1):54-63. 10.1016/j.stem.2009.05.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Choi H, Lee RH, Bazhanov N, Oh JY, Prockop DJ.. Anti-inflammatory protein TSG-6 secreted by activated MSCs attenuates zymosan-induced mouse peritonitis by decreasing TLR2/NF-kappaB signaling in resident macrophages. Blood. 2011;118(2):330-338. 10.1182/blood-2010-12-327353 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Danchuk S, Ylostalo JH, Hossain F, et al. Human multipotent stromal cells attenuate lipopolysaccharide-induced acute lung injury in mice via secretion of tumor necrosis factor-alpha-induced protein 6. Stem Cell Res Ther. 2011;2(3):27. 10.1186/scrt68 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Oh JY, Roddy GW, Choi H, et al. Anti-inflammatory protein TSG-6 reduces inflammatory damage to the cornea following chemical and mechanical injury. Proc Natl Acad Sci U S A. 2010;107(39):16875-16880. 10.1073/pnas.1012451107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Qi Y, Jiang D, Sindrilaru A, et al. TSG-6 released from intradermally injected mesenchymal stem cells accelerates wound healing and reduces tissue fibrosis in murine full-thickness skin wounds. J Invest Dermatol. 2014;134(2):526-537. 10.1038/jid.2013.328 [DOI] [PubMed] [Google Scholar]
  • 32. Jiang L, Zhang Y, Liu T, et al. Exosomes derived from TSG-6 modified mesenchymal stromal cells attenuate scar formation during wound healing. Biochimie. 2020;177:40-49. 10.1016/j.biochi.2020.08.003 [DOI] [PubMed] [Google Scholar]
  • 33. Ren G, Zhang L, Zhao X, et al. Mesenchymal stem cell-mediated immunosuppression occurs via concerted action of chemokines and nitric oxide. Cell Stem Cell. 2008;2(2):141-150. 10.1016/j.stem.2007.11.014 [DOI] [PubMed] [Google Scholar]
  • 34. Han X, Yang Q, Lin L, et al. Interleukin-17 enhances immunosuppression by mesenchymal stem cells. Cell Death Differ. 2014;21(11):1758-1768. 10.1038/cdd.2014.85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Xu C, Yu P, Han X, et al. TGF-beta promotes immune responses in the presence of mesenchymal stem cells. J Immunol. 2014;192(1):103-109. 10.4049/jimmunol.1302164 [DOI] [PubMed] [Google Scholar]
  • 36. Renner P, Eggenhofer E, Rosenauer A, et al. Mesenchymal stem cells require a sufficient, ongoing immune response to exert their immunosuppressive function. Transplant Proc. 2009;41(6):2607-2611. 10.1016/j.transproceed.2009.06.119 [DOI] [PubMed] [Google Scholar]
  • 37. Chen L, Guo S, Ranzer MJ, DiPietro LA.. Toll-like receptor 4 has an essential role in early skin wound healing. J Invest Dermatol. 2013;133(1):258-267. 10.1038/jid.2012.267 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Nelson AM, Reddy SK, Ratliff TS, et al. dsRNA released by tissue damage activates TLR3 to drive skin regeneration. Cell Stem Cell. 2015;17(2):139-151. 10.1016/j.stem.2015.07.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Yang X, Han Z-P, Zhang S-S, et al. Chronic restraint stress decreases the repair potential from mesenchymal stem cells on liver injury by inhibiting TGF-beta1 generation. Cell Death Dis. 2014;5(6):e1308. 10.1038/cddis.2014.257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Chen X, Gan Y, Li W, et al. The interaction between mesenchymal stem cells and steroids during inflammation. Cell Death Dis. 2014;5(1):e1009. 10.1038/cddis.2013.537 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Gan Y, Zhang T, Chen X, et al. Steroids enable mesenchymal stromal cells to promote CD8(+) T cell proliferation via VEGF-C. Adv Sci (Weinh). 2021;8(12):2003712. 10.1002/advs.202003712 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Rodrigues M, Kosaric N, Bonham CA, Gurtner G. C.. Wound healing: a cellular perspective. Physiol Rev. 2019;99(1):665-706. 10.1152/physrev.00067.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Leibovich SJ, Ross R.. The role of the macrophage in wound repair. A study with hydrocortisone and antimacrophage serum. Am J Pathol. 1975;78(1):71-100. [PMC free article] [PubMed] [Google Scholar]
  • 44. Simpson DM, Ross R.. The neutrophilic leukocyte in wound repair a study with antineutrophil serum. J Clin Invest. 1972;51(8):2009-2023. 10.1172/JCI107007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Guillamat-Prats R. The role of MSC in wound healing, scarring and regeneration. Cells. 2021;10(7):1729. 10.3390/cells10071729 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Le Blanc K, Mougiakakos D.. Multipotent mesenchymal stromal cells and the innate immune system. Nat Rev Immunol. 2012;12(5):383-396. 10.1038/nri3209 [DOI] [PubMed] [Google Scholar]
  • 47. Chiesa S, Morbelli S, Morando S, et al. Mesenchymal stem cells impair in vivo T-cell priming by dendritic cells. Proc Natl Acad Sci U S A. 2011;108(42):17384-17389. 10.1073/pnas.1103650108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Su Y, Richmond A.. Chemokine regulation of neutrophil infiltration of skin wounds. Adv Wound Care (New Rochelle).. 2015;4(11):631-640. 10.1089/wound.2014.0559 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Munir S, Basu A, Maity P, et al. TLR4-dependent shaping of the wound site by MSCs accelerates wound healing. EMBO Rep. 2020;21(5):e48777. 10.15252/embr.201948777 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Chow L, Johnson V, Impastato R, et al. Antibacterial activity of human mesenchymal stem cells mediated directly by constitutively secreted factors and indirectly by activation of innate immune effector cells. Stem Cells Transl Med. 2020;9(2):235-249. 10.1002/sctm.19-0092 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Wang J, Hossain M, Thanabalasuriar A, et al. Visualizing the function and fate of neutrophils in sterile injury and repair. Science. 2017;358(6359):111-116. 10.1126/science.aam9690 [DOI] [PubMed] [Google Scholar]
  • 52. Zheng Z, Li Y-N, Jia S, et al. Lung mesenchymal stromal cells influenced by Th2 cytokines mobilize neutrophils and facilitate metastasis by producing complement C3. Nat Commun. 2021;12(1):6202. 10.1038/s41467-021-26460-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Elliott MR, Koster KM, Murphy PS.. Efferocytosis signaling in the regulation of macrophage inflammatory responses. J Immunol. 2017;198(4):1387-1394. 10.4049/jimmunol.1601520 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Su VY, Lin CS, Hung SC, Yang KY.. Mesenchymal stem cell-conditioned medium induces neutrophil apoptosis associated with inhibition of the NF-kappaB pathway in endotoxin-induced acute lung injury. Int J Mol Sci. 2019;20(9):2208. 10.3390/ijms20092208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Zhu S, Yu Y, Ren Y, et al. The emerging roles of neutrophil extracellular traps in wound healing. Cell Death Dis. 2021;12(11):984. 10.1038/s41419-021-04294-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Wang G, Cao K, Liu K, et al. Kynurenic acid, an IDO metabolite, controls TSG-6-mediated immunosuppression of human mesenchymal stem cells. Cell Death Differ. 2018;25(7):1209-1223. 10.1038/s41418-017-0006-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Ionescu L, Byrne RN, van Haaften T, et al. Stem cell conditioned medium improves acute lung injury in mice: in vivo evidence for stem cell paracrine action. Am J Physiol Lung Cell Mol Physiol. 2012;303(11):L967-L977. 10.1152/ajplung.00144.2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Shang Q, Chu Y, Li Y, et al. Adipose-derived mesenchymal stromal cells promote corneal wound healing by accelerating the clearance of neutrophils in cornea. Cell Death Dis. 2020;11(8):707. 10.1038/s41419-020-02914-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Devalaraja RM, Nanney L B, Du J, et al. Delayed wound healing in CXCR2 knockout mice. J Invest Dermatol. 2000;115(2):234-244. 10.1046/j.1523-1747.2000.00034.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Dovi J. V, He LK, DiPietro LA.. Accelerated wound closure in neutrophil-depleted mice. J Leukoc Biol. 2003;73(4):448-455. 10.1189/jlb.0802406 [DOI] [PubMed] [Google Scholar]
  • 61. Goren I, Allmann N, Yogev N, et al. A transgenic mouse model of inducible macrophage depletion: effects of diphtheria toxin-driven lysozyme M-specific cell lineage ablation on wound inflammatory, angiogenic, and contractive processes. Am J Pathol. 2009;175(1):132-147. 10.2353/ajpath.2009.081002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Mirza R, DiPietro LA, Koh TJ.. Selective and specific macrophage ablation is detrimental to wound healing in mice. Am J Pathol. 2009;175(6):2454-2462. 10.2353/ajpath.2009.090248 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Hu MS, Walmsley GG, Barnes LA, et al. Delivery of monocyte lineage cells in a biomimetic scaffold enhances tissue repair. JCI Insight. 2017;2(19):e96260. 10.1172/jci.insight.96260 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Lucas T, Waisman A, Ranjan R, et al. Differential roles of macrophages in diverse phases of skin repair. J Immunol. 2010;184(7):3964-3977. 10.4049/jimmunol.0903356 [DOI] [PubMed] [Google Scholar]
  • 65. Zhu Z, Ding J, Ma Z, Iwashina T, Tredget EE.. Systemic depletion of macrophages in the subacute phase of wound healing reduces hypertrophic scar formation. Wound Repair Regen. 2016;24(4):644-656. 10.1111/wrr.12442 [DOI] [PubMed] [Google Scholar]
  • 66. Liu J, Qiu X, Lv Y, et al. Apoptotic bodies derived from mesenchymal stem cells promote cutaneous wound healing via regulating the functions of macrophages. Stem Cell Res Ther. 2020;11(1):507. 10.1186/s13287-020-02014-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. He X, Dong Z, Cao Y, et al. MSC-derived exosome promotes m2 polarization and enhances cutaneous wound healing. Stem Cells Int. 2019;2019:7132708. 10.1155/2019/7132708 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Ti D, Hao H, Tong C, et al. LPS-preconditioned mesenchymal stromal cells modify macrophage polarization for resolution of chronic inflammation via exosome-shuttled let-7b. J Transl Med. 2015;13:308. 10.1186/s12967-015-0642-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Liu W, Yu M, Xie D, et al. Melatonin-stimulated MSC-derived exosomes improve diabetic wound healing through regulating macrophage M1 and M2 polarization by targeting the PTEN/AKT pathway. Stem Cell Res Ther 2020;11(1):259. 10.1186/s13287-020-01756-x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Zhang QZ, Su W-R, Shi S-H, et al. Human gingiva-derived mesenchymal stem cells elicit polarization of m2 macrophages and enhance cutaneous wound healing. Stem Cells. 2010;28(10):1856-1868. 10.1002/stem.503 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Du L, Lin L, Li Q, et al. IGF-2 preprograms maturing macrophages to acquire oxidative phosphorylation-dependent anti-inflammatory properties. Cell Metab. 2019;29(6):1363-1375.e8. 10.1016/j.cmet.2019.01.006 [DOI] [PubMed] [Google Scholar]
  • 72. Wang X, et al. IGF2R-initiated proton rechanneling dictates an anti-inflammatory property in macrophages. Sci Adv. 2020;42:eabb7389. 10.1126/sciadv.abb7389 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Galli SJ, Grimbaldeston M, Tsai M.. Immunomodulatory mast cells: negative, as well as positive, regulators of immunity. Nat Rev Immunol. 2008;8(6):478-486. 10.1038/nri2327 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Shiota N, Nishikori Y, Kakizoe E, et al. Pathophysiological role of skin mast cells in wound healing after scald injury: study with mast cell-deficient W/W(V) mice. Int Arch Allergy Immunol. 2010;151(1):80-88. 10.1159/000232573 [DOI] [PubMed] [Google Scholar]
  • 75. Nauta A. C, Grova M, Montoro DT, et al. Evidence that mast cells are not required for healing of splinted cutaneous excisional wounds in mice. PLoS One. 2013;8(3):e59167. 10.1371/journal.pone.0059167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Egozi EI, Ferreira AM, Burns AL, Gamelli RL, Dipietro LA.. Mast cells modulate the inflammatory but not the proliferative response in healing wounds. Wound Repair Regen. 2003;11(1):46-54. 10.1046/j.1524-475x.2003.11108.x [DOI] [PubMed] [Google Scholar]
  • 77. Abe M, Kurosawa M, Ishikawa O, Miyachi Y.. Effect of mast cell-derived mediators and mast cell-related neutral proteases on human dermal fibroblast proliferation and type I collagen production. J Allergy Clin Immunol. 2000;106(1 Pt 2):S78-S84. 10.1067/mai.2000.106058 [DOI] [PubMed] [Google Scholar]
  • 78. Garbuzenko E, Nagler A, Pickholtz D, et al. Human mast cells stimulate fibroblast proliferation, collagen synthesis and lattice contraction: a direct role for mast cells in skin fibrosis. Clin Exp Allergy. 2002;32(2):237-246. 10.1046/j.1365-2222.2002.01293.x [DOI] [PubMed] [Google Scholar]
  • 79. Weller K, Foitzik K, Paus R, Syska W, Maurer M.. Mast cells are required for normal healing of skin wounds in mice. FASEB J. 2006;20(13):2366-2368. 10.1096/fj.06-5837fje [DOI] [PubMed] [Google Scholar]
  • 80. Nazari M, Ni Nathan C, Lüdke A, et al. Mast cells promote proliferation and migration and inhibit differentiation of mesenchymal stem cells through PDGF. J Mol Cell Cardiol. 2016;94:32-42. 10.1016/j.yjmcc.2016.03.007 [DOI] [PubMed] [Google Scholar]
  • 81. Kim HS, Yun J-W, Shin T-H, et al. Human umbilical cord blood mesenchymal stem cell-derived PGE2 and TGF-beta1 alleviate atopic dermatitis by reducing mast cell degranulation. Stem Cells. 2015;33(4):1254-1266. 10.1002/stem.1913 [DOI] [PubMed] [Google Scholar]
  • 82. Cho KA, Cha J-E, Kim J, et al. Mesenchymal stem cell-derived exosomes attenuate TLR7-mediated mast cell activation. Tissue Eng Regen Med. 2022;19(1):117-129. 10.1007/s13770-021-00395-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Su W, Wan Q, Huang J, et al. Culture medium from TNF-alpha-stimulated mesenchymal stem cells attenuates allergic conjunctivitis through multiple antiallergic mechanisms. J Allergy Clin Immunol. 2015;136(2):423-32.e8. 10.1016/j.jaci.2014.12.1926 [DOI] [PubMed] [Google Scholar]
  • 84. Balaji S, Watson Carey L, Ranjan R, et al. Chemokine involvement in fetal and adult wound healing. Adv Wound Care (New Rochelle). 2015;4(11):660-672. 10.1089/wound.2014.0564 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Ong CJ, Ip S, Teh SJ, et al. A role for T helper 2 cells in mediating skin fibrosis in tight-skin mice. Cell Immunol. 1999;196(1):60-68. 10.1006/cimm.1999.1537 [DOI] [PubMed] [Google Scholar]
  • 86. Wallace VA, Kondo S, Kono T, et al. A role for CD4+ T cells in the pathogenesis of skin fibrosis in tight skin mice. Eur J Immunol. 1994;24(6):1463-1466. 10.1002/eji.1830240634 [DOI] [PubMed] [Google Scholar]
  • 87. Kuang S, He F, Liu G, et al. CCR2-engineered mesenchymal stromal cells accelerate diabetic wound healing by restoring immunological homeostasis. Biomaterials. 2021;275:120963. 10.1016/j.biomaterials.2021.120963 [DOI] [PubMed] [Google Scholar]
  • 88. Hu CH, Tseng Y-W, Chiou C-Y, et al. Bone marrow concentrate-induced mesenchymal stem cell conditioned medium facilitates wound healing and prevents hypertrophic scar formation in a rabbit ear model. Stem Cell Res Ther. 2019;10(1):275. 10.1186/s13287-019-1383-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Duffy MM, Ritter T, Ceredig R, Griffin MD.. Mesenchymal stem cell effects on T-cell effector pathways. Stem Cell Res Ther. 2011;2(4):34. 10.1186/scrt75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Hu W, Shang R, Yang J, et al. Skin gammadelta T cells and their function in wound healing. Front Immunol. 2022;13:875076. 10.3389/fimmu.2022.875076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315(26):1650-1659. 10.1056/NEJM198612253152606 [DOI] [PubMed] [Google Scholar]
  • 92. Calvo F, Ranftl R, Hooper S, et al. Cdc42EP3/BORG2 and septin network enables mechano-transduction and the emergence of cancer-associated fibroblasts. Cell Rep. 2015;13(12):2699-2714. 10.1016/j.celrep.2015.11.052 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Krall JA, Reinhardt F, Mercury OA, et al. The systemic response to surgery triggers the outgrowth of distant immune-controlled tumors in mouse models of dormancy. Sci Transl Med. 2018;10(436):eaan3464. 10.1126/scitranslmed.aan3464 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Sundaram GM, Ismail HM, Bashir M, et al. EGF hijacks miR-198/FSTL1 wound-healing switch and steers a two-pronged pathway toward metastasis. J Exp Med. 2017;214(10):2889-2900. 10.1084/jem.20170354 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Schafer M, Werner S.. Cancer as an overhealing wound: an old hypothesis revisited. Nat Rev Mol Cell Biol. 2008;9(8):628-638. 10.1038/nrm2455 [DOI] [PubMed] [Google Scholar]
  • 96. Galleu A, Riffo-Vasquez Y, Trento C, et al. Apoptosis in mesenchymal stromal cells induces in vivo recipient-mediated immunomodulation. Sci Transl Med. 2017;9(416):eaam7828. 10.1126/scitranslmed.aam7828 [DOI] [PubMed] [Google Scholar]
  • 97. Galland S, Vuille J, Martin P, et al. Tumor-derived mesenchymal stem cells use distinct mechanisms to block the activity of natural killer cell subsets. Cell Rep. 2017;20(12):2891-2905. 10.1016/j.celrep.2017.08.089 [DOI] [PubMed] [Google Scholar]
  • 98. Guilloton F, Caron G, Ménard C, et al. Mesenchymal stromal cells orchestrate follicular lymphoma cell niche through the CCL2-dependent recruitment and polarization of monocytes. Blood. 2012;119(11):2556-2567. 10.1182/blood-2011-08-370908 [DOI] [PubMed] [Google Scholar]
  • 99. Ren G, Zhao X, Wang Y, et al. CCR2-dependent recruitment of macrophages by tumor-educated mesenchymal stromal cells promotes tumor development and is mimicked by TNFalpha. Cell Stem Cell. 2012;11(6):812-824. 10.1016/j.stem.2012.08.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Giallongo C, Romano A, Parrinello NL, et al. Mesenchymal stem cells (MSC) regulate activation of granulocyte-like myeloid derived suppressor cells (G-MDSC) in chronic myeloid leukemia patients. PLoS One. 2016;11(7):e0158392. 10.1371/journal.pone.0158392 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Razmkhah M, Jaberipour M, Erfani N, et al. Adipose derived stem cells (ASCs) isolated from breast cancer tissue express IL-4, IL-10 and TGF-beta1 and upregulate expression of regulatory molecules on T cells: do they protect breast cancer cells from the immune response? Cell Immunol. 2011;266(2):116-122. 10.1016/j.cellimm.2010.09.005 [DOI] [PubMed] [Google Scholar]
  • 102. Yang L, Liu Q, Zhang X, et al. DNA of neutrophil extracellular traps promotes cancer metastasis via CCDC25. Nature. 2020;583(7814):133-138. 10.1038/s41586-020-2394-6 [DOI] [PubMed] [Google Scholar]
  • 103. Rasulov MF, Vasilchenkov AV, Onishchenko NA, 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(1):141-144. 10.1007/s10517-005-0232-3 [DOI] [PubMed] [Google Scholar]
  • 104. Mansilla E, Marín GH, Berges M, et al. Cadaveric bone marrow mesenchymal stem cells: first experience treating a patient with large severe burns. Burns Trauma. 2015;3:17. 10.1186/s41038-015-0018-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Xu Y, Huang S, Fu X.. Autologous transplantation of bone marrow-derived mesenchymal stem cells: a promising therapeutic strategy for prevention of skin-graft contraction. Clin Exp Dermatol. 2012;37(5):497-500. 10.1111/j.1365-2230.2011.04260.x [DOI] [PubMed] [Google Scholar]
  • 106. Arkoulis N, Watson S, Weiler-Mithoff E.. Stem cell enriched dermal substitutes for the treatment of late burn contractures in patients with major burns. Burns. 2018;44(3):724-726. 10.1016/j.burns.2017.09.026 [DOI] [PubMed] [Google Scholar]
  • 107. Abo-Elkheir W, Hamza F, Elmofty AM, 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(3):23-35. [PMC free article] [PubMed] [Google Scholar]
  • 108. Falanga V, Iwamoto S, Chartier M, 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(6):1299-1312. 10.1089/ten.2006.0278 [DOI] [PubMed] [Google Scholar]
  • 109. Dash NR, Dash SN, Routray P, Mohapatra S, Mohapatra PC.. Targeting nonhealing ulcers of lower extremity in human through autologous bone marrow-derived mesenchymal stem cells. Rejuvenation Res. 2009;12(5):359-366. 10.1089/rej.2009.0872 [DOI] [PubMed] [Google Scholar]
  • 110. Lafosse A, Desmet C, Aouassar N, et al. Autologous adipose stromal cells seeded onto a human collagen matrix for dermal regeneration in chronic wounds: clinical proof of concept. Plast Reconstr Surg. 2015;136(2):279-295. 10.1097/PRS.0000000000001437 [DOI] [PubMed] [Google Scholar]
  • 111. Qin HL, Zhu XH, Zhang B, Zhou L, Wang WY.. Clinical evaluation of human umbilical cord mesenchymal stem cell transplantation after angioplasty for diabetic foot. Exp Clin Endocrinol Diabetes. 2016;124(8):497-503. 10.1055/s-0042-103684 [DOI] [PubMed] [Google Scholar]
  • 112. Zeng X, Tang Y, Hu K, et al. Three-week topical treatment with placenta-derived mesenchymal stem cells hydrogel in a patient with diabetic foot ulcer: A case report. Medicine (Baltim). 2017;96(51):e9212. 10.1097/MD.0000000000009212 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Lataillade JJ, Doucet C, Bey E, et al. New approach to radiation burn treatment by dosimetry-guided surgery combined with autologous mesenchymal stem cell therapy. Regen Med. 2007;2(5):785-794. 10.2217/17460751.2.5.785 [DOI] [PubMed] [Google Scholar]
  • 114. Benderitter M, Gourmelon P, Bey E, et al. New emerging concepts in the medical management of local radiation injury. Health Phys. 2010;98(6):851-857. 10.1097/HP.0b013e3181c9f79a [DOI] [PubMed] [Google Scholar]
  • 115. Portas M, Mansilla E, Drago H, et al. Use of human cadaveric mesenchymal stem cells for cell therapy of a chronic radiation-induced skin lesion: a case report. Radiat Prot Dosimetry. 2016;171(1):99-106. 10.1093/rpd/ncw206 [DOI] [PubMed] [Google Scholar]
  • 116. Garcia-Olmo D, García-Arranz M, Herreros D, et al. A phase I clinical trial of the treatment of Crohn’s fistula by adipose mesenchymal stem cell transplantation. Dis Colon Rectum. 2005;48(7):1416-1423. 10.1007/s10350-005-0052-6 [DOI] [PubMed] [Google Scholar]
  • 117. Garcia-Olmo D, Garcia-Arranz M, Herreros D.. Expanded adipose-derived stem cells for the treatment of complex perianal fistula including Crohn’s disease. Expert Opin Biol Ther. 2008;8(9):1417-1423. 10.1517/14712598.8.9.1417 [DOI] [PubMed] [Google Scholar]
  • 118. Yoshikawa T, Mitsuno H, Nonaka I, et al. Wound therapy by marrow mesenchymal cell transplantation. Plast Reconstr Surg. 2008;121(3):860-877. 10.1097/01.prs.0000299922.96006.24 [DOI] [PubMed] [Google Scholar]
  • 119. Sarasua JG, López SP, Viejo MA, et al. Treatment of pressure ulcers with autologous bone marrow nuclear cells in patients with spinal cord injury. J Spinal Cord Med. 2011;34(3):301-307. 10.1179/2045772311Y.0000000010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Olascoaga A, Vilar-Compte D, Poitevin-Chacon A, Contreras-Ruiz J.. Wound healing in radiated skin: pathophysiology and treatment options. Int Wound J. 2008;5(2):246-257. 10.1111/j.1742-481X.2008.00436.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Chapel A, Francois S, Douay L, Benderitter M, Voswinkel J.. Fifteen years of preclinical and clinical experiences about biotherapy treatment of lesions induced by accidental irradiation and radiotherapy. World J Stem Cells. 2013;5(3):68-72. 10.4252/wjsc.v5.i3.68 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Voswinkel J, Francois S, Simon J-M, et al. Use of mesenchymal stem cells (MSC) in chronic inflammatory fistulizing and fibrotic diseases: a comprehensive review. Clin Rev Allergy Immunol. 2013;45(2):180-192. 10.1007/s12016-012-8347-6 [DOI] [PubMed] [Google Scholar]
  • 123. Oien RF, Hakansson A, Ovhed I, Hansen BU.. Wound management for 287 patients with chronic leg ulcers demands 12 full-time nurses. Leg ulcer epidemiology and care in a well-defined population in southern Sweden. Scand J Prim Health Care. 2000;18(4):220-225. 10.1080/028134300448788 [DOI] [PubMed] [Google Scholar]
  • 124. Li XY, Zheng Z-H, Li X-Y, et al. Treatment of foot disease in patients with type 2 diabetes mellitus using human umbilical cord blood mesenchymal stem cells: response and correction of immunological anomalies. Curr Pharm Des. 2013;19(27):4893-4899. 10.2174/13816128113199990326 [DOI] [PubMed] [Google Scholar]
  • 125. Ankrum JA, Ong JF, Karp JM.. Mesenchymal stem cells: immune evasive, not immune privileged. Nat Biotechnol. 2014;32(3):252-260. 10.1038/nbt.2816 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Serena C, Keiran N, Ceperuelo-Mallafre V, et al. Obesity and type 2 diabetes alters the immune properties of human adipose derived stem cells. Stem Cells. 2016;34(10):2559-2573. 10.1002/stem.2429 [DOI] [PubMed] [Google Scholar]
  • 127. Serena C, Keiran N, Madeira A, et al. Crohn’s disease disturbs the immune properties of human adipose-derived stem cells related to inflammasome activation. Stem Cell Rep. 2017;9(4):1109-1123. 10.1016/j.stemcr.2017.07.014 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.


Articles from Stem Cells Translational Medicine are provided here courtesy of Oxford University Press

RESOURCES