Next Article in Journal
Interleukin-6 in Rheumatoid Arthritis
Next Article in Special Issue
Premature Activation of Immune Transcription Programs in Autoimmune-Predisposed Mouse Embryonic Stem Cells and Blastocysts
Previous Article in Journal
The Biological and Clinical Role of the Long Non-Coding RNA LOC642852 in Ovarian Carcinoma
Previous Article in Special Issue
The Roles of Immune Cells in the Pathogenesis of Fibrosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Cell Therapies to Improve Regeneration and Revascularization of Non-Healing Wounds

1
Cardiovascular Biology Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy
2
Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy
3
Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
*
Author to whom correspondence should be addressed.
These authors equally contributed as first authors.
Int. J. Mol. Sci. 2020, 21(15), 5235; https://doi.org/10.3390/ijms21155235
Submission received: 29 June 2020 / Revised: 20 July 2020 / Accepted: 21 July 2020 / Published: 23 July 2020
(This article belongs to the Special Issue Immune Cell Mediated Tissue Regeneration)

Abstract

:
With the increased prevalence of chronic diseases, non-healing wounds place a significant burden on the health system and the quality of life of affected patients. Non-healing wounds are full-thickness skin lesions that persist for months or years. While several factors contribute to their pathogenesis, all non-healing wounds consistently demonstrate inadequate vascularization, resulting in the poor supply of oxygen, nutrients, and growth factors at the level of the lesion. Most existing therapies rely on the use of dermal substitutes, which help the re-epithelialization of the lesion by mimicking a pro-regenerative extracellular matrix. However, in most patients, this approach is not efficient, as non-healing wounds principally affect individuals afflicted with vascular disorders, such as peripheral artery disease and/or diabetes. Over the last 25 years, innovative therapies have been proposed with the aim of fostering the regenerative potential of multiple immune cell types. This can be achieved by promoting cell mobilization into the circulation, their recruitment to the wound site, modulation of their local activity, or their direct injection into the wound. In this review, we summarize preclinical and clinical studies that have explored the potential of various populations of immune cells to promote skin regeneration in non-healing wounds and critically discuss the current limitations that prevent the adoption of these therapies in the clinics.

1. The Clinical Problem

The skin is the largest organ in mammals, which shields the organism from thermal, mechanical, and chemical damage; toxins; and microorganisms, serving as a protective barrier at the interface between the human body and the surrounding environment. Loss of skin integrity often results in physiologic imbalance and disability, or even death, with significant impact on the healthcare system [1,2]. Healing of any wound requires a coordinated response of multiple cell types, either residing in the various skin layers (epidermis and dermis) or recruited from the circulation [3]. This can be schematically divided in an early inflammatory phase, followed by a proliferative and regenerative phase, associated with massive extracellular matrix (ECM) remodeling [3,4].
This process is often compromised in common diseases that are highly prevalent in the aged population and include diabetes, obesity, and vascular disorders. As such, the incidence of chronic non-healing wounds is progressively increasing. Recent studies report a general prevalence of non-healing wounds of approximately 1–2%, similar to the one of heart failure [5,6,7].
Clinically, a chronic wound is defined as a full-thickness skin defect that fails to heal after 3 months of standard care [8]. Chronic wounds are generally classified into vascular ulcers (caused by either venous or arterial diseases), diabetic ulcers, and pressure ulcers. Additional, less common, etiologies include infections [9], immune diseases [10,11], traumas [12,13], large-area burns [14,15], and post-surgery complications [4,16,17,18]. Despite recent progresses in revascularization procedures and in the use of dermal and epidermal substitutes to improve healing, these ulcers often persist for more than one year and tend to recur in up to 70% of patients, significantly compromising their quality of life [19,20]. In 2020, the rate of both minor and major limb amputations is still very high, with reported incidences of 14–24% in patients with foot ulcers over the last decades [8].
Despite a heterogeneous etiology, most chronic wounds are characterized by persistent inflammation [20]. Excessive levels of inflammatory cytokines, proteolytic enzymes, and reactive oxygen species (ROS) damage both cellular and extracellular components, creating a proinflammatory feedback loop [20,21] Moreover, proteases degrade both growth factors and their receptors, resulting in cell cycle arrest and inhibition of proliferation and regeneration [22,23,24].
A common feature of all chronic wounds is insufficient vascularization, which represents a major impediment to healing and regeneration [25,26]. Appropriate supply of nutrients and growth factors, as well as maintenance of oxygen homeostasis, are essential to support cell proliferation, migration, and recruitment of immune cell populations [3,27], which play a fundamental role in both phases of the healing process.
The following chapters describe the existing models, commonly used to evaluate the efficacy of innovative therapies for skin regeneration. In particular, we consider various immune cell types, belonging to both innate and adaptive immunity, that contribute to wound healing and have been considered for their therapeutic potential in either preclinical or clinical studies. These therapeutic strategies rely on (i) immune cell mobilization into the circulation, (ii) cell recruitment and homing at the level of the skin, (iii) modulation of their pro-regenerative activity, and (iv) direct cell implantation to the wound site. Although we could not include all publications available in this expanding field, representative studies supporting each of these approaches have been cited. The most relevant approaches are schematically shown in Figure 1 and further details are provided in Table 1.

2. Animal Models of Wound Healing

Both small and large animal models have been developed to assess the regenerative potential of immune cells in wound healing [61]. Obvious differences in the anatomy and physiology of the skin in different species justify the use of multiple models, as no single one can reliably reproduce the clinical situation [61,62], and, more importantly, the comorbidities that are often responsible for chronic wounds [63,64].
Because chronic wounds usually occur in people affected by diabetes, ischemia, and mechanical pressure [8,65], these conditions are generally induced in animals, prior to the generation of an acute wound [66,67,68,69].
Mice are the most common species used for this purpose, as their housing is relatively cheap and affordable by most laboratories, they can be genetically manipulated for mechanistic investigation, and state-of-the-art protocols to induce co-morbidities (i.e., diabetes and ischemia) are available [70]. Yet, the mechanism of wound closure is very different in mice and humans. While murine wounds essentially close by contraction, in humans, wound healing relies on the formation of a new tissue, composed by tightly packed fibroblasts, keratinocytes, and endothelial and inflammatory cells (granulation tissue), which is progressively reabsorbed and replaced by re-epithelialization [71]. Preferential healing by contraction in rodents is mainly due to the presence of a thick layer of subcutaneous striated muscle tissue (named panniculus carnosus), absent in humans, and by the presence of active myofibroblasts. Several strategies have been developed to limit wound contraction in mice (i.e., the application of either a silicone splint around the wound or a dorsal skinfold chamber), allowing for the formation of granulation tissue and re-epithelialization, similar to what happens in human wound healing [71,72,73]. Combined with intravital microscopy, the dorsal skin fold chamber allows high-resolution real-time imaging of re-vascularization during skin regeneration [74]. While not yet used at a large extent, the possibility to monitor the formation of new blood vessels appears critical to develop new therapies improving wound healing and predict their efficacy in the clinics, as the whole skin regeneration requires an appropriate supply of nutrients and oxygen [75].
The evaluation of in vivo efficacy and safety of human cell therapies requires the use of immunocompromised mice, i.e., NSG (NOD scid gamma) mice, which lack B and T lymphocytes, as well as natural killer (NK) cells [44,76,77,78]. These mice are extensively used in the preclinical assessment of human cell-based therapies, as they allow the engraftment of human cells, but obviously do not recapitulate the complex immune response required for wound healing.
In conclusion, mice are very useful for the dissection of genetic, cellular, and molecular mechanisms of skin regeneration, but they cannot be considered the gold standard for therapeutic applications. Pigs share significant anatomical and physiological similarities to humans, in terms of epidermal and dermal thickness and structure, dense dermal collagen and elastin fibers, epidermal turnover time (approximately 30 days), pattern and structure of hair follicles, and biological response to growth factors [61,79,80]. Moreover, the mechanism of wound healing is very similar in pigs and humans, relying on the formation of granulation tissue, followed by re-epithelialization with minimal skin contraction [64]. Finally, the colonization of the porcine skin by immune cells is quite comparable to the human and different from the murine one, as also discussed later in more detail [79,80,81,82,83]. Because of these numerous similarities, pigs are considered the gold standard in the transition from preclinical to human studies. Yet, their use is importantly limited by high costs, absence of transgenic models for cell tracing, and a need for both sophisticated instrumentation and skilled personnel [64,79].

3. Neutrophils

Neutrophils are part of the innate immune system; they do not physiologically reside in the skin, but they are rapidly recruited to the wound site after injury [21,84]. They produce antimicrobial substances and proteases, most of which are contained in their cytoplasmic granules. They also generate cytokines and growth factors to recruit other components of the inflammatory system and support angiogenesis, keratinocyte, and fibroblast proliferation. Upon activation, neutrophils release proteins and decondensed chromatin that together form extracellular fibers able to trap bacteria. These neutrophil extracellular traps (NETs) degrade virulence factors and kill bacterial cells [85]. While their beneficial role is exerted during the early inflammatory phase of wound healing, their prolonged persistence caused by extensive damage and/or ongoing microbial contamination can inhibit the late reparative phase due to continuous release of proteases that degrade the ECM [86]. Thus, inhibition of their persistent activity has been proposed as a therapeutic strategy in preclinical experimentation, although it has not been translated to the clinics yet.
In an attempt to identify the molecular determinants of persistent neutrophil activation in diabetic conditions, Umehara and colleagues investigated specific miRNAs that are differentially expressed by neutrophils in diabetic and healthy mice and identified miR-129-2-3p as one of the most downregulated [28]. Consistent with the capacity of this miRNA to inhibit the expression of proinflammatory genes (i.e., Caspase-6 and C-C chemokine receptor type 2, Ccr2), its topical application to a wound of diabetic mice resulted in improved healing, although the cell type responsible for this in vivo effect has not been clearly identified.
Furthermore, neutrophils isolated from fresh whole blood obtained from diabetic individuals produce an increased amount of NETs [29]. High glucose correlates with elevated calcium levels in neutrophils and enhanced activation of calcium-dependent enzymes that are essential for chromatin decondensation (i.e., peptidylarginine deiminase 4, PAD4). Consistently, genetic inactivation of PAD4 or systemic NETosis inhibition by DNaseI improved wound healing and re-epithelialization in diabetic mice.
As any inflammatory cell, neutrophils are recruited to the wound by chemoattractant molecules that often bind receptors expressed by multiple cell types, thus allowing a coordinated multicellular response. One example is ChemR23, a receptor abundantly expressed by neutrophils but also by platelets and macrophages. This receptor binds the anti-inflammatory and pro-resolving peptide Chemerin15 (C15), which is derived from the cleavage of the chemoattractant protein chemerin, mostly known for its role in adipocyte differentiation and lipolysis [87]. C15 exerts reduced chemoattractive and enhanced anti-inflammatory activities compared to full-length chemerin [88]. Accordingly, its topical delivery as a gel on cutaneous burn wounds reduced the extension of granulation tissue, accelerating closure and re-epithelialization [30], as measured by fluorescence spinning disk intravital imaging. These data are consistent with previous evidence showing that C15/ChemR23 signaling suppresses integrin clustering in neutrophils, thereby reducing their trans-endothelial migration [89].

4. Monocytes/Macrophages

As in any response to tissue injury, circulating monocytes are massively recruited to the site of the skin wound, where they differentiate into macrophages able to mediate both destructive and reparative functions [90,91]. Various monocyte subtypes can be recognized based on the expression level of multiple cell surface markers. During the early phase of wound healing, CD11b+/Ly6Chi cells, commonly named M1 macrophages, produce abundant levels of proinflammatory molecules, including IL-1β, TNF-α, IL-6, and iNOS (inducible NO synthase) [92,93]. During the second phase, proinflammatory M1 macrophages switch toward an anti-inflammatory and pro-regenerative M2 phenotype (CD11b+/Ly6Clo), characterized by the secretion of Transforming Growth Factor (TGF)-β1, Vascular Endothelial Growth Factor (VEGF)-A, Arginase-1, and IL-10 [93,94]. As any classification, the distinction between M1 and M2 macrophages appears to be over simplistic. Macrophages recruited during the early phase sometimes express M2 markers and both macrophage types derive from the same pool of circulating monocytes [95,96]. Thus, it is very difficult, if not unrealistic, to strictly control macrophage polarization for therapeutic purposes, as highlighted in recent reviews [90,92,97].
In pathological states, like diabetes and obesity, monocyte recruitment, their differentiation into macrophages, and macrophage polarization are often impaired [93,98]. Hypertrophic adipocytes produce a plethora of adipokines that attract M1 macrophages and create a feedforward loop, resulting in a chronic low-grade inflammatory state [99,100] and lack of transition to an effective reparative phase [93,98].
Macrophage-based cell therapies that have reached clinical experimentation rely on their recruitment, activity modulation, or direct injection into the wound [97]. The most exploited strategy has been the local injection of human granulocyte-macrophage colony stimulating factor (hGM-CSF), which promoted healing in both refractory chronic skin ulcers and second-degree burns [32,33,34]. Approaches so far considered for their capacity to activate macrophages are based on the use of compounds that are naturally produced by microorganisms and can be purified as clinical-grade molecules. Among these are β-glucans, natural components of the cell walls of bacteria and fungi, which constitute the principal components of several Asiatic medicinal products, and the mycoplasma-derived macrophage-activating lipopeptide-2 (MALP-2) [35,36]. The local application of these immunomodulators has been reported as safe and potentially useful to improve healing of human wounds, although further studies will be needed to definitely prove efficacy [35,36]. Finally, the local injection of autologous macrophages has been attempted to treat both deep sternal wound infections following open heart surgery and refractory human ulcers in diabetic feet [37,38]. While these studies confirm the feasibility of the approach, no conclusions can be drawn in terms of safety and efficacy.
Because the initial expectations raised by these cellular therapies have not been met by the results of clinical studies, more preclinical work is needed to better define the appropriate timing of macrophage administration [101] and the mechanisms by which they could be of real benefit in skin regeneration. Over the last six years, additional animal studies have been undertaken to identify novel molecules able to boost both macrophage recruitment and polarization. Perhaps the most promising one is CXCL12 (SDF1), which recruits macrophages through its binding to CXCR4. The local application of lactic acid bacteria transformed with a CXCL12-expressing plasmid accelerated wound closure in healthy mice, in mouse models of hyperglycemia and peripheral ischemia, as well as in an ex vivo model of human skin [31]. In addition, exposure to CXCL12 favored M2 polarization and secretion of TGF-β. Similar results on macrophage polarization were obtained by using exosomes derived from a macrophage cell line (Raw264.7) [102]. Macrophage polarization is known to depend on multiple epigenetic changes, which are often impaired in diabetes [39,40]. At least two methyltransferases, MLL1 and Dnmt1, appear upregulated in macrophages of diabetic patients, in which they impose an activation mark on the promoter of proinflammatory genes (i.e., IL-1β, NOS2, and TNF-α) and inhibit the expression of factors inducing macrophage differentiation, respectively. The genetic inactivation of these methyltransferases has been shown to improve wound healing in animal models. However, the translation of this approach to humans is still unproven.
In conclusion, several open questions remain, which at present do not support the clinical use of these cells. First, the correlation between the mouse and human macrophage phenotype has been poorly proven. Second, the appropriate timing for macrophage therapy application has not been defined either in preclinical or clinical settings. Finally, as also highlighted for other cell populations, the design of clinical trials has been so far of poor quality, in terms of both patient recruitment and clinical endpoints.

5. Dendritic Cells

Dendritic cells (DCs) are antigen presenting cells that prime T cell responses. In the epidermis, DCs are called Langerhans cells (LCs) and derive their name from the surface receptor CD207 (Langerin). They exhibit a unique behavior, characterized by rhythmic extension and retraction of their dendrites in intercellular spaces between keratinocytes [3,103]. In the dermis, there are two subtypes of resident DCs, also named type 1 and type 2 myeloid/conventional DCs, which trigger Th1 and Th2 responses, respectively [104,105]. Epidermal and dermal DCs serve as first-line defenders, presenting new antigens to T cells within the dermis and migrating into draining lymph nodes where they continue to activate T cell-mediated adaptive responses [3,106].
In addition, plasmacytoid dendritic cells (pDCs) do not physiologically reside in the skin but are recruited together with neutrophils in case of injury [107]. They are particularly active during the early phase, when they produce IFN-α/β in response to nucleic acids released by damaged cells and sensed by the intracellular toll-like receptor 7 and 9 (TLR7 and TLR9).
Preclinical evidence of their positive role stems from multiple animal models of DC depletion generated using either transgenic animals or neutralizing antibodies. DC depletion invariably resulted in a suppressed early inflammatory response, reduced re-epithelialization, and decreased revascularization of healing wounds [41,107]. Conversely, systemic treatment with the DC-specific growth factor recombinant FMS-like tyrosine kinase-3 ligand (FL) significantly accelerated wound closure when delivered during the early but not the late phase of wound healing. Despite these promising insights into the role of DCs in wound healing, their therapeutic relevance in humans is still completely unexplored.

6. Lymphocytes

The adaptive immune response is orchestrated by lymphocytes, a heterogeneous population that include B cells, responsible for the humoral response, and T cells, which mainly exert cytotoxic activity. While their major function is to combat pathogen invasion, emerging evidence points toward their role in regenerative processes, including wound healing [108]. The healthy skin is colonized by different T cell subsets, identifiable by the expression of specific T cell receptor (TCR) isoforms. Two major populations, namely γδ-TCR and αβ-TCR lymphocytes, physiologically reside in the skin of both humans and rodents [109].
The most abundant T cell types in the murine healthy skin are two specialized classes of γδ-TCR lymphocytes, namely Vγ5Vδ1 dendritic epidermal T cells (DETCs) and Vγ4 dermal lymphocytes [110]. The crucial role of γδ-TCR lymphocytes in wound healing has been first described using TCRδ−/−mice, which lack epidermal lymphocytes and show a poor skin regenerative response after injury, resulting in reduced keratinocyte proliferation [111,112].
DETCs stably reside in the epidermis and have a typical dendritic cell shape, which facilitates their interaction with keratinocytes [113]. Upon activation by infection or injury, they change into round-shaped active T cells and promote keratinocyte proliferation through the secretion of keratinocyte growth factor (KGF-1) and insulin-like growth factor-1 (IGF-1) [114]. Vγ4 dermal lymphocytes normally reside in the dermis and migrate to the epidermis in response to proinflammatory molecules, such as chemokine (C-C motif) ligand 20 (CCL20), which binds chemokine receptor 6 (CCR6) expressed on their membrane [115,116]. These cells participate in the first phase of wound healing through the secretion of tumor necrosis factor α (TNF-α) and IL-17 [117].
Therapeutic approaches exploit a positive feedback loop existing between DETCs and keratinocytes. Injured keratinocytes secrete IL-15, which stimulates IGF-1 secretion by DETCs [118]. In turn, IGF-1 promotes keratinocyte proliferation and further secretion of IL-15. The importance of this paracrine loop in skin regeneration has been demonstrated in diabetic mice, which have reduced levels of IL-15 and a low number of DETC in their epidermis, with consequent decreased levels of KGF-1 and IGF-1 and impaired wound healing. Engraftment of an extra amount of DETCs in the same mice rescued the KGF-1 and IGF-1 levels and improved wound regeneration [43,118]. A similar therapeutic effect was exerted by the administration of IL-15, which restored IGF-1 production by DETC both in vivo and in vitro [42].
In addition to DETC, Vγ4 T cells are also reduced in diabetic mice, consistent with decreased levels of both IL-17 and CCL20 [119]. Evidence supporting the role of Vγ4 T cells in skin regeneration comes from IL-17−/−mice, which are characterized by defective wound healing [120], as well as from the therapeutic activity of IL-17 in improving wound healing in diabetic mice [119]. More recently, conflicting data have been reported, showing that IL-17 inhibits IGF-1 secretion in the wound bed and that Vγ4 T cell depletion accelerates wound healing, which would indicate a negative effect of these cells in the process [116].
Additional concerns on the therapeutic potential of γδ-TCR lymphocytes for regenerative medicine stems from the fact that no human counterpart for either DETC or Vγ4 T cells has been clearly identified and that γδ-TCR cells represent less than 10% of total T cells in the human skin [121]. One single study reported the capacity of a specific subset of human γδ-TCR cells to produce IGF-1 in vitro when isolated from an acute but not from a chronic wound [122].
The αβ-TCR lymphocytes (CD4+ helper and CD8+ cytotoxic) represent the vast majority of skin-resident T cells in humans, and a consistent part of them in mice [110,122]. While in humans, they reside in the epidermis, and in mice, they are localized mainly in the dermis, proximal to hair follicles [123,124]. In addition to the resident population, circulating CD4+ and CD8+ lymphocytes are recruited to the wound bed, peaking at day 1 and 7, respectively. They exert antimicrobial function during the early inflammatory phase and recruit additional immune cells (neutrophils and macrophages) through the secretion of a variety of cytokines, including IL-1β, IL-6, TNF-α, CXCL2, and CCL2 [108,125]. Several studies highlighted the detrimental role of CD4+ and CD8+ lymphocytes in tissue regeneration and revascularization. For instance, CD4+ cell ablation promotes heart regeneration [126] and both CD4+ [127] and CD8+ [128] blockade improves angiogenesis in diabetic mice. Yet, their functional role in skin regeneration is controversial, as indicated by numerous studies reporting conflicting results, and a clear discrimination of the specific role of circulating and resident lymphocytes is often missing. For instance, both CD4 and CD8 knockout mice do not show any major deficit in wound healing, despite having reduced cytokine expression and poor macrophage and neutrophil infiltration [125]. Even more surprisingly, athymic nude mice lacking all T cells present scarless skin repair, similar to what happens during fetal development, suggesting that T cells impair re-epithelialization and favor repair by scarring [44]. Opposite data have been obtained in SCID mice transplanted with either CD8+, CD4+ lymphocytes, or their combination. In this study, CD4+ cells, alone or combined with CD8+ cells, induced robust re-epithelialization and neoangiogenesis together with reduced inflammation and scarring [45]. While the role of CD8+ and CD4+ lymphocytes in wound healing in the presence of either tissue ischemia or diabetes has not been investigated in animals, few data are available in humans. Diabetic individuals with foot ulceration showed a decreased number of total circulating lymphocytes [129], with a relative accumulation of activated T cells and increased TNFα levels, indicative of a proinflammatory condition [130]. Overall, this evidence suggests that altered balance of T cell sub-populations may impact on the closure of diabetic wounds, yet further studies are needed to characterize their precise role.
An additional class of αβ-TCR lymphocytes is composed by Tregs. These are considered the major players in immune suppression, maintaining tolerance to self-antigens, and are traditionally characterized by the expression of CD4, CD25, and the transcription factor forkhead box P3 (FOXP3). Emerging evidence indicates that they are a heterogenous population, able to promote tissue regeneration through mechanisms independent from their immune-modulatory function [131,132]. They physiologically reside in both the human and murine dermis [133,134], and rapidly accumulate to the site of wounding. Different receptors have been described as key mediators of their recruitment, including EGFR and CCR4 [135,136]. Depletion of Tregs in healthy mice resulted in delayed wound closure, associated with increased proinflammatory cytokines and macrophage recruitment [137]. Contrasting results were obtained in diabetic mice, in which either Treg depletion or inhibition of their recruitment resulted in increased collagen deposition, reduced cytokine content, and faster wound healing [136]. This again warrants additional studies to better elucidate the mechanisms through which Tregs might modulate the two phases of wounds, and in particular their capacity to control macrophage polarization, as described in the setting of myocardial infarction [138], and revascularization of ischemic tissues [127].
Finally, B lymphocytes have been shown to colonize skin wounds in rodents and humans, but very poor information is available about their function [108,139]. The topical application of naïve B cells, but not T cells, accelerated the closure of either acute or chronic skin lesions in diabetic mice, associated with enhanced collagen deposition, angiogenesis, increased TGF-β, and reduced MMP2 levels [46]. In accordance, in CD19−/−mice, B cells are hyporesponsive, and displayed impaired wound healing, decreased recruitment of neutrophils and macrophages, and an altered cytokine profile. On the other hand, transgenic overexpression of CD19, which drives B lymphocyte hyper-responsiveness, induced faster healing [140], thus supporting a positive role of these cells in wound healing. Overall, these findings suggest that a correct balance between the different lymphocyte populations during wound healing may represent a promising mechanism to investigate for future therapies.

7. Peripheral Blood Mononuclear Cells

PBMNCs represent a heterogeneous cellular pool composed by roughly 65% lymphocytes, 25% monocytes, and 10% granulocytes [141,142]. They can be easily isolated from venous blood using different methods, including gravity sedimentation, gel layering, filtering and leukapheresis [141,142,143].
A few studies have evaluated the potential of PBMNCs, without any cell-specific enrichment, to promote healing when directly applied to the wound. A seminal study in 1994 demonstrated that the topical application of autologous PBMCs to skin ulcers in patients affected by peripheral arterial disease (PAD) and post-thrombotic syndrome resulted in accelerated healing and pain relief [144].
While these cells have not been vastly used to treat chronic wounds in the following years, they were exploited in multiple trials for the treatment of people affected by ischemic diseases, and in particular myocardial infarction (MI). This improved the cardiac function, but their real regenerative potential is still unproven. Starting from the evidence that very few transplanted cells survived in the ischemic heart, an intriguing hypothesis was formulated and assumed that a large proportion of apoptotic transplanted cells could modulate local tissue reaction, by downregulating innate and adaptive immunity [145]. This led to a series of experimental studies supporting the efficacy of the secretome of apoptotic PBMNCs (APOSEC) in improving the outcome of myocardial infarction in rodents [146,147], followed by the application of a similar strategy also in wound healing [148]. A small randomized double-blinded placebo-controlled phase 1 trial on 10 volunteers was performed to establish the safety and the efficacy of autologous APOSEC on artificially created wounds with negative results [49].
A more recent application entails the seeding of a mixture of fibroblasts and PBMNCs in cellular sheets, which can be topically applied to the wound. Incorporation of PBMNCs was shown to produce a variety of cytokines (i.e., VEGF, Hepatocyte Growth Factor, HGF, TGF-β, CXCL1, and CXCL2) and to promote revascularization and wound closure in diabetic mice [47,48].

8. Endothelial Progenitor Cells

Highly controversial is the assumption that PBMNC also contains a population of CD34+ endothelial progenitor cells (EPCs), which could contribute to vessel formation through their direct incorporation into nascent vascular tubes. This concept was put forward by Asahara in 1997 and assumes that a vasculogenesis-like process might occur in adulthood [149]. The idea was embraced with enthusiasm by the scientific community, as it could pave the way to novel clinical strategies for tissue revascularization.
After more than 20 years of preclinical and clinical trials using EPCs, their real capacity to differentiate into functional endothelial cells and become incorporated into new vessels, providing clinical benefit, has never been proved. Major concerns have been raised on the method used to prove and quantify their differentiation into endothelial cells [150,151,152].
In an attempt to better characterize these cells, CD34+ EPCs have been expanded ex vivo and distinguished as early and late EPCs depending on time of appearance of endothelial cells in the culture [150,151,152]. Early and late EPCs appeared to exhibit a distinct phenotype and, thus, they were reclassified as either hematopoietic (CD45+/CD133hi) or non-hematopoietic (CD45/CD133lo) EPCs. Early hematopoietic EPCs do not generate endothelial cells able to form colonies and, when transplanted into lethally irradiated recipient mice, give rise to a minimal number of endothelial cells (no more than 1%) that persist for a few months after transplantation [153,154]. Further analyses revealed that these cells are monocytic in nature and might be assimilated to M2 macrophages, able to secrete proangiogenic molecules. Thus, the only way these cells appear to contribute to angiogenesis is through a paracrine mechanism and not through their direct incorporation into vascular structures.
In contrast, non-hematopoietic EPCs, forming late endothelial colonies, can be derived from umbilical and peripheral blood but also from other tissues, including white adipose tissue, intestine, and liver [155,156,157,158]. These cells have been variably named over time, until a recent consensus statement on the nomenclature of endothelial progenitors has recommended the use of the term “endothelial colony forming cells” or “ECFCs” [152]. In the following sections, we will still keep the term EPC, as authors did not always discriminate between hematopoietic and non-hematopoietic origin in most old studies. The majority of the clinical trials using either hematopoietic EPCs or ECFCs have focused on myocardial infarction and peripheral arterial disease, with only a few studies considering the effect of these cells on chronic wounds [55,159].
As for other rare populations of circulating cells, the simplest therapeutic strategies consist in either their mobilization from the periphery or their enhanced recruitment at the level of the wound [155]. Pre-clinical studies have largely explored this second strategy. Among the most relevant chemoattractants for EPCs is SDF-1/CXCL12, which binds the CXCR4 receptor, widely and constitutively expressed by cells of both hematopoietic and endothelial lineage. This pathway is compromised in diabetic patients, as hyperglycemia is known to reduce SDF1 expression through inactivation of its transcriptional regulator hypoxia-inducible factor-1 alpha (HIF-1α) [50]. In an attempt to rescue the homing of EPCs through this pathway, the administration of recombinant SDF-1 into the skin of diabetic mice restored EPC recruitment and accelerated wound closure. An alternative strategy consisted in the pharmacological inhibition of dipeptidyl peptidase-4 (DPP4), a membrane-bound extracellular peptidase that cleaves and inactivates SDF-1 [51], with analogous promising results. The clinical translation of these approaches is still at its infancy, with only few human studies showing the actual increase in the number of circulating EPCs upon delivery of the DPP4 inhibitor or human recombinant G-CSF (NCT02694575 and NCT01102699).
Other studies have tried to enhance the EPC viability and proliferation at the site of the wound. Starting from the evidence that angiogenesis during the menstrual cycle largely depends on estrogen and that the latter increases the colony-forming capacity of EPCs in culture [160,161], the topical administration of estrogen has been successfully validated as a treatment to accelerate wound healing in diabetic mice [52]. However, to what extent this therapeutic effect can be ascribed to EPCs or other estrogen-responsive cells participating in the healing process (i.e., keratinocytes and fibroblasts) remains an open question [162,163,164].
Finally, EPCs could be directly transplanted into the wound. This has been attempted in multiple preclinical models, using either syngeneic or human EPCs in immunocompromised animals [53,54,165]. One trial assessed the effect of the intra-arterial delivery of CD133+ EPCs for the treatment of diabetic foot in 53 patients [55]. This treatment prevented any amputation and resulted in a significant increase in limb perfusion, paralleled by increased circulating levels of VEGF-A, and reduced levels of IL-6. Yet, the real clinical benefit derived from this treatment is still not obvious, based on the large standard deviation that is often reported in this type of analysis. In an additional trial, EPCs from diabetic patients with a non-healing foot were first mobilized with G-CSF and then purified as CD34+/VEGFR2+ cells, prior to their intramuscular injection into the same individuals [56]. Major limitations of the study, i.e., the inclusion of only five patients and the lack of a control group, do not allow any definitive conclusion about the efficacy of this approach.
In conclusion, the persistence of hemangioblasts in postnatal life, able to sustain vasculogenesis-like phenomena in adult organisms, has been harshly challenged. Similarly, EPCs of hematopoietic origin have never been reported as being able to give rise to new vessels. It seems more realistic the existence of ECFCs of non-hematopoietic lineage, although the physiological contribution of these cells to the formation of new blood vessels in adult organisms and, more importantly, their real therapeutic potential, still has to be determined. To this end, major efforts are required to define a consensus in terms of their culture methods, phenotypic characterization, therapeutic dose, and route of administration.

9. Stromal Vascular Fraction

The evidence that multiple cell types contribute to tissue regeneration in wound healing has prompted the use of heterogeneous cell populations, such as the one that can be obtained from white adipose tissue and is commonly named stromal vascular fraction (SVF). This fraction contains endothelial cells, pericytes, smooth muscle cells, immune cells, fibroblasts, preadipocytes, and a population of multipotent progenitors, often referred as adipose-derived stem cells (ASCs) [166,167]. Being of mesenchymal origin, ASCs differentiate into adipocytes, osteocytes, and chondrocytes and secrete a variety of factors able to sustain tissue regeneration and the formation of new blood vessels in a paracrine manner [168,169,170]. ASCs have been extensively tested for their capacity to accelerate wound healing in multiple models of diabetes, particularly when embedded in scaffolds [171,172]. Notably, the implantation of decellularized scaffolds recapitulates the therapeutic effect, suggesting that the latter relies on the paracrine secretion of pro-regenerative factors. Further studies have shown that ASCs, similar to the whole SVF, exerted a comparable therapeutic effect on wound healing in hyperglycemic mice, resulting in the expression of genes favoring fibroblast migration, angiogenesis, and macrophage recruitment [173], again supporting a paracrine mechanism of action. Similar conclusions were drawn upon the local or systemic delivery of human ASC exosomes, which improved wound healing by promoting fibroblasts’ migration and collagen production [174].
Consistent with the immunomodulatory function of other mesenchymal stromal cells, both ASCs and the entire SVF could contribute to wound healing by locally controlling the immune response [175]. For instance, the addition of the SVF to vascularized human dermo-epidermal skin substitutes promoted M2 macrophage polarization at the level of the wound in nude rats [57].
More recently, the idea of using the entire SVF, also including other cell types that can directly contribute to skin regeneration through their direct incorporation into the newly formed tissue, is emerging as a more potent strategy compared to isolated ASCs.
A direct comparison of human SVF with either ASCs or SVF-derived condition medium indicated that the whole SVF was the most effective in promoting angiogenesis, as well as the proliferation and migration of keratinocytes and fibroblasts [176,177]. The superior efficacy of the SVF compared to cultured ASCs was also confirmed by additional studies in nude mice [178].
This indicates that SVF might contribute to tissue regeneration through cellular mechanisms that do not rely exclusively on ASCs or secreted factors. Indeed, fluorescently labeled SVF cells implanted in mouse flanks and ischemic hind limbs were incorporated into newly formed vessels and exhibited an endothelial phenotype, with null contribution from other cell types [179].
On the basis of these promising preclinical results, the therapeutic efficacy of the SVF in improving skin regeneration has been investigated in various clinical trials. While most studies have considered a low number of patients, not allowing conclusive results [58,59,60], promising data were obtained by a trial that enrolled 28 patients with diabetic foot ulcers, who were treated with lipoaspirate SVF and compared to a control group of 26 matched patients. The SVF-treated group showed faster healing, resulting in full wound closure at 8 weeks in 100% of patients compared to 62% in the control group.
Despite these promising results, major concerns limiting the clinical use of the SVF refer to the poor standardization of purification protocols, culture conditions, time, and phenotypic characterization of ASCs and SVF cells. Recent single-cell sequencing approaches are expected to improve the characterization of this heterogeneous cell population and thus the choice of appropriate markers for their purification and therapeutic use [180,181,182,183,184].

10. Conclusions and Perspectives

The lack of effective therapies to treat non-healing wounds and prevent amputation, together with an increasing understanding of the role of various immune cells in skin regeneration and healing, have promoted the development of cellular and molecular therapies. These essentially aim at mobilizing therapeutic cells into the circulation, recruiting them and modulating their activity at the wound site, or directly supplementing them to the wound bed. However, none of these formulations can be considered a standard of care, likely due to multiple limitations. First, both preclinical and clinical studies often use different parameters to evaluate efficacy. For example, re-epithelialization and revascularization were assessed by multiple modalities and the presence of granulation tissue was variably considered as either a positive or a negative element. Second, clinical end points were mostly based on wound area and healing time, and did not consider either the type of healing (i.e., scarring versus re-epithelialization) or incidence of long-term relapse. Finally, well-designed, randomized, and placebo-controlled clinical trials are still lacking for most cell types.
By assessing the rationale and the supposed mechanism of action of these approaches, it emerges that some signaling pathways regulate the activity of multiple cell types. For example, the SDF1/CXCR4 axis has been exploited to recruit both macrophages and ECFCs, paving the way to the development of multitarget therapies able to orchestrate the participation of different cells.
This also highlights that a better understanding of the individual contribution of each cell type, in different phases of the healing process, is required to optimize and select the best cellular therapies. New tools will possibly provide simpler models for investigating the role and the therapeutic potential of immune cells in skin regeneration.
Zebrafish is an emerging model allowing rapid assessment of the healing of skin wounds, which can be easily and reproducibly generated on its flank [27,185,186]. In addition, both angiogenesis and regeneration can be visualized in live imaging through its transparent skin. To what extent this model will be exploitable to test the therapeutic potential of cellular therapies remains an open question. More suitable to this end is the use of ex vivo models of human skin, including two- and three-dimensional cultures of human cells (keratinocytes, fibroblasts, and endothelial cells), organotypic skin cultures, and debrided skin specimens [62,187].

Funding

This work was supported by grant POR-FESR SH_STR_13 “PREFER” to S.Z.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kim, H.S.; Sun, X.; Lee, J.-H.; Kim, H.-W.; Fu, X.; Leong, K.W. Advanced drug delivery systems and artificial skin grafts for skin wound healing. Adv. Drug Deliv. Rev. 2019, 146, 209–239. [Google Scholar] [CrossRef]
  2. Nussbaum, S.R.; Carter, M.J.; Fife, C.E.; DaVanzo, J.; Haught, R.; Nusgart, M.; Cartwright, D. An Economic Evaluation of the Impact, Cost, and Medicare Policy Implications of Chronic Nonhealing Wounds. Value Health 2018, 21, 27–32. [Google Scholar] [CrossRef] [Green Version]
  3. Rodrigues, M.; Kosaric, N.; Bonham, C.A.; Gurtner, G.C. Wound Healing: A Cellular Perspective. Physiol. Rev. 2019, 99, 665–706. [Google Scholar] [CrossRef] [PubMed]
  4. Eming, S.A.; Martin, P.; Tomic-Canic, M. Wound repair and regeneration: Mechanisms, signaling, and translation. Sci. Transl. Med. 2014, 6, 265sr6. [Google Scholar] [CrossRef] [Green Version]
  5. Guest, J.F.; Ayoub, N.; McIlwraith, T.; Uchegbu, I.; Gerrish, A.; Weidlich, D.; Vowden, K.; Vowden, P. Health economic burden that wounds impose on the National Health Service in the UK. Bmj Open 2015, 5, e009283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Roger, V.L. Epidemiology of Heart Failure. Circ. Res. 2013, 113, 646–659. [Google Scholar] [CrossRef] [PubMed]
  7. Heyer, K.; Herberger, K.; Protz, K.; Glaeske, G.; Augustin, M. Epidemiology of chronic wounds in Germany: Analysis of statutory health insurance data: Epidemiology of chronic wounds in Germany. Wound Rep. Reg. 2016, 24, 434–442. [Google Scholar] [CrossRef]
  8. Sen, C.K. Human Wounds and Its Burden: An Updated Compendium of Estimates. Adv. Wound Care 2019, 8, 39–48. [Google Scholar] [CrossRef] [Green Version]
  9. Akhtar, S.; Hasham, S.; Abela, C.; Phipps, A.R. The use of IntegraTM in necrotizing fasciitis. Burns 2006, 32, 251–254. [Google Scholar] [CrossRef]
  10. Shanmugam, V.K.; Angra, D.; Rahimi, H.; McNish, S. Vasculitic and autoimmune wounds. J. Vasc. Surg. Venous Lymphat. Disord. 2017, 5, 280–292. [Google Scholar] [CrossRef] [Green Version]
  11. Avishai, E.; Yeghiazaryan, K.; Golubnitschaja, O. Impaired wound healing: Facts and hypotheses for multi-professional considerations in predictive, preventive and personalised medicine. EPMA J. 2017, 8, 23–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Weigert, R.; Choughri, H.; Casoli, V. Management of severe hand wounds with Integra® dermal regeneration template. J. Hand Surg. Eur. Vol. 2011, 36, 185–193. [Google Scholar] [CrossRef] [PubMed]
  13. Graham, G.P.; Helmer, S.D.; Haan, J.M.; Khandelwal, A. The Use of Integra® Dermal Regeneration Template in the Reconstruction of Traumatic Degloving Injuries. J. Burn Care Res. 2013, 34, 261–266. [Google Scholar] [CrossRef] [PubMed]
  14. Pham, C.; Greenwood, J.; Cleland, H.; Woodruff, P.; Maddern, G. Bioengineered skin substitutes for the management of burns: A systematic review. Burns 2007, 33, 946–957. [Google Scholar] [CrossRef]
  15. Böttcher-Haberzeth, S.; Biedermann, T.; Reichmann, E. Tissue engineering of skin. Burns 2010, 36, 450–460. [Google Scholar] [CrossRef]
  16. Tufaro, A.P.; Buck, D.W.; Fischer, A.C. The Use of Artificial Dermis in the Reconstruction of Oncologic Surgical Defects. Plast. Reconstr. Surg. 2007, 120, 638–646. [Google Scholar] [CrossRef]
  17. Stadelmann, W.K.; Digenis, A.G.; Tobin, G.R. Physiology and healing dynamics of chronic cutaneous wounds. Am. J. Surg. 1998, 176, 26S–38S. [Google Scholar] [CrossRef]
  18. Fonder, M.A.; Lazarus, G.S.; Cowan, D.A.; Aronson-Cook, B.; Kohli, A.R.; Mamelak, A.J. Treating the chronic wound: A practical approach to the care of nonhealing wounds and wound care dressings. J. Am. Acad. Dermatol. 2008, 58, 185–206. [Google Scholar] [CrossRef]
  19. Richmond, N.A.; Maderal, A.D.; Vivas, A.C. Evidence-based management of common chronic lower extremity ulcers: Management of chronic lower extremity ulcers. Dermatol. Ther. 2013, 26, 187–196. [Google Scholar] [CrossRef]
  20. Frykberg, R.G.; Banks, J. Challenges in the Treatment of Chronic Wounds. Adv. Wound Care 2015, 4, 560–582. [Google Scholar] [CrossRef] [Green Version]
  21. Wilgus, T.A.; Roy, S.; McDaniel, J.C. Neutrophils and Wound Repair: Positive Actions and Negative Reactions. Adv. Wound Care (New Rochelle) 2013, 2, 379–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Segel, G.B.; Halterman, M.W.; Lichtman, M.A. The paradox of the neutrophil’s role in tissue injury. J. Leukoc. Biol. 2011, 89, 359–372. [Google Scholar] [CrossRef] [PubMed]
  23. McCarty, S.M.; Percival, S.L. Proteases and Delayed Wound Healing. Adv. Wound Care 2013, 2, 438–447. [Google Scholar] [CrossRef] [PubMed]
  24. Telgenhoff, D.; Shroot, B. Cellular senescence mechanisms in chronic wound healing. Cell Death Differ. 2005, 12, 695–698. [Google Scholar] [CrossRef] [PubMed]
  25. Frueh, F.S.; Später, T.; Körbel, C.; Scheuer, C.; Simson, A.C.; Lindenblatt, N.; Giovanoli, P.; Menger, M.D.; Laschke, M.W. Prevascularization of dermal substitutes with adipose tissue-derived microvascular fragments enhances early skin grafting. Sci. Rep. 2018, 8, 10977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Demidova-Rice, T.N.; Durham, J.T.; Herman, I.M. Wound Healing Angiogenesis: Innovations and Challenges in Acute and Chronic Wound Healing. Adv. Wound Care 2012, 1, 17–22. [Google Scholar] [CrossRef]
  27. Gurevich, D.B.; Severn, C.E.; Twomey, C.; Greenhough, A.; Cash, J.; Toye, A.M.; Mellor, H.; Martin, P. Live imaging of wound angiogenesis reveals macrophage orchestrated vessel sprouting and regression. EMBO J. 2018, 37, e97786. [Google Scholar] [CrossRef]
  28. Umehara, T.; Mori, R.; Mace, K.A.; Murase, T.; Abe, Y.; Yamamoto, T.; Ikematsu, K. Identification of Specific miRNAs in Neutrophils of Type 2 Diabetic Mice: Overexpression of miRNA-129-2-3p Accelerates Diabetic Wound Healing. Diabetes 2019, 68, 617–630. [Google Scholar] [CrossRef] [Green Version]
  29. Wong, S.L.; Demers, M.; Martinod, K.; Gallant, M.; Wang, Y.; Goldfine, A.B.; Kahn, C.R.; Wagner, D.D. Diabetes primes neutrophils to undergo NETosis which severely impairs wound healing. Nat. Med. 2015, 21, 815–819. [Google Scholar] [CrossRef] [Green Version]
  30. Cash, J.L.; Bass, M.D.; Campbell, J.; Barnes, M.; Kubes, P.; Martin, P. Resolution mediator chemerin15 reprograms the wound microenvironment to promote repair and reduce scarring. Curr. Biol. 2014, 24, 1406–1414. [Google Scholar] [CrossRef] [Green Version]
  31. Vågesjö, E.; Öhnstedt, E.; Mortier, A.; Lofton, H.; Huss, F.; Proost, P.; Roos, S.; Phillipson, M. Accelerated wound healing in mice by on-site production and delivery of CXCL12 by transformed lactic acid bacteria. Proc. Natl. Acad. Sci. USA 2018, 115, 1895–1900. [Google Scholar] [CrossRef] [Green Version]
  32. Huang, G.; Sun, T.; Zhang, L.; Wu, Q.; Zhang, K.; Tian, Q.; Huo, R. Combined application of alginate dressing and human granulocyte-macrophage colony stimulating factor promotes healing in refractory chronic skin ulcers. Exp. Med. 2014, 7, 1772–1776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Yan, H.; Chen, J.; Peng, X. Recombinant human granulocyte-macrophage colony-stimulating factor hydrogel promotes healing of deep partial thickness burn wounds. Burns 2012, 38, 877–881. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, L.; Chen, J.; Han, C. A multicenter clinical trial of recombinant human GM-CSF hydrogel for the treatment of deep second-degree burns. Wound Repair Regen. 2009, 17, 685–689. [Google Scholar] [CrossRef]
  35. Zykova, S.N.; Balandina, K.A.; Vorokhobina, N.V.; Kuznetsova, A.V.; Engstad, R.; Zykova, T.A. Macrophage stimulating agent soluble yeast β-1,3/1,6-glucan as a topical treatment of diabetic foot and leg ulcers: A randomized, double blind, placebo-controlled phase II study. J. Diabetes Investig. 2014, 5, 392–399. [Google Scholar] [CrossRef] [Green Version]
  36. Niebuhr, M.; Mühlradt, P.F.; Wittmann, M.; Kapp, A.; Werfel, T. Intracutaneous injection of the macrophage-activating lipopeptide-2 (MALP-2) which accelerates wound healing in mice--a phase I trial in 12 patients. Exp. Dermatol. 2008, 17, 1052–1056. [Google Scholar] [CrossRef]
  37. Orenstein, A.; Kachel, E.; Zuloff-Shani, A.; Paz, Y.; Sarig, O.; Haik, J.; Smolinsky, A.K.; Mohr, R.; Shinar, E.; Danon, D. Treatment of deep sternal wound infections post-open heart surgery by application of activated macrophage suspension. Wound Repair Regen. 2005, 13, 237–242. [Google Scholar] [CrossRef]
  38. Zuloff-Shani, A.; Kachel, E.; Frenkel, O.; Orenstein, A.; Shinar, E.; Danon, D. Macrophage suspensions prepared from a blood unit for treatment of refractory human ulcers. Transfus. Apher. Sci. 2004, 30, 163–167. [Google Scholar] [CrossRef]
  39. Kimball, A.S.; Joshi, A.; Carson, W.F.; Boniakowski, A.E.; Schaller, M.; Allen, R.; Bermick, J.; Davis, F.M.; Henke, P.K.; Burant, C.F.; et al. The Histone Methyltransferase MLL1 Directs Macrophage-Mediated Inflammation in Wound Healing and Is Altered in a Murine Model of Obesity and Type 2 Diabetes. Diabetes 2017, 66, 2459–2471. [Google Scholar] [CrossRef] [Green Version]
  40. Yan, J.; Tie, G.; Wang, S.; Tutto, A.; DeMarco, N.; Khair, L.; Fazzio, T.G.; Messina, L.M. Diabetes impairs wound healing by Dnmt1-dependent dysregulation of hematopoietic stem cells differentiation towards macrophages. Nat. Commun. 2018, 9, 1–13. [Google Scholar] [CrossRef] [Green Version]
  41. Vinish, M.; Cui, W.; Stafford, E.; Bae, L.; Hawkins, H.; Cox, R.; Toliver-Kinsky, T. Dendritic cells modulate burn wound healing by enhancing early proliferation. Wound Repair Regen. 2016, 24, 6–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Wang, Y.; Bai, Y.; Li, Y.; Liang, G.; Jiang, Y.; Liu, Z.; Liu, M.; Hao, J.; Zhang, X.; Hu, X.; et al. IL-15 Enhances Activation and IGF-1 Production of Dendritic Epidermal T Cells to Promote Wound Healing in Diabetic Mice. Front. Immunol. 2017, 8, 1557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Liu, Z.; Xu, Y.; Chen, L.; Xie, J.; Tang, J.; Zhao, J.; Shu, B.; Qi, S.; Chen, J.; Liang, G.; et al. Dendritic epidermal T cells facilitate wound healing in diabetic mice. Am. J. Transl. Res. 2016, 8, 2375–2384. [Google Scholar] [PubMed]
  44. Gawronska-Kozak, B.; Bogacki, M.; Rim, J.-S.; Monroe, W.T.; Manuel, J.A. Scarless skin repair in immunodeficient mice: Scarless skin repair in immundeficient mice. Wound Repair Regen. 2006, 14, 265–276. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, X.; Balaji, S.; Steen, E.H.; Li, H.; Rae, M.M.; Blum, A.J.; Miao, Q.; Butte, M.J.; Bollyky, P.L.; Keswani, S.G. T Lymphocytes Attenuate Dermal Scarring by Regulating Inflammation, Neovascularization, and Extracellular Matrix Remodeling. Adv. Wound Care 2019, 8, 527–537. [Google Scholar] [CrossRef] [PubMed]
  46. Sîrbulescu, R.F.; Boehm, C.K.; Soon, E.; Wilks, M.Q.; Ilieş, I.; Yuan, H.; Maxner, B.; Chronos, N.; Kaittanis, C.; Normandin, M.D.; et al. Mature B cells accelerate wound healing after acute and chronic diabetic skin lesions: Mature B cells accelerate wound healing. Wound Rep. Reg. 2017, 25, 774–791. [Google Scholar] [CrossRef]
  47. Ueno, K.; Takeuchi, Y.; Samura, M.; Tanaka, Y.; Nakamura, T.; Nishimoto, A.; Murata, T.; Hosoyama, T.; Hamano, K. Treatment of refractory cutaneous ulcers with mixed sheets consisting of peripheral blood mononuclear cells and fibroblasts. Sci. Rep. 2016, 6, 28538. [Google Scholar] [CrossRef] [Green Version]
  48. Mizoguchi, T.; Ueno, K.; Takeuchi, Y.; Samura, M.; Suzuki, R.; Murata, T.; Hosoyama, T.; Morikage, N.; Hamano, K. Treatment of Cutaneous Ulcers with Multilayered Mixed Sheets of Autologous Fibroblasts and Peripheral Blood Mononuclear Cells. Cell Physiol. Biochem. 2018, 47, 201–211. [Google Scholar] [CrossRef]
  49. Simader, E.; Traxler, D.; Kasiri, M.M.; Hofbauer, H.; Wolzt, M.; Glogner, C.; Storka, A.; Mildner, M.; Gouya, G.; Geusau, A.; et al. Safety and tolerability of topically administered autologous, apoptotic PBMC secretome (APOSEC) in dermal wounds: A randomized Phase 1 trial (MARSYAS I). Sci. Rep. 2017, 7, 6216. [Google Scholar] [CrossRef] [Green Version]
  50. Gallagher, K.A.; Liu, Z.-J.; Xiao, M.; Chen, H.; Goldstein, L.J.; Buerk, D.G.; Nedeau, A.; Thom, S.R.; Velazquez, O.C. Diabetic impairments in NO-mediated endothelial progenitor cell mobilization and homing are reversed by hyperoxia and SDF-1α. J. Clin. Invest 2007, 117, 1249–1259. [Google Scholar] [CrossRef] [Green Version]
  51. Whittam, A.J.; Maan, Z.N.; Duscher, D.; Barrera, J.A.; Hu, M.S.; Fischer, L.H.; Khong, S.; Kwon, S.H.; Wong, V.W.; Walmsley, G.G.; et al. Small molecule inhibition of dipeptidyl peptidase-4 enhances bone marrow progenitor cell function and angiogenesis in diabetic wounds. Transl. Res. 2019, 205, 51–63. [Google Scholar] [CrossRef] [PubMed]
  52. Zhuge, Y.; Regueiro, M.M.; Tian, R.; Li, Y.; Xia, X.; Vazquez-Padron, R.; Elliot, S.; Thaller, S.R.; Liu, Z.-J.; Velazquez, O.C. The effect of estrogen on diabetic wound healing is mediated through increasing the function of various bone marrow-derived progenitor cells. J. Vasc. Surg. 2018, 68, 127S–135S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Suh, W.; Kim, K.L.; Kim, J.-M.; Shin, I.-S.; Lee, Y.-S.; Lee, J.-Y.; Jang, H.-S.; Lee, J.-S.; Byun, J.; Choi, J.-H.; et al. Transplantation of Endothelial Progenitor Cells Accelerates Dermal Wound Healing with Increased Recruitment of Monocytes/Macrophages and Neovascularization. STEM CELLS 2005, 23, 1571–1578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kado, M.; Tanaka, R.; Arita, K.; Okada, K.; Ito-Hirano, R.; Fujimura, S.; Mizuno, H. Human peripheral blood mononuclear cells enriched in endothelial progenitor cells via quality and quantity controlled culture accelerate vascularization and wound healing in a porcine wound model. Cell Transpl. 2018, 27, 1068–1079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Zhang, X.; Lian, W.; Lou, W.; Han, S.; Lu, C.; Zuo, K.; Su, H.; Xu, J.; Cao, C.; Tang, T.; et al. Transcatheter Arterial Infusion of Autologous CD133+ Cells for Diabetic Peripheral Artery Disease. Available online: https://www.hindawi.com/journals/sci/2016/6925357/ (accessed on 22 May 2020).
  56. Tanaka, R.; Masuda, H.; Kato, S.; Imagawa, K.; Kanabuchi, K.; Nakashioya, C.; Yoshiba, F.; Fukui, T.; Ito, R.; Kobori, M.; et al. Autologous G-CSF-mobilized peripheral blood CD34+ cell therapy for diabetic patients with chronic nonhealing ulcer. Cell Transpl. 2014, 23, 167–179. [Google Scholar] [CrossRef]
  57. Klar, A.S.; Michalak-Mićka, K.; Biedermann, T.; Simmen-Meuli, C.; Reichmann, E.; Meuli, M. Characterization of M1 and M2 polarization of macrophages in vascularized human dermo-epidermal skin substitutes in vivo. Pediatr. Surg. Int. 2018, 34, 129–135. [Google Scholar] [CrossRef]
  58. Didangelos, T.; Koliakos, G.; Kouzi, K.; Arsos, G.; Kotzampassi, K.; Tziomalos, K.; Karamanos, D.; Hatzitolios, A.I. Accelerated healing of a diabetic foot ulcer using autologous stromal vascular fraction suspended in platelet-rich plasma. Regen. Med. 2018, 13, 277–281. [Google Scholar] [CrossRef]
  59. Nilforoushzadeh, M.A.; Sisakht, M.M.; Amirkhani, M.A.; Seifalian, A.M.; Banafshe, H.R.; Verdi, J.; Nouradini, M. Engineered skin graft with stromal vascular fraction cells encapsulated in fibrin–collagen hydrogel: A clinical study for diabetic wound healing. J. Tissue Eng. Regen. Med. 2020, 14, 424–440. [Google Scholar] [CrossRef]
  60. Cervelli, V.; Gentile, P.; De Angelis, B.; Calabrese, C.; Di Stefani, A.; Scioli, M.G.; Curcio, B.C.; Felici, M.; Orlandi, A. Application of enhanced stromal vascular fraction and fat grafting mixed with PRP in post-traumatic lower extremity ulcers. Stem Cell Res. 2011, 6, 103–111. [Google Scholar] [CrossRef]
  61. Grada, A.; Mervis, J.; Falanga, V. Research Techniques Made Simple: Animal Models of Wound Healing. J. Investig. Dermatol. 2018, 138, 2095–2105. [Google Scholar] [CrossRef] [Green Version]
  62. Ud-Din, S.; Bayat, A. Non-animal models of wound healing in cutaneous repair: In silico, in vitro, ex vivo, and in vivo models of wounds and scars in human skin: Models of wound repair in human skin. Wound Rep. Reg. 2017, 25, 164–176. [Google Scholar] [CrossRef]
  63. Lindblad, W.J. Considerations for selecting the correct animal model for dermal wound-healing studies. J. Biomater. Sci. Polym. Ed. 2008, 19, 1087–1096. [Google Scholar] [CrossRef] [PubMed]
  64. Nunan, R.; Harding, K.G.; Martin, P. Clinical challenges of chronic wounds: Searching for an optimal animal model to recapitulate their complexity. Dis. Model. Mech. 2014, 7, 1205–1213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Powers, J.G.; Higham, C.; Broussard, K.; Phillips, T.J. Wound healing and treating wounds. J. Am. Acad. Dermatol. 2016, 74, 607–625. [Google Scholar] [CrossRef]
  66. Wong, V.W.; Sorkin, M.; Glotzbach, J.P.; Longaker, M.T.; Gurtner, G.C. Surgical Approaches to Create Murine Models of Human Wound Healing. J. Biomed. Biotechnol. 2011, 2011, 1–8. [Google Scholar] [CrossRef] [PubMed]
  67. Kim, J.H.; Martins-Green, M. Protocol to Create Chronic Wounds in Diabetic Mice. JoVE 2019, 57656. [Google Scholar] [CrossRef]
  68. Chien, S.; Wilhelmi, B.J. A Simplified Technique for Producing an Ischemic Wound Model. JoVE 2012, 3341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Trujillo, A.N.; Kesl, S.L.; Sherwood, J.; Wu, M.; Gould, L.J. Demonstration of the Rat Ischemic Skin Wound Model. JoVE 2015, 52637. [Google Scholar] [CrossRef] [Green Version]
  70. King, A.J. The use of animal models in diabetes research: Animal models of diabetes. Br. J. Pharmacol. 2012, 166, 877–894. [Google Scholar] [CrossRef] [Green Version]
  71. Galiano, R.D.; Michaels, V.J.; Dobryansky, M.; Levine, J.P.; Gurtner, G.C. Quantitative and reproducible murine model of excisional wound healing. Wound Repair Regen. 2004, 12, 485–492. [Google Scholar] [CrossRef]
  72. Dunn, L.; Prosser, H.C.G.; Tan, J.T.M.; Vanags, L.Z.; Ng, M.K.C.; Bursill, C.A. Murine Model of Wound Healing. JoVE 2013, 75, e50265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Laschke, M.; Menger, M. The dorsal skinfold chamber: Window into the dynamic interaction of biomaterials with their surrounding host tissue. eCM 2011, 22, 147–167. [Google Scholar] [CrossRef] [PubMed]
  74. Sckell, A.; Leunig, M. Dorsal Skinfold Chamber Preparation in Mice: Studying Angiogenesis by Intravital Microscopy. In Angiogenesis Protocols; Martin, S.G., Hewett, P.W., Eds.; Methods in Molecular Biology; Springer: New York, NY, USA, 2016; Volume 1430, pp. 251–263. ISBN 978-1-4939-3626-7. [Google Scholar]
  75. Gurtner, G.C.; Werner, S.; Barrandon, Y.; Longaker, M.T. Wound repair and regeneration. Nature 2008, 453, 314–321. [Google Scholar] [CrossRef] [PubMed]
  76. Shultz, L.D.; Schweitzer, P.A.; Christianson, S.W.; Gott, B.; Schweitzer, I.B.; Tennent, B.; McKenna, S.; Mobraaten, L.; Rajan, T.V.; Greiner, D.L. Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J. Immunol. 1995, 154, 180–191. [Google Scholar] [PubMed]
  77. Shultz, L.D.; Lyons, B.L.; Burzenski, L.M.; Gott, B.; Chen, X.; Chaleff, S.; Kotb, M.; Gillies, S.D.; King, M.; Mangada, J.; et al. Human Lymphoid and Myeloid Cell Development in NOD/LtSz-scid IL2R γ null Mice Engrafted with Mobilized Human Hemopoietic Stem Cells. J. Immunol. 2005, 174, 6477–6489. [Google Scholar] [CrossRef] [Green Version]
  78. Shultz, L.D.; Brehm, M.A.; Garcia-Martinez, J.V.; Greiner, D.L. Humanized mice for immune system investigation: Progress, promise and challenges. Nat. Rev. Immunol. 2012, 12, 786–798. [Google Scholar] [CrossRef]
  79. Seaton, M.; Hocking, A.; Gibran, N.S. Porcine Models of Cutaneous Wound Healing. ILAR J. 2015, 56, 127–138. [Google Scholar] [CrossRef]
  80. Sullivan, T.P.; Eaglstein, W.H.; Davis, S.C.; Mertz, P. The pig as a model for human wound healing. Wound Repair Regen. 2001, 9, 66–76. [Google Scholar] [CrossRef]
  81. Pabst, R. The pig as a model for immunology research. Cell Tissue Res. 2020, 380, 287–304. [Google Scholar] [CrossRef] [PubMed]
  82. Mair, K.H.; Sedlak, C.; Käser, T.; Pasternak, A.; Levast, B.; Gerner, W.; Saalmüller, A.; Summerfield, A.; Gerdts, V.; Wilson, H.L.; et al. The porcine innate immune system: An update. Dev. Comp. Immunol. 2014, 45, 321–343. [Google Scholar] [CrossRef] [PubMed]
  83. Summerfield, A.; Meurens, F.; Ricklin, M.E. The immunology of the porcine skin and its value as a model for human skin. Mol. Immunol. 2015, 66, 14–21. [Google Scholar] [CrossRef] [PubMed]
  84. Soehnlein, O.; Steffens, S.; Hidalgo, A.; Weber, C. Neutrophils as protagonists and targets in chronic inflammation. Nat. Rev. Immunol. 2017, 17, 248–261. [Google Scholar] [CrossRef] [PubMed]
  85. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef] [PubMed]
  86. Kovtun, A.; Messerer, D.a.C.; Scharffetter-Kochanek, K.; Huber-Lang, M.; Ignatius, A. Neutrophils in Tissue Trauma of the Skin, Bone, and Lung: Two Sides of the Same Coin. J. Immunol. Res. 2018, 2018, 8173983. [Google Scholar] [CrossRef]
  87. Goralski, K.B.; McCarthy, T.C.; Hanniman, E.A.; Zabel, B.A.; Butcher, E.C.; Parlee, S.D.; Muruganandan, S.; Sinal, C.J. Chemerin, a Novel Adipokine That Regulates Adipogenesis and Adipocyte Metabolism. J. Biol. Chem. 2007, 282, 28175–28188. [Google Scholar] [CrossRef] [Green Version]
  88. Cash, J.L.; Christian, A.R.; Greaves, D.R. Chemerin Peptides Promote Phagocytosis in a ChemR23- and Syk-Dependent Manner. J. Immunol. 2010, 184, 5315–5324. [Google Scholar] [CrossRef] [Green Version]
  89. Cash, J.L.; Bena, S.; Headland, S.E.; McArthur, S.; Brancaleone, V.; Perretti, M. Chemerin15 inhibits neutrophil-mediated vascular inflammation and myocardial ischemia-reperfusion injury through ChemR23. EMBO. Rep. 2013, 14, 999–1007. [Google Scholar] [CrossRef]
  90. Krzyszczyk, P.; Schloss, R.; Palmer, A.; Berthiaume, F. The Role of Macrophages in Acute and Chronic Wound Healing and Interventions to Promote Pro-wound Healing Phenotypes. Front. Physiol. 2018, 9, 419. [Google Scholar] [CrossRef]
  91. Theret, M.; Mounier, R.; Rossi, F. The origins and non-canonical functions of macrophages in development and regeneration. Development 2019, 146. [Google Scholar] [CrossRef] [Green Version]
  92. Boniakowski, A.E.; Kimball, A.S.; Joshi, A.; Schaller, M.; Davis, F.M.; denDekker, A.; Obi, A.T.; Moore, B.B.; Kunkel, S.L.; Gallagher, K.A. Macrophage chemokine receptor CCR2 plays a crucial role in macrophage recruitment and regulated inflammation in wound healing. Eur. J. Immunol. 2018, 48, 1445–1455. [Google Scholar] [CrossRef]
  93. Kimball, A.; Schaller, M.; Joshi, A.; Davis Frank, M.; denDekker, A.; Boniakowski, A.; Bermick, J.; Obi, A.; Moore, B.; Henke Peter, K.; et al. Ly6CHi Blood Monocyte/Macrophage Drive Chronic Inflammation and Impair Wound Healing in Diabetes Mellitus. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 1102–1114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Arnold, L.; Henry, A.; Poron, F.; Baba-Amer, Y.; van Rooijen, N.; Plonquet, A.; Gherardi, R.K.; Chazaud, B. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. J. Exp. Med. 2007, 204, 1057–1069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Willenborg, S.; Lucas, T.; van Loo, G.; Knipper, J.A.; Krieg, T.; Haase, I.; Brachvogel, B.; Hammerschmidt, M.; Nagy, A.; Ferrara, N.; et al. CCR2 recruits an inflammatory macrophage subpopulation critical for angiogenesis in tissue repair. Blood 2012, 120, 613–625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Varga, T.; Mounier, R.; Gogolak, P.; Poliska, S.; Chazaud, B.; Nagy, L. Tissue LyC6- macrophages are generated in the absence of circulating LyC6- monocytes and Nur77 in a model of muscle regeneration. J. Immunol. 2013, 191, 5695–5701. [Google Scholar] [CrossRef] [Green Version]
  97. Snyder, R.J.; Lantis, J.; Kirsner, R.S.; Shah, V.; Molyneaux, M.; Carter, M.J. Macrophages: A review of their role in wound healing and their therapeutic use. Wound Repair Regen. 2016, 24, 613–629. [Google Scholar] [CrossRef]
  98. Lumeng, C.N.; Bodzin, J.L.; Saltiel, A.R. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J. Clin. Invest. 2007, 117, 175–184. [Google Scholar] [CrossRef] [Green Version]
  99. Thomas, D.; Apovian, C. Macrophage functions in lean and obese adipose tissue. Metab. Clin. Exp. 2017, 72, 120–143. [Google Scholar] [CrossRef]
  100. Torres-Castro, I.; Arroyo-Camarena, Ú.D.; Martínez-Reyes, C.P.; Gómez-Arauz, A.Y.; Dueñas-Andrade, Y.; Hernández-Ruiz, J.; Béjar, Y.L.; Zaga-Clavellina, V.; Morales-Montor, J.; Terrazas, L.I.; et al. Human monocytes and macrophages undergo M1-type inflammatory polarization in response to high levels of glucose. Immunol. Lett. 2016, 176, 81–89. [Google Scholar] [CrossRef]
  101. Dachir, S.; Cohen, M.; Sahar, R.; Graham, J.; Eisenkraft, A.; Horwitz, V.; Kadar, T. Beneficial effects of activated macrophages on sulfur mustard-induced cutaneous burns, an in vivo experience. Cutan. Ocul. Toxicol. 2014, 33, 317–326. [Google Scholar] [CrossRef]
  102. Li, M.; Wang, T.; Tian, H.; Wei, G.; Zhao, L.; Shi, Y. Macrophage-derived exosomes accelerate wound healing through their anti-inflammation effects in a diabetic rat model. Artif. Cellsnanomedicineand. Biotechnol. 2019, 47, 3793–3803. [Google Scholar] [CrossRef] [Green Version]
  103. Clausen, B.E.; Stoitzner, P. Functional Specialization of Skin Dendritic Cell Subsets in Regulating T Cell Responses. Front. Immunol. 2015, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Kim, T.-G.; Kim, S.H.; Lee, M.-G. The Origin of Skin Dendritic Cell Network and Its Role in Psoriasis. Int. J. Mol. Sci. 2017, 19. [Google Scholar] [CrossRef] [Green Version]
  105. Kaiko, G.E.; Horvat, J.C.; Beagley, K.W.; Hansbro, P.M. Immunological decision-making: How does the immune system decide to mount a helper T-cell response? Immunology 2008, 123, 326–338. [Google Scholar] [CrossRef] [PubMed]
  106. Landén, N.X.; Li, D.; Ståhle, M. Transition from inflammation to proliferation: A critical step during wound healing. Cell. Mol. Life Sci. 2016, 73, 3861–3885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Gregorio, J.; Meller, S.; Conrad, C.; Di Nardo, A.; Homey, B.; Lauerma, A.; Arai, N.; Gallo, R.L.; DiGiovanni, J.; Gilliet, M. Plasmacytoid dendritic cells sense skin injury and promote wound healing through type I interferons. J. Exp. Med. 2010, 207, 2921–2930. [Google Scholar] [CrossRef] [PubMed]
  108. Boyce, D.E.; Jones, W.D.; Ruge, F.; Harding, K.G.; Moore, K. The role of lymphocytes in human dermal wound healing. Br. J. Dermatol. 2000, 143, 59–65. [Google Scholar] [CrossRef]
  109. Cruz, M.S.; Diamond, A.; Russell, A.; Jameson, J.M. Human αβ and γδ T Cells in Skin Immunity and Disease. Front. Immunol. 2018, 9, 1304. [Google Scholar] [CrossRef]
  110. Sumaria, N.; Roediger, B.; Ng, L.G.; Qin, J.; Pinto, R.; Cavanagh, L.L.; Shklovskaya, E.; Fazekas de St. Groth, B.; Triccas, J.A.; Weninger, W. Cutaneous immunosurveillance by self-renewing dermal γδ T cells. J. Exp. Med. 2011, 208, 505–518. [Google Scholar] [CrossRef] [Green Version]
  111. Jameson, J.M.; Cauvi, G.; Witherden, D.A.; Havran, W.L. A Keratinocyte-Responsive γδ TCR Is Necessary for Dendritic Epidermal T Cell Activation by Damaged Keratinocytes and Maintenance in the Epidermis. J. Immunol. 2004, 172, 3573–3579. [Google Scholar] [CrossRef] [Green Version]
  112. Jameson, J.M.; Cauvi, G.; Sharp, L.L.; Witherden, D.A.; Havran, W.L. γδ T cell–induced hyaluronan production by epithelial cells regulates inflammation. J. Exp. Med. 2005, 201, 1269–1279. [Google Scholar] [CrossRef] [Green Version]
  113. Chodaczek, G.; Papanna, V.; Zal, M.A.; Zal, T. Body-barrier surveillance by epidermal γδ TCRs. Nat. Immunol. 2012, 13, 272–282. [Google Scholar] [CrossRef]
  114. Havran, W.L.; Jameson, J.M. Epidermal T Cells and Wound Healing. J. Immunol. 2010, 184, 5423–5428. [Google Scholar] [CrossRef]
  115. Cai, Y.; Shen, X.; Ding, C.; Qi, C.; Li, K.; Li, X.; Jala, V.R.; Zhang, H.; Wang, T.; Zheng, J.; et al. Pivotal Role of Dermal IL-17-Producing γδ T Cells in Skin Inflammation. Immunity 2011, 35, 596–610. [Google Scholar] [CrossRef] [Green Version]
  116. Li, Y.; Wang, Y.; Zhou, L.; Liu, M.; Liang, G.; Yan, R.; Jiang, Y.; Hao, J.; Zhang, X.; Hu, X.; et al. Vγ4 T Cells Inhibit the Pro-healing Functions of Dendritic Epidermal T Cells to Delay Skin Wound Closure Through IL-17A. Front. Immunol. 2018, 9, 240. [Google Scholar] [CrossRef] [Green Version]
  117. Li, Y.; Wu, J.; Luo, G.; He, W. Functions of Vγ4 T Cells and Dendritic Epidermal T Cells on Skin Wound Healing. Front. Immunol. 2018, 9, 1099. [Google Scholar] [CrossRef] [Green Version]
  118. Liu, Z.; Liang, G.; Gui, L.; Li, Y.; Liu, M.; bai, Y.; Zhang, X.; Hu, X.; Chen, J.; Huang, C.; et al. Weakened IL-15 Production and Impaired mTOR Activation Alter Dendritic Epidermal T Cell Homeostasis in Diabetic Mice. Sci. Rep. 2017, 7, 6028. [Google Scholar] [CrossRef] [Green Version]
  119. Liu, Z.; Xu, Y.; Zhang, X.; Liang, G.; Chen, L.; Xie, J.; Tang, J.; Zhao, J.; Shu, B.; Qi, S.; et al. Defects in dermal Vγ4 γ δ T cells result in delayed wound healing in diabetic mice. Am. J. Transl. Res. 2016, 8, 2667–2680. [Google Scholar]
  120. MacLeod, A.S.; Hemmers, S.; Garijo, O.; Chabod, M.; Mowen, K.; Witherden, D.A.; Havran, W.L. Dendritic epidermal T cells regulate skin antimicrobial barrier function. J. Clin. Invest. 2013, 123, 4364–4374. [Google Scholar] [CrossRef]
  121. Clark, R.A. Skin-Resident T Cells: The Ups and Downs of On Site Immunity. J. Investig. Dermatol. 2010, 130, 362–370. [Google Scholar] [CrossRef] [Green Version]
  122. Toulon, A.; Breton, L.; Taylor, K.R.; Tenenhaus, M.; Bhavsar, D.; Lanigan, C.; Rudolph, R.; Jameson, J.; Havran, W.L. A role for human skin–resident T cells in wound healing. J. Exp. Med. 2009, 206, 743–750. [Google Scholar] [CrossRef] [Green Version]
  123. Adachi, T.; Kobayashi, T.; Sugihara, E.; Yamada, T.; Ikuta, K.; Pittaluga, S.; Saya, H.; Amagai, M.; Nagao, K. Hair follicle-derived IL-7 and IL-15 mediate skin-resident memory T cell homeostasis and lymphoma. Nat. Med. 2015, 21, 1272–1279. [Google Scholar] [CrossRef]
  124. Watanabe, R.; Gehad, A.; Yang, C.; Scott, L.L.; Teague, J.E.; Schlapbach, C.; Elco, C.P.; Huang, V.; Matos, T.R.; Kupper, T.S.; et al. Human skin is protected by four functionally and phenotypically discrete populations of resident and recirculating memory T cells. Sci. Transl. Med. 2015, 7, 279ra39. [Google Scholar] [CrossRef] [Green Version]
  125. Chen, L.; Mehta, N.D.; Zhao, Y.; DiPietro, L.A. Absence of CD4 or CD8 lymphocytes changes infiltration of inflammatory cells and profiles of cytokine expression in skin wounds, but does not impair healing. Exp. Derm. 2014, 23, 189–194. [Google Scholar] [CrossRef] [Green Version]
  126. Li, J.; Liang, C.; Yang, K.Y.; Huang, X.; Han, M.Y.; Li, X.; Chan, V.W.; Chan, K.S.; Liu, D.; Huang, Z.-P.; et al. Specific ablation of CD4+ T-cells promotes heart regeneration in juvenile mice. Theranostics 2020, 10, 8018–8035. [Google Scholar] [CrossRef]
  127. Leung, O.M.; Li, J.; Li, X.; Chan, V.W.; Yang, K.Y.; Ku, M.; Ji, L.; Sun, H.; Waldmann, H.; Tian, X.Y.; et al. Regulatory T Cells Promote Apelin-Mediated Sprouting Angiogenesis in Type 2 Diabetes. Cell Rep. 2018, 24, 1610–1626. [Google Scholar] [CrossRef] [Green Version]
  128. Liang, C.; Yang, K.Y.; Chan, V.W.; Li, X.; Fung, T.H.W.; Wu, Y.; Tian, X.Y.; Huang, Y.; Qin, L.; Lau, J.Y.W.; et al. CD8+ T-cell plasticity regulates vascular regeneration in type-2 diabetes. Theranostics 2020, 10, 4217–4232. [Google Scholar] [CrossRef]
  129. Fejfarová, V.; Jirkovská, A.; Dubský, M.; Game, F.; Vydláková, J.; Sekerková, A.; Franeková, J.; Kučerová, M.; Stříž, I.; Petkov, V.; et al. An Alteration of Lymphocytes Subpopulations and Immunoglobulins Levels in Patients with Diabetic Foot Ulcers Infected Particularly by Resistant Pathogens. J. Diabetes Res. 2016, 2016, 1–9. [Google Scholar] [CrossRef]
  130. Moura, J.; Rodrigues, J.; Gonçalves, M.; Amaral, C.; Lima, M.; Carvalho, E. Impaired T-cell differentiation in diabetic foot ulceration. Cell Mol. Immunol. 2017, 14, 758–769. [Google Scholar] [CrossRef] [Green Version]
  131. Zacchigna, S.; Martinelli, V.; Moimas, S.; Colliva, A.; Anzini, M.; Nordio, A.; Costa, A.; Rehman, M.; Vodret, S.; Pierro, C.; et al. Paracrine effect of regulatory T cells promotes cardiomyocyte proliferation during pregnancy and after myocardial infarction. Nat. Commun. 2018, 9, 2432. [Google Scholar] [CrossRef]
  132. Li, J.; Yang, K.Y.; Tam, R.C.Y.; Chan, V.W.; Lan, H.Y.; Hori, S.; Zhou, B.; Lui, K.O. Regulatory T-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner. Theranostics 2019, 9, 4324–4341. [Google Scholar] [CrossRef]
  133. Sanchez Rodriguez, R.; Pauli, M.L.; Neuhaus, I.M.; Yu, S.S.; Arron, S.T.; Harris, H.W.; Yang, S.H.-Y.; Anthony, B.A.; Sverdrup, F.M.; Krow-Lucal, E.; et al. Memory regulatory T cells reside in human skin. J. Clin. Invest. 2014, 124, 1027–1036. [Google Scholar] [CrossRef] [Green Version]
  134. Gratz, I.K.; Truong, H.-A.; Yang, S.H.-Y.; Maurano, M.M.; Lee, K.; Abbas, A.K.; Rosenblum, M.D. Cutting Edge: Memory Regulatory T Cells Require IL-7 and Not IL-2 for Their Maintenance in Peripheral Tissues. J. Immunol. 2013, 190, 4483–4487. [Google Scholar] [CrossRef] [Green Version]
  135. Nosbaum, A.; Prevel, N.; Truong, H.-A.; Mehta, P.; Ettinger, M.; Scharschmidt, T.C.; Ali, N.H.; Pauli, M.L.; Abbas, A.K.; Rosenblum, M.D. Cutting Edge: Regulatory T Cells Facilitate Cutaneous Wound Healing. J. Immunol. 2016, 196, 2010–2014. [Google Scholar] [CrossRef]
  136. Barros, J.F.; Waclawiak, I.; Pecli, C.; Borges, P.A.; Georgii, J.L.; Ramos-Junior, E.S.; Canetti, C.; Courau, T.; Klatzmann, D.; Kunkel, S.L.; et al. Role of Chemokine Receptor CCR4 and Regulatory T Cells in Wound Healing of Diabetic Mice. J. Investig. Dermatol. 2019, 139, 1161–1170. [Google Scholar] [CrossRef]
  137. Haertel, E.; Joshi, N.; Hiebert, P.; Kopf, M.; Werner, S. Regulatory T cells are required for normal and activin-promoted wound repair in mice. Eur. J. Immunol. 2018, 48, 1001–1013. [Google Scholar] [CrossRef] [Green Version]
  138. Weirather, J.; Hofmann, U.D.W.; Beyersdorf, N.; Ramos, G.C.; Vogel, B.; Frey, A.; Ertl, G.; Kerkau, T.; Frantz, S. Foxp3+ CD4+ T cells improve healing after myocardial infarction by modulating monocyte/macrophage differentiation. Circ. Res. 2014, 115, 55–67. [Google Scholar] [CrossRef]
  139. Richards, A.M.; Floyd, D.C.; Terenghi, G.; McGrouther, D.A. Cellular changes in denervated tissue during wound healing in a rat model. Br. J. Dermatol. 1999, 140, 1093–1099. [Google Scholar] [CrossRef]
  140. Iwata, Y.; Yoshizaki, A.; Komura, K.; Shimizu, K.; Ogawa, F.; Hara, T.; Muroi, E.; Bae, S.; Takenaka, M.; Yukami, T.; et al. CD19, a Response Regulator of B Lymphocytes, Regulates Wound Healing through Hyaluronan-Induced TLR4 Signaling. Am. J. Pathol. 2009, 175, 649–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. He, H.; Tang, L.; Jiang, N.; Zheng, R.; Li, W.; Gu, Y.; Wang, M. Characterization of peripheral blood mononuclear cells isolated using two kinds of leukocyte filters. Transfus. Clin. Et Biol. 2020, 27, 10–17. [Google Scholar] [CrossRef] [PubMed]
  142. Grievink, H.W.; Luisman, T.; Kluft, C.; Moerland, M.; Malone, K.E. Comparison of Three Isolation Techniques for Human Peripheral Blood Mononuclear Cells: Cell Recovery and Viability, Population Composition, and Cell Functionality. Biopreservation Biobanking 2016, 14, 410–415. [Google Scholar] [CrossRef]
  143. Spaltro, G.; Straino, S.; Gambini, E.; Bassetti, B.; Persico, L.; Zoli, S.; Zanobini, M.; Capogrossi, M.C.; Spirito, R.; Quarti, C.; et al. Characterization of the Pall Celeris system as a point-of-care device for therapeutic angiogenesis. Cytotherapy 2015, 17, 1302–1313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Holzinger, C.; Zuckermann, A.; Kopp, C.; Schöllhammer, A.; Imhof, M.; Zwölfer, W.; Baumgartner, I.; Magometschnigg, H.; Weissinger, E.; Wolner, E. Treatment of non-healing skin ulcers with autologous activated mononuclear cells. Eur. J. Vasc. Surg. 1994, 8, 351–356. [Google Scholar] [CrossRef]
  145. Thum, T.; Bauersachs, J.; Poole-Wilson, P.A.; Volk, H.-D.; Anker, S.D. The dying stem cell hypothesis: Immune modulation as a novel mechanism for progenitor cell therapy in cardiac muscle. J. Am. Coll. Cardiol. 2005, 46, 1799–1802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Ankersmit, H.J.; Hoetzenecker, K.; Dietl, W.; Soleiman, A.; Horvat, R.; Wolfsberger, M.; Gerner, C.; Hacker, S.; Mildner, M.; Moser, B.; et al. Irradiated cultured apoptotic peripheral blood mononuclear cells regenerate infarcted myocardium. Eur. J. Clin. Investig. 2009, 39, 445–456. [Google Scholar] [CrossRef] [PubMed]
  147. Lichtenauer, M.; Mildner, M.; Hoetzenecker, K.; Zimmermann, M.; Podesser, B.K.; Sipos, W.; Berényi, E.; Dworschak, M.; Tschachler, E.; Gyöngyösi, M.; et al. Secretome of apoptotic peripheral blood cells (APOSEC) confers cytoprotection to cardiomyocytes and inhibits tissue remodelling after acute myocardial infarction: A preclinical study. Basic Res. Cardiol. 2011, 106, 1283–1297. [Google Scholar] [CrossRef] [Green Version]
  148. Beer, L.; Mildner, M.; Gyöngyösi, M.; Ankersmit, H.J. Peripheral blood mononuclear cell secretome for tissue repair. Apoptosis 2016, 21, 1336–1353. [Google Scholar] [CrossRef] [Green Version]
  149. Asahara, T.; Murohara, T.; Sullivan, A.; Silver, M.; van der Zee, R.; Li, T.; Witzenbichler, B.; Schatteman, G.; Isner, J.M. Isolation of putative progenitor endothelial cells for angiogenesis. Science 1997, 275, 964–967. [Google Scholar] [CrossRef]
  150. Kaushik, K.; Das, A. Endothelial progenitor cell therapy for chronic wound tissue regeneration. Cytotherapy 2019, 21, 1137–1150. [Google Scholar] [CrossRef]
  151. Chopra, H.; Hung, M.K.; Kwong, D.L.; Zhang, C.F.; Pow, E.H.N. Insights into Endothelial Progenitor Cells: Origin, Classification, Potentials, and Prospects. Stem Cells Int. 2018, 2018, 9847015. [Google Scholar] [CrossRef]
  152. Medina, R.J.; Barber, C.L.; Sabatier, F.; Dignat-George, F.; Melero-Martin, J.M.; Khosrotehrani, K.; Ohneda, O.; Randi, A.M.; Chan, J.K.Y.; Yamaguchi, T.; et al. Endothelial Progenitors: A Consensus Statement on Nomenclature. Stem Cells Transl. Med. 2017, 6, 1316–1320. [Google Scholar] [CrossRef]
  153. Bailey, A.S.; Willenbring, H.; Jiang, S.; Anderson, D.A.; Schroeder, D.A.; Wong, M.H.; Grompe, M.; Fleming, W.H. Myeloid lineage progenitors give rise to vascular endothelium. Proc. Natl. Acad. Sci. USA 2006, 103, 13156–13161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Medina, R.J.; O’Neill, C.L.; O’Doherty, T.M.; Knott, H.; Guduric-Fuchs, J.; Gardiner, T.A.; Stitt, A.W. Myeloid angiogenic cells act as alternative M2 macrophages and modulate angiogenesis through interleukin-8. Mol. Med. 2011, 17, 1045–1055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Lin, R.-Z.; Moreno-Luna, R.; Muñoz-Hernandez, R.; Li, D.; Jaminet, S.-C.S.; Greene, A.K.; Melero-Martin, J.M. Human white adipose tissue vasculature contains endothelial colony-forming cells with robust in vivo vasculogenic potential. Angiogenesis 2013, 16, 735–744. [Google Scholar] [CrossRef] [Green Version]
  156. Aicher, A.; Rentsch, M.; Sasaki, K.-I.; Ellwart Joachim, W.; Fändrich, F.; Siebert, R.; Cooke, J.P.; Dimmeler, S.; Heeschen, C. Nonbone Marrow-Derived Circulating Progenitor Cells Contribute to Postnatal Neovascularization Following Tissue Ischemia. Circ. Res. 2007, 100, 581–589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Timmermans, F.; Van Hauwermeiren, F.; De Smedt, M.; Raedt, R.; Plasschaert, F.; De Buyzere, M.L.; Gillebert, T.C.; Plum, J.; Vandekerckhove, B. Endothelial Outgrowth Cells Are Not Derived From CD133+ Cells or CD45+ Hematopoietic Precursors. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 1572–1579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Fujisawa, T.; Tura-Ceide, O.; Hunter, A.; Mitchell, A.; Vesey, A.; Medine, C.; Gallogly, S.; Hadoke, P.W.F.; Keith, C.; Sproul, A.; et al. Endothelial Progenitor Cells Do Not Originate From the Bone Marrow. Circulation 2019, 140, 1524–1526. [Google Scholar] [CrossRef]
  159. Keighron, C.; Lyons, C.J.; Creane, M.; O’Brien, T.; Liew, A. Recent Advances in Endothelial Progenitor Cells Toward Their Use in Clinical Translation. Front. Med. (Lausanne) 2018, 5. [Google Scholar] [CrossRef] [Green Version]
  160. Matsubara, Y.; Matsubara, K. Estrogen and progesterone play pivotal roles in endothelial progenitor cell proliferation. Reprod. Biol. Endocrinol. 2012, 10, 2. [Google Scholar] [CrossRef] [Green Version]
  161. Zhao, X.; Huang, L.; Yin, Y.; Fang, Y.; Zhao, J.; Chen, J. Estrogen induces endothelial progenitor cells proliferation and migration by estrogen receptors and PI3K-dependent pathways. Microvasc. Res. 2008, 75, 45–52. [Google Scholar] [CrossRef] [PubMed]
  162. Gilliver, S.C.; Ashworth, J.J.; Ashcroft, G.S. The hormonal regulation of cutaneous wound healing. Clin. Dermatol. 2007, 25, 56–62. [Google Scholar] [CrossRef]
  163. Zhou, T.; Yang, Z.; Chen, Y.; Chen, Y.; Huang, Z.; You, B.; Peng, Y.; Chen, J. Estrogen Accelerates Cutaneous Wound Healing by Promoting Proliferation of Epidermal Keratinocytes via Erk/Akt Signaling Pathway. Cell. Physiol. Biochem. 2016, 38, 959–968. [Google Scholar] [CrossRef] [PubMed]
  164. Stevenson, S.; Nelson, L.D.; Sharpe, D.T.; Thornton, M.J. 17β-Estradiol regulates the secretion of TGF-β by cultured human dermal fibroblasts. J. Biomater. Sci. Polym. Ed. 2008, 19, 1097–1109. [Google Scholar] [CrossRef] [PubMed]
  165. Wang, C.; Wang, Q.; Gao, W.; Zhang, Z.; Lou, Y.; Jin, H.; Chen, X.; Lei, B.; Xu, H.; Mao, C. Highly efficient local delivery of endothelial progenitor cells significantly potentiates angiogenesis and full-thickness wound healing. Acta Biomater. 2018, 69, 156–169. [Google Scholar] [CrossRef] [PubMed]
  166. Sun Yuan; Chen Song; Zhang Xicheng; Pei Ming Significance of Cellular Cross-Talk in Stromal Vascular Fraction of Adipose Tissue in Neovascularization. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 1034–1044. [CrossRef] [PubMed]
  167. Ramakrishnan, V.M.; Boyd, N.L. The Adipose Stromal Vascular Fraction as a Complex Cellular Source for Tissue Engineering Applications. Tissue Eng. Part B Rev. 2018, 24, 289–299. [Google Scholar] [CrossRef] [PubMed]
  168. Bora, P.; Majumdar, A.S. Adipose tissue-derived stromal vascular fraction in regenerative medicine: A brief review on biology and translation. Stem Cell Res. 2017, 8, 145. [Google Scholar] [CrossRef]
  169. Traktuev, D.O.; Prater, D.N.; Merfeld-Clauss, S.; Sanjeevaiah, A.R.; Saadatzadeh, M.R.; Murphy, M.; Johnstone, B.H.; Ingram, D.A.; March, K.L. Robust Functional Vascular Network Formation In Vivo by Cooperation of Adipose Progenitor and Endothelial Cells. Circ. Res. 2009, 104, 1410–1420. [Google Scholar] [CrossRef]
  170. Klar, A.S.; Güven, S.; Zimoch, J.; Zapiórkowska, N.A.; Biedermann, T.; Böttcher-Haberzeth, S.; Meuli-Simmen, C.; Martin, I.; Scherberich, A.; Reichmann, E.; et al. Characterization of vasculogenic potential of human adipose-derived endothelial cells in a three-dimensional vascularized skin substitute. Pediatr. Surg. Int. 2016, 32, 17–27. [Google Scholar] [CrossRef]
  171. Sack, B.S.; Mauney, J.R.; Estrada, C.R. Silk fibroin scaffolds for urologic tissue engineering. Curr. Urol. Rep. 2016, 17, 16. [Google Scholar] [CrossRef]
  172. Navone, S.E.; Pascucci, L.; Dossena, M.; Ferri, A.; Invernici, G.; Acerbi, F.; Cristini, S.; Bedini, G.; Tosetti, V.; Ceserani, V.; et al. Decellularized silk fibroin scaffold primed with adipose mesenchymal stromal cells improves wound healing in diabetic mice. Stem Cell Res. Ther. 2014, 5, 7. [Google Scholar] [CrossRef] [Green Version]
  173. Bi, H.; Li, H.; Zhang, C.; Mao, Y.; Nie, F.; Xing, Y.; Sha, W.; Wang, X.; Irwin, D.M.; Tan, H. Stromal vascular fraction promotes migration of fibroblasts and angiogenesis through regulation of extracellular matrix in the skin wound healing process. Stem Cell Res. Ther. 2019, 10, 302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Hu, L.; Wang, J.; Zhou, X.; Xiong, Z.; Zhao, J.; Yu, R.; Huang, F.; Zhang, H.; Chen, L. Exosomes derived from human adipose mensenchymal stem cells accelerates cutaneous wound healing via optimizing the characteristics of fibroblasts. Sci. Rep. 2016, 6, 32933. [Google Scholar] [CrossRef] [PubMed]
  175. Bowles, A.C.; Wise, R.M.; Gerstein, B.Y.; Thomas, R.C.; Ogelman, R.; Febbo, I.; Bunnell, B.A. Immunomodulatory Effects of Adipose Stromal Vascular Fraction Cells Promote Alternative Activation Macrophages to Repair Tissue Damage. STEM CELLS 2017, 35, 2198–2207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Deng, C.; He, Y.; Feng, J.; Dong, Z.; Yao, Y.; Lu, F. Conditioned medium from 3D culture system of stromal vascular fraction cells accelerates wound healing in diabetic rats. Regen. Med. 2019, 14, 925–937. [Google Scholar] [CrossRef]
  177. Sun, M.; He, Y.; Zhou, T.; Zhang, P.; Gao, J.; Lu, F. Adipose Extracellular Matrix/Stromal Vascular Fraction Gel Secretes Angiogenic Factors and Enhances Skin Wound Healing in a Murine Model. Available online: https://www.hindawi.com/journals/bmri/2017/3105780/ (accessed on 26 May 2020).
  178. Chae, D.-S.; Han, S.; Son, M.; Kim, S.-W. Stromal vascular fraction shows robust wound healing through high chemotactic and epithelialization property. Cytotherapy 2017, 19, 543–554. [Google Scholar] [CrossRef]
  179. Koh, Y.J.; Koh, B.I.; Kim, H.; Joo, H.J.; Jin, H.K.; Jeon, J.; Choi, C.; Lee, D.H.; Chung, J.H.; Cho, C.H.; et al. Stromal vascular fraction from adipose tissue forms profound vascular network through the dynamic reassembly of blood endothelial cells. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 1141–1150. [Google Scholar] [CrossRef] [Green Version]
  180. Burl, R.B.; Ramseyer, V.D.; Rondini, E.A.; Pique-Regi, R.; Lee, Y.-H.; Granneman, J.G. Deconstructing Adipogenesis Induced by β3-Adrenergic Receptor Activation with Single-Cell Expression Profiling. Cell Metab. 2018, 28, 300–309.e4. [Google Scholar] [CrossRef] [Green Version]
  181. Hepler, C.; Shan, B.; Zhang, Q.; Henry, G.H.; Shao, M.; Vishvanath, L.; Ghaben, A.L.; Mobley, A.B.; Strand, D.; Hon, G.C.; et al. Identification of functionally distinct fibro-inflammatory and adipogenic stromal subpopulations in visceral adipose tissue of adult mice. eLife 2018, 7, e39636. [Google Scholar] [CrossRef]
  182. Merrick, D.; Sakers, A.; Irgebay, Z.; Okada, C.; Calvert, C.; Morley, M.P.; Percec, I.; Seale, P. Identification of a mesenchymal progenitor cell hierarchy in adipose tissue. Science 2019, 364, eaav2501. [Google Scholar] [CrossRef]
  183. Rennert, R.C.; Januszyk, M.; Sorkin, M.; Rodrigues, M.; Maan, Z.N.; Duscher, D.; Whittam, A.J.; Kosaraju, R.; Chung, M.T.; Paik, K.; et al. Microfluidic single-cell transcriptional analysis rationally identifies novel surface marker profiles to enhance cell-based therapies. Nat. Commun. 2016, 7, 11945. [Google Scholar] [CrossRef]
  184. Schwalie, P.C.; Dong, H.; Zachara, M.; Russeil, J.; Alpern, D.; Akchiche, N.; Caprara, C.; Sun, W.; Schlaudraff, K.-U.; Soldati, G.; et al. A stromal cell population that inhibits adipogenesis in mammalian fat depots. Nature 2018, 559, 103–108. [Google Scholar] [CrossRef] [PubMed]
  185. Hoodless, L.J.; Lucas, C.D.; Duffin, R.; Denvir, M.A.; Haslett, C.; Tucker, C.S.; Rossi, A.G. Genetic and pharmacological inhibition of CDK9 drives neutrophil apoptosis to resolve inflammation in zebrafish in vivo. Sci. Rep. 2016, 6, 36980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Richardson, R.; Slanchev, K.; Kraus, C.; Knyphausen, P.; Eming, S.; Hammerschmidt, M. Adult Zebrafish as a Model System for Cutaneous Wound-Healing Research. J. Investig. Dermatol. 2013, 133, 1655–1665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Pastar, I.; Wong, L.L.; Egger, A.N.; Tomic-Canic, M. Descriptive vs mechanistic scientific approach to study wound healing and its inhibition: Is there a value of translational research involving human subjects? Exp. Dermatol. 2018, 27, 551–562. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The figure shows examples of cellular therapies proposed and exploited for the treatment of non-healing wounds, grouped according to four main approaches: cell mobilization into the circulation, cell recruitment to the wound site, modulation of cell function/activity, and direct application of cells to the wound site. Studies that reached the clinical stage are in bold. APOSEC, apoptotic secretome; C15, chemerin 15; DETC, dendritic epidermal T cells; Dnmt1, DNA (cytosine-5)-methyltransferase 1; DPP4, dipeptidyl peptidase-4; EPCs, endothelial progenitor cells; FL, FMS-like tyrosine kinase-3 ligand; GM-CSF, granulocyte-macrophage colony stimulating factor; IL-15, interleukin-15; MALP2, macrophage-activating lipopeptide-2; MLL1, mixed-lineage leukemia 1; PAD4, peptidyl-arginine deiminase 4; PBMCs, peripheral blood mononucleated cells; SDF1, stromal cell-derived factor-1; SVF, stromal vascular fraction.
Figure 1. The figure shows examples of cellular therapies proposed and exploited for the treatment of non-healing wounds, grouped according to four main approaches: cell mobilization into the circulation, cell recruitment to the wound site, modulation of cell function/activity, and direct application of cells to the wound site. Studies that reached the clinical stage are in bold. APOSEC, apoptotic secretome; C15, chemerin 15; DETC, dendritic epidermal T cells; Dnmt1, DNA (cytosine-5)-methyltransferase 1; DPP4, dipeptidyl peptidase-4; EPCs, endothelial progenitor cells; FL, FMS-like tyrosine kinase-3 ligand; GM-CSF, granulocyte-macrophage colony stimulating factor; IL-15, interleukin-15; MALP2, macrophage-activating lipopeptide-2; MLL1, mixed-lineage leukemia 1; PAD4, peptidyl-arginine deiminase 4; PBMCs, peripheral blood mononucleated cells; SDF1, stromal cell-derived factor-1; SVF, stromal vascular fraction.
Ijms 21 05235 g001
Table 1. Preclinical and clinical studies based on cell therapy for non-healing wounds.
Table 1. Preclinical and clinical studies based on cell therapy for non-healing wounds.
Cell TypeApproachPreclinical DataClinical Studies
NeutrophilsModulationTopical application of miR-129-2-3p on wounds of diabetic mice [28]-
Genetic inactivation of PAD4 in diabetic mice [29]-
RecruitmentTopical delivery of C15 on burn wounds in mice [30]-
Monocytes and MacrophagesRecruitment and ModulationLocal expression of SDF-1 through transformed bacteria in mice with diabetes and peripheral ischemia and ex vivo model of human skins [31] -
Recruitment-Local injection of hGM-CSF [32,33,34]
-Local application of β−glucans and MALP-2 [35,36]
Direct application-Local injection of autologous macrophages in diabetic foot [37,38]
ModulationGenetic inactivation of MLL1 and Dnmt1 in mice [39,40] -
Dendritic cellsModulationSystemic administration of FL in burn wounds in mice [41] -
Lymphocytes (DETC)ModulationIL-15 administration in diabetic mice [42] -
Lymphocytes (DETC)Direct applicationDETC engraftment in diabetic mice [43] -
Lymphocytes (CD4+ CD8+)Cell transplantation in athymic nude mice [44] and SCID mice [45] -
Lymphocytes (B cells)Topical application in diabetic mice [46] -
Peripheral blood mononuclear cellsDirect applicationApplication of cell sheets composed of fibroblasts and PBMCs in diabetic mice [47,48] Topical application of APOSEC in healthy volunteers [49]
Endothelial progenitorsRecruitmentLocal administration of recombinant SDF1 in diabetic mice [50] Pharmacological inhibition of DPP4 in diabetic wounds [51]
ModulationTopical administration of estrogens in diabetic mice [52] -
Direct applicationLocal transplantation of human EPCs in immunocompromised mice [53] or model of burn wound in pigs [54] Intra-arterial delivery of CD133+ EPCs in diabetic foot patients [55]
Mobilization and Direct application-Systemic administration of GM-CSF followed by isolation of CD34+/VEGFR2+ cells and intramuscular injection in non-healing foot in diabetic patients [56]
Stromal vascular fractionDirect applicationSVF seeding on human epidermal skin substitutes applied in nude rats [57] Direct application of autologous SVF on diabetic ulcers [58,59] or post-traumatic lower extremity ulcers [60]

Share and Cite

MDPI and ACS Style

Groppa, E.; Colliva, A.; Vuerich, R.; Kocijan, T.; Zacchigna, S. Immune Cell Therapies to Improve Regeneration and Revascularization of Non-Healing Wounds. Int. J. Mol. Sci. 2020, 21, 5235. https://doi.org/10.3390/ijms21155235

AMA Style

Groppa E, Colliva A, Vuerich R, Kocijan T, Zacchigna S. Immune Cell Therapies to Improve Regeneration and Revascularization of Non-Healing Wounds. International Journal of Molecular Sciences. 2020; 21(15):5235. https://doi.org/10.3390/ijms21155235

Chicago/Turabian Style

Groppa, Elena, Andrea Colliva, Roman Vuerich, Tea Kocijan, and Serena Zacchigna. 2020. "Immune Cell Therapies to Improve Regeneration and Revascularization of Non-Healing Wounds" International Journal of Molecular Sciences 21, no. 15: 5235. https://doi.org/10.3390/ijms21155235

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop