Abstract
As regenerative medicine matures as a field, more promising technologies are being translated from the benchtop to the clinic. However, many of these strategies are designed with otherwise healthy hosts in mind and validated in animal models without other co-morbidities. In reality, many of the patient populations benefiting from drug delivery and tissue engineering-based devices to enhance wound healing also have significant underlying immunodeficiency. Specifically, patients suffering from diabetes, malignancy, human immunodeficiency virus, post organ transplantation, and other compromised states have significant pleotropic immune defects that affect wound healing. In this work, we review the role of different immune cells in the regenerative process, highlight the effect of several common immunocompromised states on wound healing, and discuss different drug delivery strategies for overcoming immunodeficiencies.
Keywords: Tissue engineering, regenerative medicine, immunity, immunosuppression, immunodeficient, infection
Graphical Abstract
1.0 Introduction and Motivation
The immune system plays a key role in both resisting infection as well as wound healing and tissue regeneration. Inevitably, nearly all drug delivery vehicles and scaffolds for tissue engineering purposes will elicit a response from the host immune system in vivo. While this response may range from a general foreign body reaction to an antigen-specific cell activation, the interactions between immune system and biomaterials can significantly alter the success of any implanted therapeutic device. Activation of specific immune components can either favorably or adversely affect tissue regeneration [1]. In addition to interacting with the host immune system, drug delivery vehicles and other foreign materials used in tissue engineering may also serve as a nidus for infection even in an otherwise healthy host [2–4]. These considerations (immune response and susceptibility to infection) become even more complicated in immunodeficient populations.
In many cases, the populations that will most benefit from drug delivery and tissue engineering-based strategies for wound healing have immunodeficiencies. For example, diabetic foot ulcers are estimated to represent an annual cost of $9.1–13.2 billion dollars in the USA [5] and have long been a target for regenerative medicine strategies [6]. However, diabetic-related immunodeficiencies such as decreased macrophage activation, inhibited vascularization and ability for immune cells to access the ulcer, and other complex host factors represent long-standing challenges in diabetic wound healing [7, 8]. Similar immunodeficiency-related challenges are present in strategies to regenerate tissues in populations with large wounds resulting from malignancy and tumor resection, trauma and/or burns, infection, and other clinical scenarios due to either 1) local immunosuppression from disruption of local vascularization or 2) systemic immunosuppression from disease-related factors [9–12]. Furthermore, tissues and organs generated in ex vivo bioreactor systems from allogeneic cell sources will require chemically-induced immunosuppression after implantation to prevent rejection by their host, although it is noted that allogeneic mesenchymal stem cells may have immunomodulatory abilities that protect against adverse immune responses [13]. Finally, due to advances in both disease-specific treatment and supportive care, immunocompromised populations such as organ transplant recipients and patients with human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS) have increased life expectancy and will benefit from wound repair strategies for similar reasons as the immunocompetent population. Ultimately, it is important to understand the role of the immune system in tissue regeneration due to the number of clinical scenarios in which patients with tissue deficiencies will also have co-morbid immune deficiencies.
Given the role of the immune system in wound healing and the specific challenges associated in tissue regeneration in the immunocompromised host, this area is of active interest to researchers working in the fields of drug delivery and regenerative medicine. In this work, we will discuss 1) the role of various components of the immune system in wound healing; 2) the effect of immunodeficiency in areas relevant to drug delivery/regenerative medicine; and 3) current advancements in harnessing drug delivery strategies to improve outcomes in immunocompromised populations.
2.0 Immune Cells and Wound Healing
There are a variety of important mediators of wound healing and inflammation within the innate and adaptive arms of the immune system. The innate immune system is a nonspecific defense response and includes neutrophils, macrophages, dendritic cells, complement proteins, and natural killer cells. The adaptive immune system is an antigen-specific response and includes B and T lymphocytes. Classically the immune system is studied for its role in protection against infection, involvement in inflammatory and autoimmune disease, and regulatory effects inhibiting the development of cancer. However, the cellular components of the immune system are also instrumental in wound healing. Following tissue damage, the immune system elicits a coordinated and organized response. Wound healing can be divided into four phases: hemostasis (clotting of blood); inflammation (sterilization and debridement); proliferation (expansion of host cells); and tissue remodeling [14]. These phases roughly correlate with the timeline of immune cell entry to the wound bed. Following initial injury and hemostasis, neutrophils enter a wound site after the first 24 hours, followed by macrophages in 48–96 hours, and lymphocytes after 120 hours [1]. Each cell type has a specific role in the promotion of wound healing (Figure 1).
Figure 1.
Summary of Roles of Immune Cells in Wound Healing
2.1 Neutrophils
Produced in the bone marrow as progenitors of myeloid lineage, neutrophils are one of the initial components of the immune system to reach a wound. In a non-specific fashion, neutrophils eliminate invading pathogens, secrete reactive oxygen species, and clear cellular debris from the wound bed. While these processes are necessary to initiate healing, an exuberant neutrophilic inflammatory response can destroy healthy tissue and impede regeneration [15, 16]. In fact, in certain models, it has been shown that depletion of neutrophils in sterile wounds results in more rapid healing, such as a murine model in which an 85% reduction of neutrophil concentration resulted in significantly greater cutaneous wound re-epithelialization [17, 18]. It should be noted that neutrophil concentration alone may be insufficient to predict effect on healing response. Host conditions such as diabetes can prime neutrophils to become more inflammatory and toxic to wound healing; for example, at baseline, 0.2% of neutrophils in normal hosts produce extracellular traps (cytotoxic proteins) while 2% produce extracellular traps in diabetic hosts [15]. Stimulation by inflammatory cytokines further amplified the difference in expression of cytotoxic secretion by neutrophils in diabetic compare to immunocompetent hosts. Overall, neutrophils are not strictly necessary for wound healing and may even be detrimental depending on recruited concentration and phenotype due to excessive inflammatory response. Despite the possible detriment of neutrophils, it should be noted that most non-surgical wounds are highly susceptible to infection (either from the environment or from the host’s microbiome) and infection is highly detrimental to tissue regeneration [19]. From an evolutionary standpoint, the protective role of neutrophils must outweigh their detrimental effect on healing.
Nevertheless, the presence of neutrophils also activates specific components of the wound healing responses. For example, neutrophil attachment to local endothelial and epithelial populations results in upregulation of Akt and β-catenin signaling [20] and potent modulators [21]. Knockout of C-X-C Chemokine Receptor Type 2 (CXCR2), a mediator of neutrophil recruitment to the wound bed, inhibits neutrophil entry and resulted in significantly delayed wound healing in a murine model [22]. Disease processes, such as old age and diabetes, in which neutrophil phagocytic ability is compromised has also been shown to delay wound healing [23]. Ultimately, while not necessary for tissue regeneration, neutrophils are important in the sterilization and debridement of the wound and may play a role in priming sites for healing. Recent work has highlighted the complexity of the neutrophil response in reaction to biomaterials and suggested neutrophils play a more nuanced part in host/material interactions [24].
2.2 Monocytes and Macrophages
Monocytes are progenitors of the myeloid lineage produced in the bone marrow and part of the innate immune system and differentiate into macrophages or dendritic cells based on environmental cues. Monocytes will circulate within blood vessels, migrate to a wound site, and then differentiate into specific macrophage populations [25]. The macrophage/monocyte axis is arguably the most important effector immune response following tissue injury in order to elicit and promote wound healing. Both monocytes and macrophages play major roles as major producers of cytokines and growth factors during the inflammatory and early proliferative phases of wound healing [26]. These secreted signaling molecules include factors important for angiogenesis and matrix deposition such as vascular endothelial growth factor (VEGF), insulin-like growth factor-1, transforming growth factor-β, and interleukin-10 [26–28]. In addition, macrophages prevent overgrowth of tissues once repair is complete [29]. However, not all macrophages are the same; macrophage polarization refers to the alteration of macrophage phenotype. M1 macrophages are proinflammatory in nature and phagocytose neutrophils and secrete inflammatory cytokines, whereas M2 macrophages are regarded as regenerative and secrete growth factors and migratory cytokines to promote healing [30]. Uncontrolled proinflammatory M1 polarization leads to chronic wounds rather than successful healing in both human patients and murine models of disease [31]. M1 macrophages inhibit wound healing through mechanisms such as direct tissue damage via secretion of radicals as well as induction of local fibroblasts to destroy rather than produce extracellular matrix [32]. M2 macrophages, on the other hand, play major regulatory roles in tissue remodeling and resolving inflammation [33]. M2 macrophages secrete paracrine factors that upregulate fibroblast proliferation and stimulate matrix production [32]. In addition, M2 macrophages also induce angiogenesis through secretion of growth factors [34]. Macrophages are plastic in nature and polarization has been demonstrated to be reversible under different in vitro and in vivo conditions [35]. The different factors that drive macrophage polarization during wound healing under different host conditions is an active and complex area of current research.
Unlike neutrophils, macrophages are necessary for tissue healing. Macrophage-specific β-catenin was shown to be necessary to complete wound healing in a murine knockout model [36]. By creating murine knockout models, or animal models where certain monocyte/macrophage-related pathways are selectively inhibited or removed, the importance of these cell lines and their products to wound healing has been demonstrated (Table 1). Specific ablation of macrophages, while leaving monocyte and neutrophil populations intact, also results in impaired wound healing [37].
Table 1.
Examples of Murine Macrophage-Related Knockout Models With Impaired Wound Healing
Factor | Type | Produced by | Effect | Reference |
---|---|---|---|---|
IL-6 | Cytokine | Macrophages | Neutrophil adhesion | Gallucci et al. [40] |
GM-CSF | Cytokine | Damaged keratinocytes | Neutrophil and macrophage recruitment | Fang et al. [41] |
MCP-1 | Cytokine | Macrophages | Macrophage phenotype | Low et al. [42] |
CX3CR1 | Receptor | Macrophages | Macrophage recruitment/phenotype | Ishida et al. [43] |
CXCR2 | Receptor | Damaged keratinocytes and endothelial cells | Neutrophil and monocyte recruitment | Devalaraja et al. [22] |
MyD88 | Cytoplasmic adapter | Macrophages | Macrophage phenotype | Macedo et al. [44] |
IL-6 = interleukin-6; GM-CSF = granulocyte-macrophage colony-stimulating factor; MCP-1 = monocyte chemotactic protein-1; CX3CR1 = C-X3-C Motif Chemokine Receptor 1; CXCR2 = C-X-C Motif Chemokine Receptor Type 2; MyD88 = myeloid differentiation primary response gene 88
However, in an unfavorable environment, monocytes and macrophages may be unable to mount a sufficient regenerative response. For example, even extremely low levels of bacterial endotoxin (5 ng/kg body weight injected once every three days for ten days) inhibit the wound healing response of monocytes in a murine cutaneous defect [38]. As arguably the most important immune cell type in tissue repair, there is great interest in developing drug delivery techniques to create favorable environments and locally recruit specific macrophage populations to wound sites to enhance tissue healing in the immunoregenerative biomaterial strategy [39]. Delivery of specific cytokines or use of specific biomaterials can preferentially promote M2 over M1 macrophage phenotype and stimulate wound healing.
2.3 Lymphocytes
Including T and B lymphocytes (adaptive immune cells) and natural killer (NK) cells (innate immune cells), the lymphocytes also have important roles in response to injury. Regulatory T lymphocytes are the predominant lymphocyte present in skin and become activated upon injury [45]. Ablation of regulatory T lymphocytes results in delayed wound healing due to inefficient transition from inflammatory to proliferative phases, suggesting lymphocytes are important modulators of the inflammatory reaction to tissue injury [46]. In addition, T lymphocytes specifically are important regulators of angiogenesis and collagen deposition. Through activation of C-X-C Motif Chemokine Receptor 3 by local secretion of C-X-C Motif Chemokine Ligands 9, 10, and 11 at the wound site, T lymphocytes are involved in the maturation of the angiogenic response and “prune” immature vessels [47]. When challenged with specific antigens, T lymphocytes secrete factors which induce dermal fibroblasts to proliferate and produce collagen [48]. The roles of NK cells and B lymphocytes in wound healing are less clear; both cell types are involved in mitigation of infectious processes.
However, as is the case with neutrophils, it does not appear that lymphocytes are necessary for successful wound healing. In murine models of CD4 deficiency and CD8 deficiency, neutrophil recruitment was altered and cytokine expression was changed but ultimately wound healing was not impaired [49].
2.4 Immune Cells and Angiogenesis
Angiogenesis is key to the regeneration of tissues. As discussed above, immune cells play various roles in initiating and regulating the angiogenic response. Hypoxia, or lack of oxygen within a tissue, recruits immune cells to a damaged site [50]. In addition to sterilizing the wound bed, macrophages drive the initial sprouting of local endothelial cells [51]. As the wound evolves and angiogenesis is initiated, there is a large amount of molecular cross-talk between vascular endothelium and the immune system. For example, Wnt signaling produced by the endothelium is recognized by Frizzled receptors on T lymphocytes, neutrophils, and macrophages and promotes upregulation of matrix metalloproteases from these immune cells [50], allowing for immune cells to more quickly extravagate to the wound site as well as begin the remodeling process by degrading damaged tissue. As the healing response evolves, T cells are actively engaged to reduce/redirect immature and superfluous vessels as the tissue matures [47]. In wound beds where immune cells exhibit pro-inflammatory phenotypes (such as inflammatory neutrophils and M1 polarized macrophages), angiogenesis has been shown to be inhibited [52]. The interplay between immunity and angiogenesis is an important factor in the wound healing response.
3.0 Diseases of Immunodeficiency and Wound Healing
Not all immunodeficient states are created equal. Even subtle differences in immune populations can drastically alter wound healing. For example, in a study of tissue recovery after limb ischemia, two different strains of mice were studied (inbred Balb/C nude mice versus outbred athymic nude mice) [53]. Despite the same genetic mutation and resulting immune deficit (absent thymus from an autosomal recessive mutation in Foxn1nu), Balb/C mice had significantly impaired recovery compared to outbred mice (50±10% to 75±20%). The authors theorized that the outbred mice had greater monocyte activation (possibly from greater genetic heterogeneity), resulting in increased angiogenesis. Genetic background has been shown to significantly affect angiogenesis associated with wound healing among different mouse strains [54]. Therefore, understanding immunodeficiency and its impact on wound healing in the greater context of specific disease states is critical when designing strategies to augment regeneration in immunocompromised hosts.
In addition to complex host disease states, local tissue damage (such as from trauma or combat-related wounds) produces states of local immunosuppression due to compromised vasculature and necrotic tissue. These wound beds are particularly at risk for infection and penetration of therapeutics such as antibiotics is limited [55]. In composite wounds, where multiple tissue types are damaged (i.e. skin, fascia, muscle, nerve, and bone), healing becomes increasingly challenging and the role of the immune system in the repair of these complex wound beds is an area that warrants further research.
In this review, diabetes mellitus, HIV/AIDS, and malignancy will be discussed as examples of immunodeficient states with altered wound healing. However, many other host conditions are associated with immunodeficiency and pose additional specific challenges for regeneration, such as organ transplant recipients on immunosuppressants and patients with inborn genetic immunodeficiencies [56, 57].
3.1 Diabetes Mellitus
In chronic wounds associated with diabetes mellitus, the inflammatory phase is upregulated without progression to the proliferative phase [26]. Recruited neutrophils and macrophages (primarily M1 polarization) secrete more proinflammatory factors and cause additional damage to host tissues [15, 58]. While most likely a multi-factorial issue, diabetes results in the downregulation of Cysteine-rich protein 61 (CCN1), causing decreased neutrophil efferocytosis (autophagy) and decreased M2 polarization of macrophages; delivery of CCN1 can restore normal wound healing in diabetic mice [59].
In addition to altering the phenotype of immune cells and promoting inflammation, diabetes mellitus is a chronic disease which also results in significant microvascular disease [60]. With decreased blood supply, there is limited access by peripheral immune cells as well as circulating stem cells that can stimulate a healing response. In addition to wound access, diabetes affects stem cells at their source; recent data have shown that diabetes induces epigenetic changes in bone marrow progenitor cells that is then passed down to wound macrophages and drives greater inflammatory interleukin-12 production, resulting in decreased wound healing [61]. Relevant to the field of tissue engineering, it has also been found in a murine model that autologous adipocyte-derived stem cells from diabetic hosts have decreased inherent wound healing capacity compared to allogeneic isolates from non-diabetic animals [62]. In addition to altering immune cell phenotype, inhibiting immune system access to the wound bed, and negatively impacting the signaling between immune cells and stem cells, diabetes also mitigates healing due to the susceptibility of wound to infection. Given the changes in immune phenotype and necrotic tissue from vascular disruption, diabetic wounds are often chronically infected which results in further tissue damage [63]. For these reasons, 1) restoring normal immune phenotype, 2) providing access of immune cells to the wound site through vascularization, 3) artificially delivering growth factors and cytokines normally produced by healthy macrophages and other local tissues to the wound, and 4) releasing antibiotics locally to treat and prevent infection are strategies being pursued within drug delivery for the treatment of diabetic wounds.
3.2 HIV/AIDS
As of 2013, there were estimated to be 1.7–2.1 million new cases of HIV globally per year [64]. With advances in highly active antiretroviral therapy (HAART), patients with HIV/AIDS now have extended life expectancy, suffer from similar medical issues as the general population, and may also benefit from drug delivery/tissue engineering strategies. HIV is a disease in which CD4 cells specifically are targeted and destroyed. As noted previously, T lymphocytes may augment wound healing (such as through enhancement of angiogenesis) but are not necessary. There is controversy regarding the effects of HIV on wound healing. In clinical studies, it does not appear HIV positivity affects healing time after male circumcision, in the healing of tibial fracture, or in the osteointegration of dental implants [65–68]. However, in all of these cases, HIV may increase the likelihood of infection and subsequent inhibition of healing compared to the general population. It is possible that current HAART therapy, the negative effects of HIV infection on wound healing may be masked [67]. In older clinical work published before the widespread use of HAART, it was found that patients with HIV had significantly worsened wound healing after anorectal surgery (attributed to loss of inflammation-modulating CD4 cells) [69].
Therefore, in designing drug delivery strategies to aid in wound healing in populations with HIV/AIDS, it is important to consider the patient’s current CD4 levels. Pre-treatment with HAART and adequate antibiotic prophylaxis may be critical in the success of regenerative medicine therapies in the setting of HIV/AIDS.
3.3 Malignancy
Patients suffering from malignancy often have complex tissue defects from either local invasion and destruction by the tumor or due to resection in order to surgically remove the tumor. Leveraging tissue engineering and drug delivery to heal wounds in cancer patients has long been a goal of regenerative medicine [70, 71]. However, cancer and its treatment (chemotherapy and radiation therapy) can be inhibitory to wound healing. For example, cutaneous wounds in a rat model of hepatoma had decreased mechanical integrity compared to healthy animals after being given time to heal, suggesting impaired tissue remodeling [72]. The degree of decreased mechanical integrity correlated with the volume of tumor burden, despite the tumor being ectopic to the injury site. In addition to malnutrition, another possible mechanism contributing to inhibition of wound healing in the setting of malignancy is disruption of the bone marrow. Given that myeloid-derived cells are produced in bone marrow, in addition to other stem cell populations that may be relevant in wound healing [73], tumors that metastasize to the marrow or otherwise exhaust the bone marrow’s ability to replenish itself may affect wound healing.
Likewise, the treatments for cancer may be toxic to regenerative processes. Common chemotherapeutics that affect rapidly dividing cells, such as doxorubicin, have been shown to inhibit wound healing in cancer patients [72]. However, even as cancer treatments become molecule-specific, there is still a possibility for a profound effect on wound healing depending on the specific mechanism in question. There are many shared genes that are upregulated in both malignancy and wound healing that may be attractive targets for anti-cancer strategies [74]. For example, the chemotherapeutic bevacuzimab (anti-VEGF monoclonal antibody) increased post-surgical wound healing complication rates in patients with colorectal cancer from 3 to 13% (not statistically significant) [75], as well as has resulted in tissue fistulas and necrosis in the treatment of head and neck cancer [76]. While an attractive target against tumor growth, VEGF is a critical angiogenic growth factor secreted by macrophages and other cells during tissue remodeling. Radiation therapy also locally disrupts wound healing. Through mechanisms that are not entirely understood, radiation causes overexpression of proinflammatory and matrix-producing cytokines by immune cells, resulting in uncontrolled fibrosis [77]. There is also downregulation of matrix metalloproteinases, molecules which help remodel the extracellular matrix. Given the extensive local damage to keratinocytes and fibroblasts, drug delivery strategies may require recruitment of other cell populations to regenerate healthy tissue at the site of radiation. Lastly, check point inhibitors are a new class of therapeutics for cancer treatment. They have been known to induce autoimmune adverse reactions and affect the immune response; however, their effect on wound healing is thus far unexplored and will need to be assessed as clinical trials continue [78]. The effect of autoimmune disease and therapies to treat autoimmune disease draw similar parallels regarding wound healing. Autoimmune diseases, such as systemic lupus erythematous, rheumatic arthritis, and inflammatory bowel disease result in dysregulation of the immune response which can alter wound healing and treatments for autoimmune disease result in significant immunosuppression that has been demonstrated to negatively affect wound healing [79–81].
4.0 Drug Delivery Strategies in Immunocompromised Hosts
In immunodeficient hosts, drug delivery strategies can be utilized to recruit immune cells, alter cell phenotype, or replace certain functions of deficient immune cells in order to augment wound healing (Fig. 2). In a broad sense, there are few studies designed to investigate the role of the immune system in drug delivery. In one murine experiment, nanoparticle-mediated gene delivery was studied in immunocompetent versus athymic mice. While initial delivery was decreased in immunodeficient mice, the two groups were similar after 24 hours, suggesting that in vivo drug delivery kinetics may be unchanged in some immunocompromised models [82]. In diseases of immunodeficiency with significant vascular compromise, such as diabetes, one may expect that drug delivery strategies relying on systemic circulation may be inhibited; therefore, delivery systems featuring local release (such as those included as part of implanted tissue scaffolds) may be of greater benefit in those populations. In general, strategies can be divided as recruitment-based, phenotype-based, or replacement-based.
Figure 2.
Drug delivery and tissue engineering strategies can promote wound healing by recruitment, phenotype modification, or replacement of immune populations.
4.1 Recruitment of Immune Populations
In non-healing wounds in certain immunodeficient populations, cell types such as macrophages and neutrophils are low in number and/or are not being recruited in great enough quantity to drive the wound healing response. Therefore, there have been efforts in leveraging biomaterials to locally deliver chemokines to 1) boost the proliferation of existing immune cells and 2) recruit immune cells locally to the wound site. As described in Figure 1, the wound healing response is divided into phases featuring neutrophil, macrophage, and lymphocyte activity. As a neutrophil/macrophage chemoattractant, the local application of GM-CSF directly to wounds has been shown to promote healing in clinical studies involving chronic non-healing wounds in patients with immunodeficiencies such as diabetes, vascular compromise, and cancer [83]. However, the in vivo half-life of GM-CSF is only approximately 1.5–7 hours [84, 85]. Neutrophils and macrophages are heavily involved in the healing response for up to 120 hours (Figure 1). Therefore, there have been efforts for both wound healing as well as augmentation of vaccine efficacy to harness biomaterials as local delivery agents for GM-CSF. Poly(lactide-co-glycolide)(PLGA)/poly(lactide)-based microparticles, for example, were demonstrated to be capable of releasing biologically-active GM-CSF in a murine model at concentrations > 10 ng/mL for at least nine days and these microparticles resulted in local recruitment of neutrophils and macrophages at the site of injection [84]. Chitosan is another biomaterial that has been shown to sustain delivery of GM-CSF within murine models [86]; in addition to its ability to enhance GM-CSF local delivery, chitosan has the added benefit of inherent antimicrobial properties [87, 88]. Further work has been done to create chitosan-based microparticles for further control over delivery kinetics and deliver GM-CSF plasmid DNA [89]. Other explored vehicles include liposomes [90], adenovirus-mediated gene transfer [91], and poly(γ-glutamic acid)-based hydrogels [92].
Biomaterial-based local delivery of GM-CSF is most likely the closest technology to full clinical translation and regulatory approval given that GM-CSF has already been approved by the United States Food and Drug Administration for systemic use following induction of chemotherapy to shorten time for neutrophil recovery (among other systemic use approvals). However, other experimental agents that attract immune populations to wounds are also being explored. These newer molecules are of interest as they may not only recruit macrophages to wounds but may selectively recruit M2 (pro-regenerative) rather than M1 (pro-inflammatory) macrophages. For example, the controlled local delivery of a macrophage-recruiting chemokine (SEW2871, a sphingosine-1-phosphate agonist) within a gelatin hydrogel selectively recruited M2 macrophages and resulted in increased wound healing in a murine model [93]. The same molecule was also delivered using lactic acid-grafted gelatin micelles within gelatin hydrogels and resulted in greater macrophage presence and improved healing in critically-sized rat ulnar defect [94]. Local delivery of another sphingosine-1-phosphate agonist, FTY720, from a nanofiber scaffold composed of poly(lactide-co-glycolide) and poly(ε-caprolactone) promoted increased recruitment of M2 macrophages, vascularization, and wound healing in a murine mandibular defect model [95]. Temporally, if normal neutrophil and/or macrophage activity can be restored to the wound in an immunodeficient host, these cells may be able to initiate angiogenesis [34], tissue remodeling [33], and extracellular deposition [32] and overcome the inhibition of these activities normally seen in immunodeficient hosts.
Of note, wound healing in immunocompromised patients may respond to differently recruitment factor strategies compared to their immunocompetent hosts. For example, IL-6 is a pleotropic macrophage- and keratinocyte-secreted cytokine that aids in immune response such as neutrophil adhesion and has been shown to be important in the wound healing response in mouse models [40]. In steroid-induced immunodeficient disease models as well as IL-6 knockout models, injection of IL-6 (or IL-6 plasmid) promoted increased wound healing [40, 96]. However, these injections delayed wound healing in normal immunocompetent mice [96]. As IL-6 is present in wounds already in healthy animals, the authors of the study speculated that the additional delivered IL-6 may have caused upregulated matrix metalloproteinases that interfered with collagen deposition. These findings highlight the importance of considering host immune status when developing strategies to promote wound healing; the same delivery factors that stimulate regeneration in one host may not be appropriate for another.
4.2. Phenotype Modification
While biomaterials may be conducive as tissue scaffolds to aid in the regenerative response, they also function as foreign bodies that can elicit a pro-inflammatory phenotype from migrating immune cells. In addition, certain immunodeficient host states such as diabetes and malignancy can further predispose recruited macrophages to M1 rather than M2 polarization and impede the wound healing response [33]. As M2 macrophages are critical in the secretion of cytokines for angiogenesis and regulation of inflammation, there has been a great effort in understanding factors influencing macrophage polarization and developing drug delivery systems and biomaterials-based approaches to promote M2 macrophage recruitment.
Interleukin-4 (IL-4) is one such cytokine that is capable of driving macrophages to an M2 phenotype even in the presence of biomaterials that normally elicit M1 polarization, such as poly(methylmethacrylate) [97]. When injected directly into an implant-associated calvarial model of inflammation, IL-4 resulted in decreased inflammation and osteolysis [98]. For controlled delivery, investigators have studied the use of silk films to as biomaterials to deliver IL-4 locally and promote M2 polarization [99]. In addition to spatially-controlled IL-4 delivery, temporally-controlled IL-4 delivery with respect to macrophage polarization may be important in wound healing. There is some evidence that angiogenesis may be best stimulated by an early M1 response followed by an M2 response. Therefore, there have been efforts to develop scaffold delivery systems with early release of M1-inducing factors (such as interferon gamma) followed by sustained release of M2-inducing factors (such as IL-4). This was achieved by adsorbing interferon gamma onto a decellularized bone-based scaffold for rapid release while simultaneously binding IL-4 via biotin and streptavidin for delayed release [100]. Ultimately, in vivo results in a murine subcutaneous implant model did not show differences in M1/M2 polarization; the authors suggested that overlap in the release of both factors may have interfered with affecting macrophage phenotype.
In addition to temporal control, combination approaches of recruitment factors and polarization factors have been explored. For example, MCP-1 and IL-4 were delivered from a multidomain peptide hydrogel scaffold in mouse model to locally recruit macrophages (via MCP-1) and then drive the M2 response (via IL-4) [101]. As a better understanding of the cytokine environment in wound healing is understood, more complex approaches such as these can be designed and implemented. In addition to IL-4, other soluble factors are also being explored for polarization control. Local injections of lipoxin A4 and resolvin D1 caused increased M2 and reduced M1 macrophages within a chitosan scaffold in a mouse model, for example [102]. In contrast to delivering specific cytokines, biomaterials can also be leveraged to capture or scavenge inflammatory cytokines that may be locally detrimental to healing in immunocompromised populations. In a murine diabetic wound model, a glycosaminoglycan-based hydrogel was capable of scavenging inflammatory cytokines and improving wound healing [103].
However, drug delivery is not the only way biomaterials can be leveraged to influence macrophage polarization. The material properties of scaffolds themselves can be designed to drive macrophage phenotype. Pore size [104], fiber diameter [105], surface microstructures [106], and mechanical loading [107] are all physical cues whose modulation has demonstrated to influence polarization. While the focus of tissue engineering has classically been on modulating material properties to optimize migration/differentiation of native cells of the desired tissue type (ie create materials conducive to osteoblasts for bone tissue engineering), maximizing macrophage polarization by scaffold design may be equally important to elicit a regenerative environment within an implant. This may be even more critical in the immunocompromised host, where immune cells that would typically aid in the healing response are further depleted or inhibited.
4.3 Replacement of Immune Cell Functions
Lastly, many efforts within drug delivery and tissue engineering to augment healing in immunocompromised hosts involve replacing the factors produced in wound healing under normal conditions. Replacement of growth factors that are normally secreted or induced by macrophages in large wounds has been shown to augment healing with local application in clinical studies [108, 109]. For example, initiation and regulation of angiogenesis is a major role of the immune system, a process inhibited in diabetic hosts [27, 47, 110]. However, local delivery of angiogenic growth factors such as basic fibroblast growth factor and VEGF have been shown to enhance wound repair in diabetic animal models, as demonstrated using delivery systems such as chitosan scaffolds [111], fibrin scaffolds (with or without PLGA microparticles) [112], and poly(ethylene glycol)/poly(lactide) electrospun fibers [113]. As macrophages are responsible for inducing endothelial sprouting through growth factor secretion, replacing these growth factors early in the wound healing process may be critical in preventing chronic wound development [32]. In addition to angiogenesis, immune cells also produce growth factors regulating tissue deposition and recruitment of matrix-producing cells, such as epidermal growth factor [114]. In large burn wounds in humans and animal models, considered immunodeficient wound environment [115], local delivery of epidermal growth factor has been shown to enhance wound healing [108, 116]. Drug delivery vehicles such as chitosan gels [117, 118], liposomes within a chitosan matrix [119], fibrin scaffolds with xenogeneic human keratinocytes [120], and hyaluronan [121] have all shown efficacy in improving regeneration in animal models of burn wounds. Other growth factors associated with macrophages have shown similar efficacy in the treatment of large wounds; replacing/augmenting macrophage secretion with local delivery-based strategies have shown benefit using keratinocyte growth factor, platelet derived growth factor, transforming growth factor-beta, and other cytokines in immunocompetent and immunocompromised animal models [122–125].
However, lack of appropriate growth factor secretion is not the only challenged presented wounds in immunodeficient populations. These sites are also inherent to infection which further creates tissue damage and inhibits the healing response. Sterilization of the wound bed is in fact part of the first phase of the wound healing process (Figure 1). To replace the infection-resistant function of the immune system, local delivery of antibiotics to treat or prevent infection has been a longstanding goal within the field with some success in translation of biomaterials-based products to market [126, 127]. Polymer-based delivery of antibiotics as well as the synthesis of inherently antimicrobial polymers and antifouling materials are areas that have been reviewed recently [87, 128, 129]. Emulsions and oils are other vehicles being explored that have inherent antimicrobial activity [130, 131]. In addition to delivering antibiotics via polymer-based delivery vehicles, a recent innovative approach involved loading neutrophil-like cells themselves with antifungal agents ex vivo, injection of the cells into an immunocompromised murine model featuring infection, and leveraging the cells as delivery vehicles [132]. In strategies featuring local antibiotic delivery, some caution should be exercised; there is evidence that certain antibiotics have pleotropic effects, including inhibition of neutrophil-generated matrix metalloproteinases necessary for tissue remodeling, that can inhibit the regenerative response [133, 134] although this magnitude of this effect is still under study [135]. Another approach to replace the antimicrobial activity of the immune system in deficient patients has been delivery of antibodies or plasmids for the expression of antibodies to artificially replace B-lymphocyte immunity. For example, local application of polyclonal immunoglobulin to infected burn wounds in a mouse model significantly increased wound healing [136]. If a specific pathogen is recognized as a threat to wound recovery in a population, there are also strategies to deliver monoclonal antibodies. For example, local delivery of an adeno-associated virus vector was used to deliver anti-influenza antibody and prevented immunocompromised mice from dying of influenza [137]. A similar approach to deliver monoclonal antibodies was shown to prevent respiratory syncytial virus infection [138]. By replacing artificially replacing the immune system’s role in promoting angiogenesis, secreting regenerative growth factors, and preventing infection of the wound bed, drug delivery approaches can facilitate healing in the immunocompromised host.
4.4 Considerations for the Immunocompromised Host
As discussed in this work, patient populations featuring immunodeficiencies present unique challenges when designing regenerative medicine strategies. Specific factors relating to the host and drug delivery vehicle that must be taken under consideration are summarized in Table 2.
Table 2.
Examples of Considerations When Designing Drug Delivery Strategies for Immunocompromised Hosts
Host Factors | Drug Delivery Factors | ||
---|---|---|---|
Factor | Examples | Factor | Examples |
Specific immunodeficiency | -Deficiencies in neutrophils, macrophages, B cells, or T cells -Complex pleotropic host disease states (diabetes, systemic lupus erythematosus, etc.) |
Purpose/goal of delivered therapeutic | -Recruitment of immune cells -Stimulation of specific immune phenotype -Replacement of immune function (antibiotic, antibody angiogenic factor, etc.) |
Evolution of host immunodeficiency | -Acute immune insult followed by gradual resolution (burn, trauma, etc.) -Chronic immunodeficiency (diabetes, HIV/AIDs, etc.) -Waxing/waning immunodeficiency (need for intermittent chemotherapy, autoimmune disease, etc.) |
Spatial delivery requirements | -Systemic versus local delivery -Concentration gradient -Release from the center of the wound to the periphery versus from the periphery to the center |
Angiogenic potential | -Vascularization of the wound site -Ability of the host to initiate angiogenesis |
Temporal delivery requirements | -Duration of therapy -Release kinetics (Continuous release, burst release, pulse release, etc.) |
Presence of pathogens | -“Clean” versus “dirty” wound (for example, tissue loss from resection of a tumor versus tissue loss from a motor vehicle accident) -Proximity to microbiome (skin, oral cavity, gastrointestinal system, genitourinary system, etc.) |
Requirement for tissue scaffold and scaffold considerations | -Large versus small defects -Type of scaffold (synthetic polymer, naturally-derived material, composite, etc.) -Scaffold delivery (injection, self-assembly, surgical insertion, etc.) |
Other co-morbidities | -Nutritional status -Genetic conditions -Socioeconomic and psychologic burdens of disease, access to healthcare |
Immune response to vehicle/scaffold | -Impact on macrophage polarization -Fibrous capsule formation versus tissue incorporation -Response to and clearance of degradation products |
Regarding the host, the specific immunodeficiency is critical. For example, as diabetics have deficiencies in converting macrophages from M1 to M2, strategies for the release of factors such as IL-4 may be beneficial in treating a chronic ulcer. On the other hand, in a patient with immunodeficiencies due to chemotherapy induction, the challenge to wound healing is not necessarily macrophage polarization but bone marrow suppression and lack of numbers of neutrophils and macrophages. In this example patient, delivery strategies to extend the half-life of GM-CSF as well as to provide local antibiotic wound coverage may be desirable. Another key host factor is the vascularization to the wound site and the host’s angiogenic potential. Diabetes, burns, implantation of foreign bodies, and other immunodeficiencies can compromise wound vascularization and deficiencies in macrophages and lymphocytes can inhibit angiogenic potential. In addition, the wounds of immunocompromised patients are at increased risk for fungal infections, which can further inhibit angiogenesis [139, 140]. In addition to inhibiting angiogenesis, pathogens can further disrupt healing by directly damaging surrounding tissue and increasing inflammation within the wound bed. Immunocompromised patients are highly susceptible to infection and often benefit from antibiotic prophylaxis; due to decreased angiogenic potential, these same patients may benefit from local antibiotic delivery strategies due to disruption of systemic vascularization to wound sites. Lastly, many patient populations with immunodeficiencies may suffer from other co-morbidities such as decreased renal or hepatic clearance that may affect drug delivery approaches [141, 142].
In the design of the drug delivery vehicle, there are specific factors that may require special attention in the immunocompromised host. As discussed earlier, as angiogenesis may be decreased due to macrophage and/or lymphocyte deficiencies, systemic approaches (intravenous injection, oral intake, etc) may not result in vehicle or therapeutic appropriately accessing the wound site. In addition, temporal factors such as the duration of release as well as the sequential release of different cytokines has been demonstrated to significantly affect the immune system’s ability to augment wound healing [100]. In addition, the need for a tissue scaffold in conjunction with a drug delivery vehicle should be evaluated. In some critically-sized tissue defects, a scaffold may be required for healing to proceed; in others, delivery of cytokines or growth factors may be sufficient. Finally, the immune response to the delivery device in question must also be considered. As common polymers leveraged in drug delivery have been shown to stimulate M1 polarization without further augmentation [97], these vehicles and/or scaffolds may require modification such as additional loading of M2-inducing cytokines or topographical changes that have been demonstrated to promote M2 polarization. In addition, the increased inflammatory conditions that may be present in the wound of immunodeficient patients may cause increased biodegradation rates of drug delivery vehicles fabricated from commonly used aliphatic polyesters [143, 144], requiring evaluation of release kinetics under different physiologic conditions. Lastly, in immunocompromised patients, wounds are particularly vulnerable to infections which further inhibits the healing response. A new and exciting area of research involves the microbiome (including bacteriome and mycobiome) and its effects on host inflammation and wound healing [145, 146]. Given that many wounds are adjacent to microbiome communities, such as the skin or oral mucosa, this field may hold relevance for treating wounds in immunocompromised populations. The development of drug delivery and tissue engineering-based approaches for the immunocompromised host requires special considerations to optimize success.
5.0 Conclusions
Given the importance of the immune system in initiating and regulating wound healing, patients with immunodeficiencies present unique challenges to the field of regenerative medicine. In this work, we have discussed the roles of specific cells within the immune system, highlighted changes in wound healing in several specific common immunodeficient states that may benefit from drug delivery approaches, and reviewed current strategies in the literature that may benefit immunocompromised populations.
While there has been significant progress in understanding the roles of different cells in the immune system in wound healing, the effect of different cytokines on the regenerative process, and how different immunodeficient states affect tissue repair, there remains much work to be done to understand how these challenges can be overcome using drug delivery strategies. The vast majority of in vivo studies are performed in immunocompetent animal models. As has been discussed in this work and demonstrated in the literature, even subtle immunodeficiencies can greatly affect healing. With advances in medicine permitting greater life expectancy in certain immunodeficient populations, such as those with HIV/AIDS, organ transplant recipients, survivors of malignancy, and diabetics, there is a great need to develop and validate strategies in animal models with similar co-morbidities. Biologists and immunologists have developed a plethora of animal models featuring specific immunodeficiencies through chemical or genetic manipulation; many of these models have been validated mechanistically and have well-understood deficiencies in specific immune cell lines. Collaborations between bioengineers, immunologists, materials scientists, veterinarian staff, and physicians may be fruitful in developing physiologically-relevant platforms to evaluate drug delivery strategies for common immunocompromised populations.
Fortunately, there has been a great deal of work done in the development of drug delivery vehicles for wound healing in the immunocompetent host. By understanding the unique challenges in their immunocompromised counterparts, these strategies can be more rapidly modified as required and validated in the appropriate models rather than starting from scratch. There is a real clinical need to enhance wound healing in patient populations with immunodeficiencies. The fields of drug delivery and tissue engineering stand to make a large impact in improving healthcare outcomes by developing strategies to improve wound healing in the immunocompromised host.
Acknowledgments
This work was supported by the Army, Navy, NIH, Air Force, VA and Health Affairs to support the AFIRM II effort, under Award No. W81XWH-14-2-0004. The U.S. Army Medical Research Acquisition Activity, 820 Chandler Street, Fort Detrick MD 21702-5014 is the awarding and administering acquisition office. In addition, further support has been provided by the John S. Dunn Foundation and the National Institutes of Health (R01 AR068073). A.M.T. would like to thank the Baylor College of Medicine Medical Scientist Training Program and the Barrow Scholars Program. DPK acknowledges the Texas 4000 endowment.
List of Abbreviations
- CCN1
cysteine-rich protein 61
- CX3CR1
C-X3-C motif chemokine receptor 1
- CXCR2
C-X-C motif chemokine receptor type 2
- GM-CSF
granulocyte-macrophage colony-stimulating factor
- HAART
highly active antiretroviral therapy
- HIV/AIDS
human immunodeficiency virus/acquired immunodeficiency syndrome
- IL-6
interleukin-6
- MCP-1
monocyte chemotactic protein-1
- MyD88
myeloid differentiation primary response gene 88
- NK
natural killer
- PLGA
poly(lactide-co-glycolide)
- VEGF
vascular endothelial growth factor
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Park JE, Barbul A. Understanding the role of immune regulation in wound healing. Am J Surg. 2004;187:11S–16S. doi: 10.1016/S0002-9610(03)00296-4. [DOI] [PubMed] [Google Scholar]
- 2.Busscher HJ, van der Mei HC, Subbiahdoss G, Jutte PC, van den Dungen JJ, Zaat SA, Schultz MJ, Grainger DW. Biomaterial-associated infection: locating the finish line in the race for the surface. Sci Transl Med. 2012;4:153rv110. doi: 10.1126/scitranslmed.3004528. [DOI] [PubMed] [Google Scholar]
- 3.Darouiche RO. Treatment of infections associated with surgical implants. N Engl J Med. 2004;350:1422–1429. doi: 10.1056/NEJMra035415. [DOI] [PubMed] [Google Scholar]
- 4.Shah SR, Tatara AM, D’Souza RN, Mikos AG, Kasper FK. Evolving strategies for preventing biofilm on implantable materials. Materials Today. 2013;16:177–182. [Google Scholar]
- 5.Rice JB, Desai U, Cummings AK, Birnbaum HG, Skornicki M, Parsons NB. Burden of diabetic foot ulcers for medicare and private insurers. Diabetes Care. 2014;37:651–658. doi: 10.2337/dc13-2176. [DOI] [PubMed] [Google Scholar]
- 6.MacNeil S. Biomaterials for tissue engineering of skin. Materials today. 2008;11:26–35. [Google Scholar]
- 7.Maruyama K, Asai J, Ii M, Thorne T, Losordo DW, D’Amore PA. Decreased macrophage number and activation lead to reduced lymphatic vessel formation and contribute to impaired diabetic wound healing. Am J Pathol. 2007;170:1178–1191. doi: 10.2353/ajpath.2007.060018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Galkowska H, Wojewodzka U, Olszewski WL. Low recruitment of immune cells with increased expression of endothelial adhesion molecules in margins of the chronic diabetic foot ulcers. Wound Repair Regen. 2005;13:248–254. doi: 10.1111/j.1067-1927.2005.130306.x. [DOI] [PubMed] [Google Scholar]
- 9.Serafini P, Borrello I, Bronte V. Myeloid suppressor cells in cancer: recruitment, phenotype, properties, and mechanisms of immune suppression. Semin Cancer Biol. 2006;16:53–65. doi: 10.1016/j.semcancer.2005.07.005. [DOI] [PubMed] [Google Scholar]
- 10.Faist E, Schinkel C, Zimmer S. Update on the mechanisms of immune suppression of injury and immune modulation. World J Surg. 1996;20:454–459. doi: 10.1007/s002689900071. [DOI] [PubMed] [Google Scholar]
- 11.Vanzant EL, Lopez CM, Ozrazgat-Baslanti T, Ungaro R, Davis R, Cuenca AG, Gentile LF, Nacionales DC, Cuenca AL, Bihorac A, Leeuwenburgh C, Lanz J, Baker HV, McKinley B, Moldawer LL, Moore FA, Efron PA. Persistent inflammation, immunosuppression, and catabolism syndrome after severe blunt trauma. J Trauma Acute Care Surg. 2014;76:21–29. doi: 10.1097/TA.0b013e3182ab1ab5. discussion 29–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Tatara AM, Shah SR, Livingston CE, Mikos AG. Infected animal models for tissue engineering. Methods. 2015;84:17–24. doi: 10.1016/j.ymeth.2015.03.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.De Miguel MP, Fuentes-Julian S, Blazquez-Martinez A, Pascual CY, Aller MA, Arias J, Arnalich-Montiel F. Immunosuppressive properties of mesenchymal stem cells: advances and applications. Curr Mol Med. 2012;12:574–591. doi: 10.2174/156652412800619950. [DOI] [PubMed] [Google Scholar]
- 14.Guo S, Dipietro LA. Factors affecting wound healing. J Dent Res. 2010;89:219–229. doi: 10.1177/0022034509359125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wong SL, Demers M, Martinod K, Gallant M, Wang Y, Goldfine AB, Kahn CR, Wagner DD. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat Med. 2015;21:815–819. doi: 10.1038/nm.3887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Mountziaris PM, Spicer PP, Kasper FK, Mikos AG. Harnessing and modulating inflammation in strategies for bone regeneration. Tissue Eng Part B Rev. 2011;17:393–402. doi: 10.1089/ten.teb.2011.0182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Dovi JV, He LK, DiPietro LA. Accelerated wound closure in neutrophil-depleted mice. J Leukoc Biol. 2003;73:448–455. doi: 10.1189/jlb.0802406. [DOI] [PubMed] [Google Scholar]
- 18.Martin P, Leibovich SJ. Inflammatory cells during wound repair: the good, the bad and the ugly. Trends Cell Biol. 2005;15:599–607. doi: 10.1016/j.tcb.2005.09.002. [DOI] [PubMed] [Google Scholar]
- 19.Percival SL. Wounds and Infection. Biofilms in Infection Prevention and Control: A Healthcare Handbook. 2014:127. [Google Scholar]
- 20.Sumagin R, Brazil JC, Nava P, Nishio H, Alam A, Luissint AC, Weber DA, Neish AS, Nusrat A, Parkos CA. Neutrophil interactions with epithelial-expressed ICAM-1 enhances intestinal mucosal wound healing. Mucosal Immunol. 2016;9:1151–1162. doi: 10.1038/mi.2015.135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Cheon SS, Wei Q, Gurung A, Youn A, Bright T, Poon R, Whetstone H, Guha A, Alman BA. Beta-catenin regulates wound size and mediates the effect of TGF-beta in cutaneous healing. FASEB J. 2006;20:692–701. doi: 10.1096/fj.05-4759com. [DOI] [PubMed] [Google Scholar]
- 22.Devalaraja RM, Nanney LB, Du J, Qian Q, Yu Y, Devalaraja MN, Richmond A. Delayed wound healing in CXCR2 knockout mice. J Invest Dermatol. 2000;115:234–244. doi: 10.1046/j.1523-1747.2000.00034.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ebaid H. Neutrophil depletion in the early inflammatory phase delayed cutaneous wound healing in older rats: improvements due to the use of un-denatured camel whey protein. Diagn Pathol. 2014;9:46. doi: 10.1186/1746-1596-9-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Selders GS, Fetz AE, Radic MZ, Bowlin GL. An overview of the role of neutrophils in innate immunity, inflammation and host-biomaterial integration. Regen Biomater. 2017;4:55–68. doi: 10.1093/rb/rbw041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Brancato SK, Albina JE. Wound macrophages as key regulators of repair: origin, phenotype, and function. The American journal of pathology. 2011;178:19–25. doi: 10.1016/j.ajpath.2010.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mirza RE, Koh TJ. Contributions of cell subsets to cytokine production during normal and impaired wound healing. Cytokine. 2015;71:409–412. doi: 10.1016/j.cyto.2014.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Schultz GS, Wysocki A. Interactions between extracellular matrix and growth factors in wound healing. Wound repair and regeneration. 2009;17:153–162. doi: 10.1111/j.1524-475X.2009.00466.x. [DOI] [PubMed] [Google Scholar]
- 28.King A, Balaji S, Le LD, Crombleholme TM, Keswani SG. Regenerative Wound Healing: The Role of Interleukin-10. Adv Wound Care (New Rochelle) 2014;3:315–323. doi: 10.1089/wound.2013.0461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.DiPietro LA. Wound healing: the role of the macrophage and other immune cells. Shock. 1995;4:233–240. [PubMed] [Google Scholar]
- 30.Sindrilaru A, Scharffetter-Kochanek K. Disclosure of the Culprits: Macrophages-Versatile Regulators of Wound Healing. Adv Wound Care (New Rochelle) 2013;2:357–368. doi: 10.1089/wound.2012.0407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Sindrilaru A, Peters T, Wieschalka S, Baican C, Baican A, Peter H, Hainzl A, Schatz S, Qi Y, Schlecht A, Weiss JM, Wlaschek M, Sunderkotter C, Scharffetter-Kochanek K. An unrestrained proinflammatory M1 macrophage population induced by iron impairs wound healing in humans and mice. J Clin Invest. 2011;121:985–997. doi: 10.1172/JCI44490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Ploeger DT, Hosper NA, Schipper M, Koerts JA, de Rond S, Bank RA. Cell plasticity in wound healing: paracrine factors of M1/M2 polarized macrophages influence the phenotypical state of dermal fibroblasts. Cell Commun Signal. 2013;11:29. doi: 10.1186/1478-811X-11-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Mantovani A, Biswas SK, Galdiero MR, Sica A, Locati M. Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol. 2013;229:176–185. doi: 10.1002/path.4133. [DOI] [PubMed] [Google Scholar]
- 34.Jetten N, Verbruggen S, Gijbels MJ, Post MJ, De Winther MP, Donners MM. Anti-inflammatory M2, but not pro-inflammatory M1 macrophages promote angiogenesis in vivo. Angiogenesis. 2014;17:109–118. doi: 10.1007/s10456-013-9381-6. [DOI] [PubMed] [Google Scholar]
- 35.Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest. 2012;122:787–795. doi: 10.1172/JCI59643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Amini-Nik S, Cambridge E, Yu W, Guo A, Whetstone H, Nadesan P, Poon R, Hinz B, Alman BA. beta-Catenin-regulated myeloid cell adhesion and migration determine wound healing. J Clin Invest. 2014;124:2599–2610. doi: 10.1172/JCI62059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Mirza R, DiPietro LA, Koh TJ. Selective and specific macrophage ablation is detrimental to wound healing in mice. The American journal of pathology. 2009;175:2454–2462. doi: 10.2353/ajpath.2009.090248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Yuan R, Geng S, Chen K, Diao N, Chu HW, Li L. Low-grade inflammatory polarization of monocytes impairs wound healing. J Pathol. 2016;238:571–583. doi: 10.1002/path.4680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ogle ME, Segar CE, Sridhar S, Botchwey EA. Monocytes and macrophages in tissue repair: Implications for immunoregenerative biomaterial design. Exp Biol Med (Maywood) 2016;241:1084–1097. doi: 10.1177/1535370216650293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Gallucci RM, Simeonova PP, Matheson JM, Kommineni C, Guriel JL, Sugawara T, Luster MI. Impaired cutaneous wound healing in interleukin-6-deficient and immunosuppressed mice. FASEB J. 2000;14:2525–2531. doi: 10.1096/fj.00-0073com. [DOI] [PubMed] [Google Scholar]
- 41.Fang Y, Gong SJ, Xu YH, Hambly BD, Bao S. Impaired cutaneous wound healing in granulocyte/macrophage colony-stimulating factor knockout mice. Br J Dermatol. 2007;157:458–465. doi: 10.1111/j.1365-2133.2007.07979.x. [DOI] [PubMed] [Google Scholar]
- 42.Low QE, Drugea IA, Duffner LA, Quinn DG, Cook DN, Rollins BJ, Kovacs EJ, DiPietro LA. Wound healing in MIP-1alpha(−/−) and MCP-1(−/−) mice. Am J Pathol. 2001;159:457–463. doi: 10.1016/s0002-9440(10)61717-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ishida Y, Gao JL, Murphy PM. Chemokine receptor CX3CR1 mediates skin wound healing by promoting macrophage and fibroblast accumulation and function. J Immunol. 2008;180:569–579. doi: 10.4049/jimmunol.180.1.569. [DOI] [PubMed] [Google Scholar]
- 44.Macedo L, Pinhal-Enfield G, Alshits V, Elson G, Cronstein BN, Leibovich SJ. Wound healing is impaired in MyD88-deficient mice: a role for MyD88 in the regulation of wound healing by adenosine A2A receptors. The American journal of pathology. 2007;171:1774–1788. doi: 10.2353/ajpath.2007.061048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Venet F, Chung CS, Monneret G, Huang X, Horner B, Garber M, Ayala A. Regulatory T cell populations in sepsis and trauma. J Leukoc Biol. 2008;83:523–535. doi: 10.1189/jlb.0607371. [DOI] [PubMed] [Google Scholar]
- 46.Nosbaum A, Prevel N, Truong HA, Mehta P, Ettinger M, Scharschmidt TC, Ali NH, Pauli ML, Abbas AK, Rosenblum MD. Cutting Edge: Regulatory T Cells Facilitate Cutaneous Wound Healing. J Immunol. 2016;196:2010–2014. doi: 10.4049/jimmunol.1502139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Bodnar RJ. Chemokine Regulation of Angiogenesis During Wound Healing. Adv Wound Care (New Rochelle) 2015;4:641–650. doi: 10.1089/wound.2014.0594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Wahl SM, Wahl LM, McCarthy JB. Lymphocyte-mediated activation of fibroblast proliferation and collagen production. J Immunol. 1978;121:942–946. [PubMed] [Google Scholar]
- 49.Chen L, Mehta ND, Zhao Y, DiPietro LA. 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 Dermatol. 2014;23:189–194. doi: 10.1111/exd.12346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kreuger J, Phillipson M. Targeting vascular and leukocyte communication in angiogenesis, inflammation and fibrosis. Nat Rev Drug Discov. 2016;15:125–142. doi: 10.1038/nrd.2015.2. [DOI] [PubMed] [Google Scholar]
- 51.Martin P, Nunan R. Cellular and molecular mechanisms of repair in acute and chronic wound healing. Br J Dermatol. 2015;173:370–378. doi: 10.1111/bjd.13954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Med. 2014;6:265sr266. doi: 10.1126/scitranslmed.3009337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Thomas D, Thirumaran A, Mallard B, Chen X, Browne S, Wheatley AM, O’Brien T, Pandit A. Variability in Endogenous Perfusion Recovery of Immunocompromised Mouse Models of Limb Ischemia. Tissue Eng Part C Methods. 2016;22:370–381. doi: 10.1089/ten.TEC.2015.0441. [DOI] [PubMed] [Google Scholar]
- 54.Marques SM, Campos PP, Castro PR, Cardoso CC, Ferreira MA, Andrade SP. Genetic background determines mouse strain differences in inflammatory angiogenesis. Microvasc Res. 2011;82:246–252. doi: 10.1016/j.mvr.2011.08.011. [DOI] [PubMed] [Google Scholar]
- 55.Akers KS, Rowan MP, Niece KL, Graybill JC, Mende K, Chung KK, Murray CK. Antifungal wound penetration of amphotericin and voriconazole in combat-related injuries: case report. BMC Infect Dis. 2015;15:184. doi: 10.1186/s12879-015-0918-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Valente JF, Hricik D, Weigel K, Seaman D, Knauss T, Siegel CT, Bodziak K, Schulak JA. Comparison of sirolimus vs. mycophenolate mofetil on surgical complications and wound healing in adult kidney transplantation. Am J Transplant. 2003;3:1128–1134. doi: 10.1034/j.1600-6143.2003.00185.x. [DOI] [PubMed] [Google Scholar]
- 57.Schappi MG, Jaquet V, Belli DC, Krause KH. Hyperinflammation in chronic granulomatous disease and anti-inflammatory role of the phagocyte NADPH oxidase. Semin Immunopathol. 2008;30:255–271. doi: 10.1007/s00281-008-0119-2. [DOI] [PubMed] [Google Scholar]
- 58.Mirza RE, Fang MM, Weinheimer-Haus EM, Ennis WJ, Koh TJ. Sustained inflammasome activity in macrophages impairs wound healing in type 2 diabetic humans and mice. Diabetes. 2014;63:1103–1114. doi: 10.2337/db13-0927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Jun JI, Kim KH, Lau LF. The matricellular protein CCN1 mediates neutrophil efferocytosis in cutaneous wound healing. Nat Commun. 2015;6:7386. doi: 10.1038/ncomms8386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Emerging Risk Factors C. Sarwar N, Gao P, Seshasai SR, Gobin R, Kaptoge S, Di Angelantonio E, Ingelsson E, Lawlor DA, Selvin E, Stampfer M, Stehouwer CD, Lewington S, Pennells L, Thompson A, Sattar N, White IR, Ray KK, Danesh J. Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease: a collaborative meta-analysis of 102 prospective studies. Lancet. 2010;375:2215–2222. doi: 10.1016/S0140-6736(10)60484-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Gallagher KA, Joshi A, Carson WF, Schaller M, Allen R, Mukerjee S, Kittan N, Feldman EL, Henke PK, Hogaboam C, Burant CF, Kunkel SL. Epigenetic changes in bone marrow progenitor cells influence the inflammatory phenotype and alter wound healing in type 2 diabetes. Diabetes. 2015;64:1420–1430. doi: 10.2337/db14-0872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Cianfarani F, Toietta G, Di Rocco G, Cesareo E, Zambruno G, Odorisio T. Diabetes impairs adipose tissue-derived stem cell function and efficiency in promoting wound healing. Wound Repair Regen. 2013;21:545–553. doi: 10.1111/wrr.12051. [DOI] [PubMed] [Google Scholar]
- 63.Falanga V. Wound healing and its impairment in the diabetic foot. Lancet. 2005;366:1736–1743. doi: 10.1016/S0140-6736(05)67700-8. [DOI] [PubMed] [Google Scholar]
- 64.Murray CJ, Ortblad KF, Guinovart C, Lim SS, Wolock TM, Roberts DA, Dansereau EA, Graetz N, Barber RM, Brown JC, Wang H, Duber HC, Naghavi M, Dicker D, Dandona L, Salomon JA, Heuton KR, Foreman K, Phillips DE, Fleming TD, Flaxman AD, Phillips BK, Johnson EK, Coggeshall MS, Abd-Allah F, Abera SF, Abraham JP, Abubakar I, Abu-Raddad LJ, Abu-Rmeileh NM, Achoki T, Adeyemo AO, Adou AK, Adsuar JC, Agardh EE, Akena D, Al Kahbouri MJ, Alasfoor D, Albittar MI, Alcala-Cerra G, Alegretti MA, Alemu ZA, Alfonso-Cristancho R, Alhabib S, Ali R, Alla F, Allen PJ, Alsharif U, Alvarez E, Alvis-Guzman N, Amankwaa AA, Amare AT, Amini H, Ammar W, Anderson BO, Antonio CA, Anwari P, Arnlov J, Arsenijevic VS, Artaman A, Asghar RJ, Assadi R, Atkins LS, Badawi A, Balakrishnan K, Banerjee A, Basu S, Beardsley J, Bekele T, Bell ML, Bernabe E, Beyene TJ, Bhala N, Bhalla A, Bhutta ZA, Abdulhak AB, Binagwaho A, Blore JD, Basara BB, Bose D, Brainin M, Breitborde N, Castaneda-Orjuela CA, Catala-Lopez F, Chadha VK, Chang JC, Chiang PP, Chuang TW, Colomar M, Cooper LT, Cooper C, Courville KJ, Cowie BC, Criqui MH, Dandona R, Dayama A, De Leo D, Degenhardt L, Del Pozo-Cruz B, Deribe K, Des Jarlais DC, Dessalegn M, Dharmaratne SD, Dilmen U, Ding EL, Driscoll TR, Durrani AM, Ellenbogen RG, Ermakov SP, Esteghamati A, Faraon EJ, Farzadfar F, Fereshtehnejad SM, Fijabi DO, Forouzanfar MH, Fra Paleo U, Gaffikin L, Gamkrelidze A, Gankpe FG, Geleijnse JM, Gessner BD, Gibney KB, Ginawi IA, Glaser EL, Gona P, Goto A, Gouda HN, Gugnani HC, Gupta R, Gupta R, Hafezi-Nejad N, Hamadeh RR, Hammami M, Hankey GJ, Harb HL, Haro JM, Havmoeller R, Hay SI, Hedayati MT, Pi IB, Hoek HW, Hornberger JC, Hosgood HD, Hotez PJ, Hoy DG, Huang JJ, Iburg KM, Idrisov BT, Innos K, Jacobsen KH, Jeemon P, Jensen PN, Jha V, Jiang G, Jonas JB, Juel K, Kan H, Kankindi I, Karam NE, Karch A, Karema CK, Kaul A, Kawakami N, Kazi DS, Kemp AH, Kengne AP, Keren A, Kereselidze M, Khader YS, Khalifa SE, Khan EA, Khang YH, Khonelidze I, Kinfu Y, Kinge JM, Knibbs L, Kokubo Y, Kosen S, Defo BK, Kulkarni VS, Kulkarni C, Kumar K, Kumar RB, Kumar GA, Kwan GF, Lai T, Balaji AL, Lam H, Lan Q, Lansingh VC, Larson HJ, Larsson A, Lee JT, Leigh J, Leinsalu M, Leung R, Li Y, Li Y, De Lima GM, Lin HH, Lipshultz SE, Liu S, Liu Y, Lloyd BK, Lotufo PA, Machado VM, Maclachlan JH, Magis-Rodriguez C, Majdan M, Mapoma CC, Marcenes W, Marzan MB, Masci JR, Mashal MT, Mason-Jones AJ, Mayosi BM, Mazorodze TT, McKay AC, Meaney PA, Mehndiratta MM, Mejia-Rodriguez F, Melaku YA, Memish ZA, Mendoza W, Miller TR, Mills EJ, Mohammad KA, Mokdad AH, Mola GL, Monasta L, Montico M, Moore AR, Mori R, Moturi WN, Mukaigawara M, Murthy KS, Naheed A, Naidoo KS, Naldi L, Nangia V, Narayan KM, Nash D, Nejjari C, Nelson RG, Neupane SP, Newton CR, Ng M, Nisar MI, Nolte S, Norheim OF, Nowaseb V, Nyakarahuka L, Oh IH, Ohkubo T, Olusanya BO, Omer SB, Opio JN, Orisakwe OE, Pandian JD, Papachristou C, Caicedo AJ, Patten SB, Paul VK, Pavlin BI, Pearce N, Pereira DM, Pervaiz A, Pesudovs K, Petzold M, Pourmalek F, Qato D, Quezada AD, Quistberg DA, Rafay A, Rahimi K, Rahimi-Movaghar V, Ur Rahman S, Raju M, Rana SM, Razavi H, Reilly RQ, Remuzzi G, Richardus JH, Ronfani L, Roy N, Sabin N, Saeedi MY, Sahraian MA, Samonte GM, Sawhney M, Schneider IJ, Schwebel DC, Seedat S, Sepanlou SG, Servan-Mori EE, Sheikhbahaei S, Shibuya K, Shin HH, Shiue I, Shivakoti R, Sigfusdottir ID, Silberberg DH, Silva AP, Simard EP, Singh JA, Skirbekk V, Sliwa K, Soneji S, Soshnikov SS, Sreeramareddy CT, Stathopoulou VK, Stroumpoulis K, Swaminathan S, Sykes BL, Tabb KM, Talongwa RT, Tenkorang EY, Terkawi AS, Thomson AJ, Thorne-Lyman AL, Towbin JA, Traebert J, Tran BX, Dimbuene ZT, Tsilimbaris M, Uchendu US, Ukwaja KN, Uzun SB, Vallely AJ, Vasankari TJ, Venketasubramanian N, Violante FS, Vlassov VV, Vollset SE, Waller S, Wallin MT, Wang L, Wang X, Wang Y, Weichenthal S, Weiderpass E, Weintraub RG, Westerman R, White RA, Wilkinson JD, Williams TN, Woldeyohannes SM, Wong JQ, Xu G, Yang YC, Yano Y, Yentur GK, Yip P, Yonemoto N, Yoon SJ, Younis M, Yu C, Jin KY, El Sayed Zaki M, Zhao Y, Zheng Y, Zhou M, Zhu J, Zou XN, Lopez AD, Vos T. Global, regional, and national incidence and mortality for HIV, tuberculosis, and malaria during 1990–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2014;384:1005–1070. doi: 10.1016/S0140-6736(14)60844-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Kigozi G, Musoke R, Kighoma N, Watya S, Serwadda D, Nalugoda F, Kiwanuka N, Nkale J, Wabwire-Mangen F, Makumbi F, Sewankambo NK, Gray RH, Wawer MJ. Male circumcision wound healing in human immunodeficiency virus (HIV)-negative and HIV-positive men in Rakai, Uganda. BJU Int. 2014;113:127–132. doi: 10.1111/bju.12406. [DOI] [PubMed] [Google Scholar]
- 66.Howard N, Phaff M, Aird J, Wicks L, Rollinson P. Does human immunodeficiency virus status affect early wound healing in open surgically stabilised tibial fractures? Bone Joint J. 2013;95:1703–1707. doi: 10.1302/0301-620X.95B12.32083. [DOI] [PubMed] [Google Scholar]
- 67.Ata-Ali J, Ata-Ali F, Di-Benedetto N, Bagán L, Bagán J-V. Does HIV infection have an impact upon dental implant osseointegration? A systematic review. Medicina oral, patologia oral y cirugia bucal. 2015;20:e347. doi: 10.4317/medoral.20408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Harrison WJ, Lewis CP, Lavy CB. Wound healing after implant surgery in HIV-positive patients. J Bone Joint Surg Br. 2002;84:802–806. doi: 10.1302/0301-620x.84b6.12641. [DOI] [PubMed] [Google Scholar]
- 69.Lord RV. Anorectal surgery in patients infected with human immunodeficiency virus: factors associated with delayed wound healing. Ann Surg. 1997;226:92–99. doi: 10.1097/00000658-199707000-00013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Vacanti JP, Langer R. Tissue engineering: the design and fabrication of living replacement devices for surgical reconstruction and transplantation. Lancet. 1999;354(Suppl 1):SI32–34. doi: 10.1016/s0140-6736(99)90247-7. [DOI] [PubMed] [Google Scholar]
- 71.Patrick CW., Jr Adipose tissue engineering: the future of breast and soft tissue reconstruction following tumor resection. Semin Surg Oncol. 2000;19:302–311. doi: 10.1002/1098-2388(200010/11)19:3<302::aid-ssu12>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
- 72.Bland KI, Palin WE, von Fraunhofer JA, Morris RR, Adcock RA, Tobin GR., 2nd Experimental and clinical observations of the effects of cytotoxic chemotherapeutic drugs on wound healing. Ann Surg. 1984;199:782–790. doi: 10.1097/00000658-198406000-00017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Wu Y, Wang J, Scott PG, Tredget EE. Bone marrow-derived stem cells in wound healing: a review. Wound Repair Regen. 2007;15(Suppl 1):S18–26. doi: 10.1111/j.1524-475X.2007.00221.x. [DOI] [PubMed] [Google Scholar]
- 74.Chang HY, Sneddon JB, Alizadeh AA, Sood R, West RB, Montgomery K, Chi JT, van de Rijn M, Botstein D, Brown PO. Gene expression signature of fibroblast serum response predicts human cancer progression: similarities between tumors and wounds. PLoS Biol. 2004;2:E7. doi: 10.1371/journal.pbio.0020007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Scappaticci FA, Fehrenbacher L, Cartwright T, Hainsworth JD, Heim W, Berlin J, Kabbinavar F, Novotny W, Sarkar S, Hurwitz H. Surgical wound healing complications in metastatic colorectal cancer patients treated with bevacizumab. J Surg Oncol. 2005;91:173–180. doi: 10.1002/jso.20301. [DOI] [PubMed] [Google Scholar]
- 76.Seiwert TY, Haraf DJ, Cohen EE, Stenson K, Witt ME, Dekker A, Kocherginsky M, Weichselbaum RR, Chen HX, Vokes EE. Phase I study of bevacizumab added to fluorouracil- and hydroxyurea-based concomitant chemoradiotherapy for poor-prognosis head and neck cancer. J Clin Oncol. 2008;26:1732–1741. doi: 10.1200/JCO.2007.13.1706. [DOI] [PubMed] [Google Scholar]
- 77.Haubner F, Ohmann E, Pohl F, Strutz J, Gassner HG. Wound healing after radiation therapy: review of the literature. Radiat Oncol. 2012;7:162. doi: 10.1186/1748-717X-7-162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Weber JS, Postow M, Lao CD, Schadendorf D. Management of Adverse Events Following Treatment With Anti-Programmed Death-1 Agents. Oncologist. 2016;21:1230–1240. doi: 10.1634/theoncologist.2016-0055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Wang AS, Armstrong EJ, Armstrong AW. Corticosteroids and wound healing: clinical considerations in the perioperative period. Am J Surg. 2013;206:410–417. doi: 10.1016/j.amjsurg.2012.11.018. [DOI] [PubMed] [Google Scholar]
- 80.Pountos I, Giannoudis PV. Effect of methotrexate on bone and wound healing. Expert Opin Drug Saf. 2017;16:535–545. doi: 10.1080/14740338.2017.1310839. [DOI] [PubMed] [Google Scholar]
- 81.Hofmann SR, Rosen-Wolff A, Tsokos GC, Hedrich CM. Biological properties and regulation of IL-10 related cytokines and their contribution to autoimmune disease and tissue injury. Clin Immunol. 2012;143:116–127. doi: 10.1016/j.clim.2012.02.005. [DOI] [PubMed] [Google Scholar]
- 82.Betker JL, Anchordoquy TJ. Assessing the effect of a nude mouse model on nanoparticle-mediated gene delivery. Drug Deliv Transl Res. 2017;7:162–167. doi: 10.1007/s13346-016-0327-6. [DOI] [PubMed] [Google Scholar]
- 83.Groves RW, Schmidt-Lucke JA. Recombinant human GM-CSF in the treatment of poorly healing wounds. Adv Skin Wound Care. 2000;13:107–112. [PubMed] [Google Scholar]
- 84.Pettit DK, Lawter JR, Huang WJ, Pankey SC, Nightlinger NS, Lynch DH, Schuh JA, Morrissey PJ, Gombotz WR. Characterization of poly(glycolide-co-D,L-lactide)/poly(D,L-lactide) microspheres for controlled release of GM-CSF. Pharm Res. 1997;14:1422–1430. doi: 10.1023/a:1012176823155. [DOI] [PubMed] [Google Scholar]
- 85.Burgess AW, Metcalf D. Serum half-life and organ distribution of radiolabeled colony stimulating factor in mice. Exp Hematol. 1977;5:456–464. [PubMed] [Google Scholar]
- 86.Zaharoff DA, Rogers CJ, Hance KW, Schlom J, Greiner JW. Chitosan solution enhances the immunoadjuvant properties of GM-CSF. Vaccine. 2007;25:8673–8686. doi: 10.1016/j.vaccine.2007.10.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Watson E, Tatara AM, Kontoyiannis DP, Mikos AG. Inherently Antimicrobial Biodegradable Polymers in Tissue Engineering. ACS Biomaterials Science & Engineering. 2016;3:1207–1220. doi: 10.1021/acsbiomaterials.6b00501. [DOI] [PubMed] [Google Scholar]
- 88.Hasan A, Waibhaw G, Tiwari S, Dharmalingam K, Shukla I, Pandey LM. Fabrication and characterization of chitosan, polyvinylpyrrolidone, and cellulose nanowhiskers nanocomposite films for wound healing drug delivery application. Journal of Biomedical Materials Research Part A. 2017 doi: 10.1002/jbm.a.36097. [DOI] [PubMed] [Google Scholar]
- 89.Sezer AD, Akbuga J. Comparison on in vitro characterization of fucospheres and chitosan microspheres encapsulated plasmid DNA (pGM-CSF): formulation design and release characteristics. AAPS PharmSciTech. 2009;10:1193–1199. doi: 10.1208/s12249-009-9324-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Kedar E, Palgi O, Golod G, Babai I, Barenholz Y. Delivery of cytokines by liposomes. III. Liposome-encapsulated GM-CSF and TNF-alpha show improved pharmacokinetics and biological activity and reduced toxicity in mice. J Immunother. 1997;20:180–193. doi: 10.1097/00002371-199705000-00003. [DOI] [PubMed] [Google Scholar]
- 91.Steinwede K, Tempelhof O, Bolte K, Maus R, Bohling J, Ueberberg B, Langer F, Christman JW, Paton JC, Ask K, Maharaj S, Kolb M, Gauldie J, Welte T, Maus UA. Local delivery of GM-CSF protects mice from lethal pneumococcal pneumonia. J Immunol. 2011;187:5346–5356. doi: 10.4049/jimmunol.1101413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Cho SH, Kim A, Shin W, Heo MB, Noh HJ, Hong KS, Cho JH, Lim YT. Photothermal-modulated drug delivery and magnetic relaxation based on collagen/poly(gamma-glutamic acid) hydrogel. Int J Nanomedicine. 2017;12:2607–2620. doi: 10.2147/IJN.S133078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kim YH, Tabata Y. Recruitment of mesenchymal stem cells and macrophages by dual release of stromal cell-derived factor-1 and a macrophage recruitment agent enhances wound closure. J Biomed Mater Res A. 2016;104:942–956. doi: 10.1002/jbm.a.35635. [DOI] [PubMed] [Google Scholar]
- 94.Kim YH, Furuya H, Tabata Y. Enhancement of bone regeneration by dual release of a macrophage recruitment agent and platelet-rich plasma from gelatin hydrogels. Biomaterials. 2014;35:214–224. doi: 10.1016/j.biomaterials.2013.09.103. [DOI] [PubMed] [Google Scholar]
- 95.Das A, Segar CE, Hughley BB, Bowers DT, Botchwey EA. The promotion of mandibular defect healing by the targeting of S1P receptors and the recruitment of alternatively activated macrophages. Biomaterials. 2013;34:9853–9862. doi: 10.1016/j.biomaterials.2013.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Gallucci RM, Sugawara T, Yucesoy B, Berryann K, Simeonova PP, Matheson JM, Luster MI. Interleukin-6 treatment augments cutaneous wound healing in immunosuppressed mice. J Interferon Cytokine Res. 2001;21:603–609. doi: 10.1089/10799900152547867. [DOI] [PubMed] [Google Scholar]
- 97.Rao AJ, Gibon E, Ma T, Yao Z, Smith RL, Goodman SB. Revision joint replacement, wear particles, and macrophage polarization. Acta Biomater. 2012;8:2815–2823. doi: 10.1016/j.actbio.2012.03.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Rao AJ, Nich C, Dhulipala LS, Gibon E, Valladares R, Zwingenberger S, Smith RL, Goodman SB. Local effect of IL-4 delivery on polyethylene particle induced osteolysis in the murine calvarium. J Biomed Mater Res A. 2013;101:1926–1934. doi: 10.1002/jbm.a.34486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Reeves AR, Spiller KL, Freytes DO, Vunjak-Novakovic G, Kaplan DL. Controlled release of cytokines using silk-biomaterials for macrophage polarization. Biomaterials. 2015;73:272–283. doi: 10.1016/j.biomaterials.2015.09.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Spiller KL, Nassiri S, Witherel CE, Anfang RR, Ng J, Nakazawa KR, Yu T, Vunjak-Novakovic G. Sequential delivery of immunomodulatory cytokines to facilitate the M1-to-M2 transition of macrophages and enhance vascularization of bone scaffolds. Biomaterials. 2015;37:194–207. doi: 10.1016/j.biomaterials.2014.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Kumar VA, Taylor NL, Shi S, Wickremasinghe NC, D’Souza RN, Hartgerink JD. Self-assembling multidomain peptides tailor biological responses through biphasic release. Biomaterials. 2015;52:71–78. doi: 10.1016/j.biomaterials.2015.01.079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Vasconcelos DP, Costa M, Amaral IF, Barbosa MA, Aguas AP, Barbosa JN. Modulation of the inflammatory response to chitosan through M2 macrophage polarization using pro-resolution mediators. Biomaterials. 2015;37:116–123. doi: 10.1016/j.biomaterials.2014.10.035. [DOI] [PubMed] [Google Scholar]
- 103.Lohmann N, Schirmer L, Atallah P, Wandel E, Ferrer RA, Werner C, Simon JC, Franz S, Freudenberg U. Glycosaminoglycan-based hydrogels capture inflammatory chemokines and rescue defective wound healing in mice. Sci Transl Med. 2017;9 doi: 10.1126/scitranslmed.aai9044. [DOI] [PubMed] [Google Scholar]
- 104.Bota PC, Collie AM, Puolakkainen P, Vernon RB, Sage EH, Ratner BD, Stayton PS. Biomaterial topography alters healing in vivo and monocyte/macrophage activation in vitro. J Biomed Mater Res A. 2010;95:649–657. doi: 10.1002/jbm.a.32893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Saino E, Focarete ML, Gualandi C, Emanuele E, Cornaglia AI, Imbriani M, Visai L. Effect of electrospun fiber diameter and alignment on macrophage activation and secretion of proinflammatory cytokines and chemokines. Biomacromolecules. 2011;12:1900–1911. doi: 10.1021/bm200248h. [DOI] [PubMed] [Google Scholar]
- 106.Bartneck M, Schulte VA, Paul NE, Diez M, Lensen MC, Zwadlo-Klarwasser G. Induction of specific macrophage subtypes by defined micro-patterned structures. Acta Biomater. 2010;6:3864–3872. doi: 10.1016/j.actbio.2010.04.025. [DOI] [PubMed] [Google Scholar]
- 107.McWhorter FY, Davis CT, Liu WF. Physical and mechanical regulation of macrophage phenotype and function. Cell Mol Life Sci. 2015;72:1303–1316. doi: 10.1007/s00018-014-1796-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Brown GL, Nanney LB, Griffen J, Cramer AB, Yancey JM, Curtsinger LJ, 3rd, Holtzin L, Schultz GS, Jurkiewicz MJ, Lynch JB. Enhancement of wound healing by topical treatment with epidermal growth factor. N Engl J Med. 1989;321:76–79. doi: 10.1056/NEJM198907133210203. [DOI] [PubMed] [Google Scholar]
- 109.Robson MC, Phillips LG, Thomason A, Robson LE, Pierce GF. Platelet-derived growth factor BB for the treatment of chronic pressure ulcers. Lancet. 1992;339:23–25. doi: 10.1016/0140-6736(92)90143-q. [DOI] [PubMed] [Google Scholar]
- 110.Martin A, Komada MR, Sane DC. Abnormal angiogenesis in diabetes mellitus. Med Res Rev. 2003;23:117–145. doi: 10.1002/med.10024. [DOI] [PubMed] [Google Scholar]
- 111.Mizuno K, Yamamura K, Yano K, Osada T, Saeki S, Takimoto N, Sakurai T, Nimura Y. Effect of chitosan film containing basic fibroblast growth factor on wound healing in genetically diabetic mice. J Biomed Mater Res A. 2003;64:177–181. doi: 10.1002/jbm.a.10396. [DOI] [PubMed] [Google Scholar]
- 112.Losi P, Briganti E, Errico C, Lisella A, Sanguinetti E, Chiellini F, Soldani G. Fibrin-based scaffold incorporating VEGF- and bFGF-loaded nanoparticles stimulates wound healing in diabetic mice. Acta Biomater. 2013;9:7814–7821. doi: 10.1016/j.actbio.2013.04.019. [DOI] [PubMed] [Google Scholar]
- 113.Yang Y, Xia T, Zhi W, Wei L, Weng J, Zhang C, Li X. Promotion of skin regeneration in diabetic rats by electrospun core-sheath fibers loaded with basic fibroblast growth factor. Biomaterials. 2011;32:4243–4254. doi: 10.1016/j.biomaterials.2011.02.042. [DOI] [PubMed] [Google Scholar]
- 114.Hardwicke J, Schmaljohann D, Boyce D, Thomas D. Epidermal growth factor therapy and wound healing—past, present and future perspectives. The Surgeon. 2008;6:172–177. doi: 10.1016/s1479-666x(08)80114-x. [DOI] [PubMed] [Google Scholar]
- 115.Tiwari V. Burn wound: How it differs from other wounds? Indian journal of plastic surgery: official publication of the Association of Plastic Surgeons of India. 2012;45:364. doi: 10.4103/0970-0358.101319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Jahovic N, Guzel E, Arbak S, Yegen BC. The healing-promoting effect of saliva on skin burn is mediated by epidermal growth factor (EGF): role of the neutrophils. Burns. 2004;30:531–538. doi: 10.1016/j.burns.2004.02.007. [DOI] [PubMed] [Google Scholar]
- 117.Alemdaroglu C, Degim Z, Celebi N, Zor F, Ozturk S, Erdogan D. An investigation on burn wound healing in rats with chitosan gel formulation containing epidermal growth factor. Burns. 2006;32:319–327. doi: 10.1016/j.burns.2005.10.015. [DOI] [PubMed] [Google Scholar]
- 118.Yenilmez E, Basaran E, Arslan R, Berkman MS, Guven UM, Baycu C, Yazan Y. Chitosan gel formulations containing egg yolk oil and epidermal growth factor for dermal burn treatment. Pharmazie. 2015;70:67–73. [PubMed] [Google Scholar]
- 119.Degim Z, Celebi N, Alemdaroglu C, Deveci M, Ozturk S, Ozogul C. Evaluation of chitosan gel containing liposome-loaded epidermal growth factor on burn wound healing. Int Wound J. 2011;8:343–354. doi: 10.1111/j.1742-481X.2011.00795.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Gwak SJ, Kim SS, Sung K, Han J, Choi CY, Kim BS. Synergistic effect of keratinocyte transplantation and epidermal growth factor delivery on epidermal regeneration. Cell Transplant. 2005;14:809–817. doi: 10.3727/000000005783982521. [DOI] [PubMed] [Google Scholar]
- 121.Matsumoto Y, Kuroyanagi Y. Development of a wound dressing composed of hyaluronic acid sponge containing arginine and epidermal growth factor. J Biomater Sci Polym Ed. 2010;21:715–726. doi: 10.1163/156856209X435844. [DOI] [PubMed] [Google Scholar]
- 122.Geer DJ, Swartz DD, Andreadis ST. Biomimetic delivery of keratinocyte growth factor upon cellular demand for accelerated wound healing in vitro and in vivo. The American journal of pathology. 2005;167:1575–1586. doi: 10.1016/S0002-9440(10)61242-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Li B, Davidson JM, Guelcher SA. The effect of the local delivery of platelet-derived growth factor from reactive two-component polyurethane scaffolds on the healing in rat skin excisional wounds. Biomaterials. 2009;30:3486–3494. doi: 10.1016/j.biomaterials.2009.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Puolakkainen PA, Twardzik DR, Ranchalis JE, Pankey SC, Reed MJ, Gombotz WR. The enhancement in wound healing by transforming growth factor-beta 1 (TGF-beta 1) depends on the topical delivery system. J Surg Res. 1995;58:321–329. doi: 10.1006/jsre.1995.1050. [DOI] [PubMed] [Google Scholar]
- 125.Chen RR, Mooney DJ. Polymeric growth factor delivery strategies for tissue engineering. Pharm Res. 2003;20:1103–1112. doi: 10.1023/a:1025034925152. [DOI] [PubMed] [Google Scholar]
- 126.Van Dyke TE, Offenbacher S, Braswell L, Lessem J. Enhancing the value of scaling and root-planing: Arestin clinical trial results. J Int Acad Periodontol. 2002;4:72–76. [PubMed] [Google Scholar]
- 127.Nelson CL, Griffin FM, Harrison BH, Cooper RE. In vitro elution characteristics of commercially and noncommercially prepared antibiotic PMMA beads. Clin Orthop Relat Res. 1992:303–309. [PubMed] [Google Scholar]
- 128.ter Boo GJ, Grijpma DW, Moriarty TF, Richards RG, Eglin D. Antimicrobial delivery systems for local infection prophylaxis in orthopedic- and trauma surgery. Biomaterials. 2015;52:113–125. doi: 10.1016/j.biomaterials.2015.02.020. [DOI] [PubMed] [Google Scholar]
- 129.Hasan A, Pandey LM. Polymers, Surface-Modified Polymers, and Self Assembled Monolayers as Surface-Modifying Agents for Biomaterials. Polymer-Plastics Technology and Engineering. 2015;54:1358–1378. [Google Scholar]
- 130.Saxena V, Hasan A, Sharma S, Pandey LM. Edible oil nanoemulsion: An organic nanoantibiotic as a potential biomolecule delivery vehicle. International Journal of Polymeric Materials and Polymeric Biomaterials. 2017:1–10. [Google Scholar]
- 131.Burt S. Essential oils: their antibacterial properties and potential applications in foods—a review. International journal of food microbiology. 2004;94:223–253. doi: 10.1016/j.ijfoodmicro.2004.03.022. [DOI] [PubMed] [Google Scholar]
- 132.Baistrocchi SR, Lee MJ, Lehoux M, Ralph B, Snarr BD, Robitaille R, Sheppard DC. Posaconazole-Loaded Leukocytes as a Novel Treatment Strategy Targeting Invasive Pulmonary Aspergillosis. J Infect Dis. 2016 doi: 10.1093/infdis/jiw513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Golub LM, Sorsa T, Lee HM, Ciancio S, Sorbi D, Ramamurthy NS, Gruber B, Salo T, Konttinen YT. Doxycycline inhibits neutrophil (PMN)-type matrix metalloproteinases in human adult periodontitis gingiva. J Clin Periodontol. 1995;22:100–109. doi: 10.1111/j.1600-051x.1995.tb00120.x. [DOI] [PubMed] [Google Scholar]
- 134.Rathbone CR, Cross JD, Brown KV, Murray CK, Wenke JC. Effect of various concentrations of antibiotics on osteogenic cell viability and activity. Journal of Orthopaedic Research. 2011;29:1070–1074. doi: 10.1002/jor.21343. [DOI] [PubMed] [Google Scholar]
- 135.Hebda PA, Whaley D, Kim HG, Wells A. Absence of inhibition of cutaneous wound healing in mice by oral doxycycline. Wound Repair Regen. 2003;11:373–379. doi: 10.1046/j.1524-475x.2003.11510.x. [DOI] [PubMed] [Google Scholar]
- 136.Felts AG, Giridhar G, Grainger DW, Slunt JB. Efficacy of locally delivered polyclonal immunoglobulin against Pseudomonas aeruginosa infection in a murine burn wound model. Burns. 1999;25:415–423. doi: 10.1016/s0305-4179(99)00017-0. [DOI] [PubMed] [Google Scholar]
- 137.Adam VS, Crosariol M, Kumar S, Ge MQ, Czack SE, Roy S, Haczku A, Tretiakova A, Wilson JM, Limberis MP. Adeno-associated virus 9-mediated airway expression of antibody protects old and immunodeficient mice against influenza virus. Clin Vaccine Immunol. 2014;21:1528–1533. doi: 10.1128/CVI.00572-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Kim E, Okada K, Beeler JA, Crim RL, Piedra PA, Gilbert BE, Gambotto A. Development of an adenovirus-based respiratory syncytial virus vaccine: preclinical evaluation of efficacy, immunogenicity, and enhanced disease in a cotton rat model. J Virol. 2014;88:5100–5108. doi: 10.1128/JVI.03194-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Ben-Ami R, Lewis RE, Leventakos K, Kontoyiannis DP. Aspergillus fumigatus inhibits angiogenesis through the production of gliotoxin and other secondary metabolites. Blood. 2009;114:5393–5399. doi: 10.1182/blood-2009-07-231209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Tatara AM, Mikos AG, Kontoyiannis DP. Factors affecting patient outcome in primary cutaneous aspergillosis. Medicine (Baltimore) 2016;95:e3747. doi: 10.1097/MD.0000000000003747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Huang E, Hewitt RG, Shelton M, Morse GD. Comparison of measured and estimated creatinine clearance in patients with advanced HIV disease. Pharmacotherapy. 1996;16:222–229. [PubMed] [Google Scholar]
- 142.Gwilt PR, Nahhas RR, Tracewell WG. The effects of diabetes mellitus on pharmacokinetics and pharmacodynamics in humans. Clin Pharmacokinet. 1991;20:477–490. doi: 10.2165/00003088-199120060-00004. [DOI] [PubMed] [Google Scholar]
- 143.Shive MS, Anderson JM. Biodegradation and biocompatibility of PLA and PLGA microspheres. Adv Drug Deliv Rev. 1997;28:5–24. doi: 10.1016/s0169-409x(97)00048-3. [DOI] [PubMed] [Google Scholar]
- 144.Ali SA, Doherty PJ, Williams DF. Molecular biointeractions of biomedical polymers with extracellular exudate and inflammatory cells and their effects on the biocompatibility, in vivo. Biomaterials. 1994;15:779–785. doi: 10.1016/0142-9612(94)90032-9. [DOI] [PubMed] [Google Scholar]
- 145.McIntyre MK, Peacock TJ, Akers KS, Burmeister DM. Initial Characterization of the Pig Skin Bacteriome and Its Effect on In Vitro Models of Wound Healing. PLoS One. 2016;11:e0166176. doi: 10.1371/journal.pone.0166176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Ghannoum M. Cooperative Evolutionary Strategy between the Bacteriome and Mycobiome. mBio. 2016;7:e01951–01916. doi: 10.1128/mBio.01951-16. [DOI] [PMC free article] [PubMed] [Google Scholar]