Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2023 Feb 20;32(5):570–587. doi: 10.1111/exd.14734

Immune cells and associated molecular markers in dermal fibrosis with focus on raised cutaneous scars

Elvis Banboye Kidzeru 1,2, Maribanyana Lebeko 1,5, Jyoti Rajan Sharma 1,3,6, Lucia Nkengazong 2, Henry Ademola Adeola 1, Hlumani Ndlovu 4, Nonhlanhla P Khumalo 1, Ardeshir Bayat 1,
PMCID: PMC10947010  PMID: 36562321

Abstract

Raised dermal scars including hypertrophic, and keloid scars as well as scalp‐associated fibrosing Folliculitis Keloidalis Nuchae (FKN) are a group of fibrotic raised dermal lesions that mostly occur following cutaneous injury. They are characterized by increased extracellular matrix (ECM) deposition, primarily excessive collagen type 1 production by hyperproliferative fibroblasts. The extent of ECM deposition is thought to be proportional to the severity of local skin inflammation leading to excessive fibrosis of the dermis. Due to a lack of suitable study models, therapy for raised dermal scars remains ill‐defined. Immune cells and their associated markers have been strongly associated with dermal fibrosis. Therefore, modulation of the immune system and use of anti‐inflammatory cytokines are of potential interest in the management of dermal fibrosis. In this review, we will discuss the importance of immune factors in the pathogenesis of raised dermal scarring. The aim here is to provide an up‐to‐date comprehensive review of the literature, from PubMed, Scopus, and other relevant search engines in order to describe the known immunological factors associated with raised dermal scarring. The importance of immune cells including mast cells, macrophages, lymphocytes, and relevant molecules such as cytokines, chemokines, and growth factors, antibodies, transcription factors, and other immune‐associated molecules as well as tissue lymphoid aggregates identified within raised dermal scars will be presented. A growing body of evidence points to a shift from proinflammatory Th1 response to regulatory/anti‐inflammatory Th2 response being associated with the development of fibrogenesis in raised dermal scarring. In summary, a better understanding of immune cells and associated molecular markers in dermal fibrosis will likely enable future development of potential immune‐modulated therapeutic, diagnostic, and theranostic targets in raised dermal scarring.

Keywords: Folliculitis Keloidalis Nuchae, hypertrophic scars, immune targets, keloid disease, raised dermal scarring

1. INTRODUCTION

Fibrosis was first described in the 19th century and the medical term “fibrosis” was derived from the Latin word “fibra” meaning fibre and the Greek/Latin suffix “oasis” which describes a physiological state or process usually being abnormal or diseased. 1 The extent and intensity of extracellular matrix (ECM) deposition by fibroblasts beyond normotrophic requirements is considered to be a characteristic hallmark of fibrotic tissue formation and is proportional to the severity of inflammation leading to the onset and subsequent development of skin fibrotic disorders. Fibrosis equally plays a pivotal role in the development of chronic diseases, and several fibrotic conditions have been described as affecting the kidney, liver, lung, eye, pancreas, intestine, brain, and joints. 2 , 3 , 4 , 5 Cutaneous fibroproliferative disorders characterized by excessive scarring can cause a visual aesthetic disfigurement as well as physcial symptoms which may lead to psychosocial issues. 6 , 7 , 8 , 9 , 10 , 11

Although they manifest differently, hypertrophic scars (HTS), keloid disease (KD), and fibrotic scalp Folliculitis Keloidalis Nuchae (FKN) have several clinical aspects that appear to be similar, as they are all raised cutaneous fibrotic disorders with common dysregulation of connective tissue pathobiology and metabolism leading to excessive collagen and ECM deposition (Figure 1). The intensity of ECM deposition (the hallmark of fibrosis) in raised dermal scars is proportional to the severity of fibrosis and has therefore been used for their characterization. 12 , 13

FIGURE 1.

FIGURE 1

The spectrum of wound healing, from normal scars (includes fine line flat and stretched scars) to raised dermal scars. Raised dermal scars comprise of hypertrophic scars and keloids, which are characterized by growth within and beyond wound margins, respectively.

Key aspects of the wound healing process play a critical role in development of fibrosis and particularly in formation of raised dermal scarring. 14 , 15 Raised dermal scarring often manifests from trauma or an injury to the deep connective tissue layer of the skin and could be considered as a complication or a pathologically abnormal form of the wound healing process. 89 Keloid disease together with other fibrotic conditions including HTS 67 and FKN 10 , 11 are the end result of uncontrolled inflammation leading to excessive collagen deposition and increased ECM production. 12 , 13 However, Keloid disease is a heterogeneous lesion, also recognized as abnormal scar tissue that manifests with uncontrolled fibroproliferative growth beyond the boundaries of the original lesions that is recalcitrant to conventional scar therapy. KD also presents with excessive dysregulated collagen deposition, leading to a progressively invasive dermal disorder with an expansile and exophytic growth pattern as well as an aggressive clinical behaviour. 16 , 17

Keloid disease prevalence is higher in individuals with pigmented skin including, African, Hispanic and Asian ethnic skins but low in the Caucasian population. 18 , 19 , 20 , 21 , 22 , 23 In a study carried out in Nigeria, patients were reported to have a family history with known registered KD symptoms, 20 and additionally a high occurrence of KD has been reported in rural black South African populations. 23 , 24 It is not clear why keloid lesions develop in some individuals and not in others, and consensus on a definitive therapy is yet to be agreed upon.

Keloid therapy most often targets hyperactive fibroblast proliferation to halt increased amount of collagen deposition, 24 , 26 and to induce shrinkage of the keloid lesion. 27 , 28 , 29 The effect of therapeutic compounds against HTS, KD, and FKN dermal scars can be investigated using several models and assays including investigating keloid derived fibroblast proliferation, migration and invasion, in order to evaluate the effect of potential anti‐fibrotic candidates. 30 , 31 The high recurrence rate that often occurs post‐conventional therapies is a clinical challege and a major cause to intensify research for identifying better therapies with minimal side effects and reduced recurrence rates. Current treatment approaches include topical anti‐scarring application, surgical excision, cryotherapy, pressure dressing, laser therapy, intralesional corticosteroid injections, and antimetabolites. 32 , 33 , 34 , 35 , 36 , 37 , 38 , 39 , 40 , 41 , 42 , 43 , 44 , 45 , 46 , 47 , 48

The aim of this review is to briefly describe cutaneous wound healing processes and the participating immune cells, then provide a comprehensive overview of the immune and associated molecular targets involved in raised dermal scarring conditions.

2. CUTANEOUS WOUND HEALING

Cutaneous wound healing in human skin is a complex process that requires a well‐orchestrated series of events for the successful restoration of the structural barrier and partial return of the complete physiological, sensory and mechanical functions of the skin. 49 The process of wound healing can be divided into four distinct but overlapping steps: hemostasis, inflammation, proliferation, and remodelling. 50 , 51 , 52 Studies have demonstrated the indispensable role of the immune system in wound healing. Hence, in this section of the review, we will evaluate the contribution of various cells of the immune system and associated chemical effectors (cytokines) involved in and facilitating cutaneous wound healing (Figure 2).

FIGURE 2.

FIGURE 2

Phases of wound healing; inflammation, proliferation, and tissue remodelling. Key immunologic phenotype (immune cells) and key chemical messengers (cytokines and chemokines) for each phase of wound healing. The inflammation phase is characterized by neutrophil recruitment, as well as pro‐inflammatory cytokine release by macrophages. Proliferation also comprises macrophage recruitment by neutrophils and mast cells, which in turn initiate resident fibroblast proliferation. Later, anti‐inflammatory cascade by macrophages is evident. During the last stage of wound healing, re‐epithelialization continues, as wound granulation tissue forms, and collagen deposition increases, due to increased cell proliferation, while T‐cells recruit late‐stage (pro‐resolving) macrophages, as well as formation of cell–cell interaction with skin cells. TGF‐β, transforming growth factor beta; VEGF, vascular endothelial growth factor; CXCR1, C‐X‐C Motif Chemokine Receptor 1; IL6, interleukin 6; INF‐γ, interferon‐gamma; IGF, insulin‐like growth factor‐1; CD4, helper T cells; bFGF, basic fibroblast growth factor; EGF, epidermal growth factor; PDGF, platelet‐derived growth factor; TNF‐α, tumor necrosis factor alpha; MMP‐2, matrix metalloproteinase‐2; TIMP‐1, tissue inhibitor of metalloproteinases 1.

2.1. Immune cells involved in would healing

2.1.1. Macrophages

Macrophages are one of the most important cells of the innate immune system that play a critical role in wound healing. 53 , 54 , 55 , 56 Macrophages are derived from monocytes that are recruited from peripheral blood to the site of injury and are essential for initiating the healing cascade. 57 , 58 The phenotype of monocyte‐derived macrophages during wound healing is influenced by the local microenvironment at the site of injury. 59 , 60 , 61 During the inflammatory phase, macrophages play a key role in driving the inflammatory response that is characterized by clearing debris and secretion of pro‐inflammatory cytokines. 51 , 53 However, at a later stage, they contribute to wound repair and control of inflammation. 53 This plasticity of macrophages is crucial for the wound healing process.

The role of monocyte‐derived macrophages during wound healing has been elucidated using C‐C chemokine receptor type 2 (CCR2) deficient mice. 54 This study showed that CCR2 knock‐out mice had impaired wound healing at all times‐points post‐injury, had reduced frequency of Ly6Chi monocytes/macrophages, and reduced production of IL‐1β plus tumor necrosis factor (TNF)‐α. 54 Finally, this study showed that the adoptive transfer of wild‐type monocytes into CCR2−/− mice restored wound healing, 54 demonstrating the importance of macrophages in wound healing. The Ly6Chi cells have been shown to produce pro‐inflammatory cytokines, including interleukin (IL)‐1β, TNF‐α, IL‐6, 62 and are indispensable for the initiation of inflammation that is required for the transition to the reparative phase of healing. 53 In fact, the absence of initial inflammation has been shown to impede the wound‐healing cascade resulting in failure of wound healing. 53 In contrast, the Ly6Clo cells have been shown to appear at the later stages of wound healing, displaying anti‐inflammatory characteristics, as well as promoting tissue remodelling, fibrosis, and secretion of transforming growth factor (TGF)‐β and IL‐10. 63 , 64 , 65

2.1.2. Neutrophils

Neutrophils are amongst the first inflammatory cells to arrive at the site of injury and they play a key role in removing debris and preventing infection by phagocytosing and killing pathogens through the release of reactive oxygen species, proteases, antimicrobial peptides, and neutrophil extracellular traps (NETs). 66 , 67 The recruitment and activation of neutrophils is driven by the chemical messenger chemokine ligand CXCL‐8, also known as IL‐8, that is secreted by activated tissue‐resident macrophages and fibroblasts, 68 , 69 and other danger associated molecular patterns (DAMPS) such as hydrogen peroxide and leukotriene B4 that are released by necrotic cells in the wound bed. 70 , 71 CXCL8 binds to CXC receptor‐1 (CXCR‐1) and CXCR2 on the surface of neutrophils, leading to their recruitment to the site of inflammation. Once neutrophils arrive in the wound, they are able to secrete CXCL8, thus, amplifying neutrophil recruitment via a pro‐inflammatory feedback loop. 72

A study by Simpson and Ross showed that neutrophils are dispensable during wound healing in guinea pigs that were depleted of neutrophils after administration of anti‐neutrophil serum. 73 This study showed that there was no difference between neutropenic and control wounds in the total wound volume occupied by mononuclear leukocytes and fibrin using histological analysis. 73 Importantly, there was no difference in the rate of wound debridement, cellularity, and degree of connective tissue formation between the two groups of animals, demonstrating that neutrophils are not essential for wound healing. 73 However, neutrophils have been shown to play an active role in secreting cytokines, chemokines, and growth factors such as TNF‐α, IL‐6, IL‐1β, CXCL8, CXCL2, monocyte chemoattractant factor‐1 (MCP‐1), vascular endothelial growth factor (VEGF), laminin 5 β‐3, and urokinase‐type plasminogen activator that are crucial for the recruitment of macrophages and T‐cells to the site of injury. 72 , 74 , 75 These molecules are also essential for angiogenesis, the proliferation of keratinocytes and fibroblasts, adhesion of keratinocytes to the dermal layer, and tissue remodelling.

2.1.3. Mast cells

Mast cells (MCs) are specialized innate immune cells that are abundant in the dermis and play a varied role across the different stages of wound healing. 76 , 77 , 78 A study by Younan and colleagues, investigated the requirement of MCs in wound healing using MCs‐deficient mice (WWv mice) that had uniform full‐thickness wound surface micro‐deformations that were induced with suction combined with open‐pore polyurethane foam. 79 This study unequivocally demonstrated that MCs are required for wound tissue granulation, cell proliferation, blood vessel sprouting, and collagen deposition during the proliferation and remodelling stages of healing. 79 This was further corroborated by subsequent studies that showed MCs activation and histamine release are essential for cutaneous wound healing in mice. 80

The contribution of MCs in wound healing also involves their ability to secrete various key factors that are essential for coordinating wound healing. For instance, MCs secrete histamine and VEGF that mediate vascular permeability and vasodilation during injury. 76 , 79 Moreover, they also secrete TNF‐α, Macrophage Inflammatory Protein 2 (MIP‐2), and IL‐8 which are involved in the recruitment of neutrophils to the site of injury. 81 , 82 They also secrete proteases such as tryptase that induces vasodilation that further enhances the trafficking of neutrophils and other inflammatory mediators to the injury site. 83 , 84 Finally, MCs contribute to angiogenesis by secreting platelet‐derived growth factor (PDGF), chymase, angiopoietin 1, and fibroblast growth factor 2 (FGF‐2). 82 , 85

2.1.4. Innate lymphoid cells

The contribution of innate lymphoid cells (ILC) in immunity, inflammation, and homeostasis is well established in the literature. 86 , 87 However, a few studies have explored their role during cutaneous wound healing. Three subsets of ILC have been defined and designated into groups 1, 2, and 3 based on their transcriptional landscape and effector functions. 88 , 89 , 90 Interestingly, several studies have identified the presence of ILC2s in human and murine skin, and revealed that these cells are elevated after cutaneous inflammation. 91 , 92 , 93 Importantly, skin ILC2s have been shown to respond to IL‐25, IL‐33, and TSLP and express type‐2 associated cytokines IL‐5 and IL‐13. 93 A series of studies by Yin and colleagues demonstrated that IL‐33, an alarmin that promotes ILC2 differentiation facilitated wound healing in an excisional wound healing model 94 and Staphylococcus aureus incision model. 95 A study by Rak and colleagues investigated the direct role of IL‐33‐dependent ILC2 responses in cutaneous wound healing after full‐thickness dermal injury in mice. 96 This study found that IL‐33 and ILC2s are elevated following cutaneous wounding in mice and humans. 96 Interestingly, depletion of ILC2 using anti‐CD90.2 monoclonal antibody delayed wound closure and impaired re‐epithelialization, a key process during wound healing. 96

2.1.5. Regulatory T‐cells

T regulatory cells (Tregs) are abundant in the tissues such as the skin and play a key role in homeostasis and regulating inflammation. 97 , 98 A study by Nosbaum and colleagues investigated the role of FoxP3‐expressing Tregs in excisional wound healing using FoxP3 express knocked‐in human diphtheria toxin receptor (FoxP3‐DTR) mice that allowed for inducible ablation of Tregs after the administration of the diphtheria toxin. 99 Depletion of Tregs delayed the kinetics of wound healing and impaired wound re‐epithelialization early after wounding, however, no significant differences were observed late after wounding. 99 Moreover, this study showed that activated Tregs characterized by expression of CD25, cytotoxic T‐lymphocyte‐associated protein 4 (CTLA‐4), and inducible T‐cell co‐stimulator (ICOS) accumulated at the site of injury, and suppressed the production of interferon (IFN)‐γ and recruitment of pro‐inflammatory macrophages. 99 Finally, it was demonstrated that conditional deletion of the epidermal growth factor receptor (EGFR) on Tregs using Foxp3creEGFRfl/fl mice delayed wound healing, showing the requirement of EGFR‐mediated Tregs function during wound healing. 99 However, Tregs have also been shown previously to negatively affect wound healing in diabetic mice. 100

2.2. Cytokines involved in wound healing

2.2.1. Tumor necrosis factor‐alpha

Tumor necrosis factor‐alpha (TNF‐α) is a pleiotropic cytokine that is secreted by keratinocytes, macrophages, MCs, and T cells. The contribution of TNF‐α during wound healing is concentration dependent – elevated levels of TNF‐α lead to a decrease in granulation tissue while low levels of TNF‐α promote collagen deposition. 101 A study by Mori and colleagues showed that mice deficient in TNF‐Rp55 had accelerated wound healing due to enhanced angiogenesis, collagen accumulation, re‐epithelialization, and leukocyte infiltration, suggesting a negative role of TNF‐α in wound healing. 102

2.2.2. IL‐17

IL‐17 is a pro‐inflammatory cytokine that has been implicated in the pathogenesis of inflammatory diseases such as psoriasis and rheumatoid arthritis. 103 , 104 bleomycin and IL‐1β‐induced pulmonary fibrosis, 105 skin sclerosis, 106 and liver fibrosis. 107 IL‐17 was demonstrated to be essential for wound closure in IL‐17−/− mice after incisional wound injury. 108 IL‐17 mediates its effects by enhancing the skin's antimicrobial barrier and regulating epidermal regeneration and differentiation. 108 , 109

2.2.3. IL‐33

IL‐33 is an alarmin that plays a central role in wound healing by facilitating the differentiation of ILC2s. 94 , 96 Administration of IL‐33 accelerated healing of incisional wounds in mice and was associated with improved re‐epithelialization, collagen deposition, expression of genes associated with the extracellular matrix, and development of alternatively activated macrophage. 94

3. ABERRANT WOUND HEALING

Aberrant wound healing is a manifestation of immune‐related perturbations in molecular and cellular processes that guide normal wound healing. 110 , 111 , 112 Such disruptions may occur at any of the stages of wound healing as outlined above. A transition, delay, or acceleration of any of these processes may result in the formation of either chronic (non‐healing wounds) or fibrotic disorders such as HTS and KD or the scalp‐associated FKN. 111 In this section of the review, we will focus on the major immune initiators and chemical mediators (cytokines and chemokines) that result in impaired healing such as prolonged inflammation, dysregulated re‐epithelialization processes, excessive ECM deposition, as well as atypical ECM remodelling (Figure 3). 6 , 113 , 114 , 115

FIGURE 3.

FIGURE 3

Wound healing phenotypic differences between hypertrophic scars and keloids. The 3 phases of wound healing (inflammation, proliferation, and tissue remodelling), with their associated upregulated or downregulated functional markers. Inflammation in HTS is characterized by the early recruitment of CD4+ T cells at the site of injury, and their subsequent polarization to Th‐2 phenotype, with the production of cytokines such as TNF‐α and IL‐1. VEGF, PDGF, FGF, IGF, TGF‐β, and CCL2 are cytokines and chemokines upregulated during the proliferation phase that is characterized by the increased proportion and longevity of pro‐fibrotic M2 macrophages, as well as the persistence of CD4+ T cells thereby allowing for increased fibroblast proliferation, myofibroblast differentiation, and increased collagen deposition. Chemical messengers such as IL‐4, IL‐5, IL‐10, IL‐13, and MMP‐2 are produced during the late stages of wound healing (the matrix formation or tissue remodelling). Th‐2, Th‐2 lymphocytes; TGF‐β, transforming growth factor beta; VEGF, vascular endothelial growth factor; HIF‐1α, hypoxia‐inducible factor 1‐alpha; CCL2, C–C motif chemokine ligand 2; IL6, interleukin 6; INF‐γ, interferon‐gamma; IGF, insulin‐like growth factor‐1; CD4, helper T cells; FGF‐1, fibroblast growth factor 1; EGF, epidermal growth factor; PDGF, platelet‐derived growth factor; TNF‐α, tumor necrosis factor alpha; MMP‐2, matrix metalloproteinase‐2; TIMP‐2, tissue inhibitor of metalloproteinases 2.

3.1. Hypertrophic scars

Hypertrophic scars (HTS) are a result of aberrant wound healing processes and are characterized by raised stiff scar tissue accompanied by redness, pain, and itch. 6 They also present with a lack of skin appendages such as hair follicles and sebaceous glands, with a consequent loss of skin stem cells, which play an essential role in replenishing cells in the skin. 116 This type of scarring mostly results from burn injuries and can be debilitating to patients. 117 In this section of the review, we look closely at the cellular (immune) processes and resulting molecular culprits associated with impaired homeostasis (prolonged inflammation, dysregulated re‐epithelialization, and excessive ECM deposition) in HTS. Furthermore, possible immune‐derived molecular targets are suggested with the aim to prevent HTS formation.

3.1.1. Prolonged inflammation

Multiple studies have implicated prolonged inflammation as a key player in the development of hypertrophic scarring post‐wounding. 110 , 118 , 119 , 120 Herein platelets, neutrophils, macrophages, MCs, and CD4+ T lymphocytes have been shown to produce proinflammatory cytokines such as IL‐1, 114 , 121 IL‐6, 122 IL‐10, 115 and TNF‐α, 121 as well as growth factors such as TGF‐β1 115 , 121 , 123 , 124 and PDGF, 114 , 121 with concomitant CCL2 chemokine production. 122 Persistent production of such molecules often result in hypertrophic scarring during the tissue remodelling stage of skin wound healing. Therefore, anti‐scarring therapies could be aimed at ameliorating this pro‐inflammatory immune phenotype in HTS. 115

A longitudinal study by Tredget et al. 125 showed that a polarized Th2 response to injury leads to increased Th2 fibrogenic cytokines (IL‐4 and IL‐10) and growth factors (TGF‐β), which in turn results in the development of fibrogenesis in HTS. Several studies have also shown increases in Th2 cytokines such as IL‐2 and IL‐4, as well as regulatory cytokines IL‐10 and TGF‐β in fibrotic conditions such as HTS. 115 , 126 , 127 Therefore, inducing a polarized Th1 cell‐mediated response, which leads to the production of pro‐apoptotic cytokines seems a plausible phenomenon in the treatment of HTS. Furthermore, since HTS fibroblasts have been shown to have low collagenase activity, coupled with nitric oxide synthase (NOS) activity, 128 increased collagenase activity, and matrix remodelling could be achieved via NOS activation by the Th1 cytokines. However, care must be taken when manipulating such cell‐mediated immune responses as a prolonged Th1 cell response (inflammation) might prove futile.

3.1.2. Dysregulated re‐epithelialization

Major players in dysregulated epithelialization resulting in hypertrophic scarring are skin resident immune cells; Langerhans cells, which produce growth factors such as insulin‐like growth factor one (IGF‐1) and fibroblast growth factor 1 (FGF‐1) 56 responsible for fibroblast proliferation and activation, as well as monocyte‐derived macrophages recruited to the site of injury. Alterations in macrophage number and phenotype can disrupt re‐epithelialization process and could dictate the level of scar formation. 53 , 56 , 129 Depletion of macrophages restricted to the early stage of the repair response (inflammatory phase) significantly reduced the formation of vascularized granulation tissue, impaired epithelialization, and resulted in minimized scar formation. 53

CXC chemokine receptor 2 (CXCR2) also known as interleukin 8 receptor beta (IL8Rb) is an essential mediator of neutrophil chemotaxis via the release of numerous keratinocyte‐derived chemokine ligands. 130 Its knockdown in wound healing mouse models has been implicated with a lack of monocyte infiltration, thereby resulting in reduced keratinocyte migration and proliferation (markers of re‐epithelialization) during wound healing. Chemotactic perturbation during the epidermal proliferation stages of wound healing therefore has the potential as a therapeutic anti‐scarring agent. A study by Dovi et al. (2003) suggested that re‐epithelialization might be improved in wounds of neutrophil‐depleted mice. 131 The investigators performed a time course neutrophil depletion by rabbit anti‐mouse neutrophil‐serum injection into mice, followed by wounding. Wound histology and re‐epithelialization protocols showed that neutropenic mice exhibited accelerated wound healing (closure), even in diabetic mouse models. Therefore, regarding excessive healing conditions such as keloid and HTS, neutrophil enrichment studies may be beneficial, with the aim of slowing the rate of re‐epithelialization, thus making neutrophils a potential therapeutic target for such raised dermal scarring conditions. 119

Taken together, these studies highlight the possibility of manipulating early immune processes such as cells (neutrophils and macrophages), as well as their associated chemokines and/or receptors in limiting the excessive epidermal proliferation and migration that happens during HTS formation.

3.1.3. Excessive ECM deposition and remodelling

The secretion of cytokines such as IL‐1, 115 IL‐4, 115 , 125 , 127 and TNF‐α 115 as well as growth factors including VEGF, TGF‐β1, and FGF 56 by macrophages, CD4+ T cells, resident Langerhans and MCs have been implicated in excessive matrix deposition, with aberrant ECM remodelling. M2 macrophages secrete factors such as TGF‐β1 that induce the proliferation of fibroblasts and their differentiation into myofibroblasts. 132 Furthermore, the development of a Th2 response has been associated with fibrogenesis. 115 , 125 , 126 For instance, a study by Wang and colleagues showed that fibroblasts cultured with CD4+ T cells that were purified from peripheral blood mononuclear‐cells (PBMCs) from burn patients exhibited increased cell proliferation, collagen synthesis, and alpha‐smooth muscle actin (α‐SMA), as well as a significant up‐regulation of TGF‐β, which inhibits Th1 cytokine expression and finally contributes to HTS formation. 123 These studies also suggest the hypothesis that prolonging polarized Th1 cell‐mediated response, which in turn results in the synthesis of anti‐fibrotic processes such as increased collagenase activity and matrix remodelling as well as manipulating macrophage phenotypic characteristics (shift from M2 to M1) could be a potential therapeutic target for HTS.

Chemokines and their receptors may also be potential targets for hypertrophic scarring. Ishida et al. 56 showed that CX3CR1 mediates the direct recruitment of bone marrow‐derived monocytes/macrophages which release profibrotic and angiogenic mediators. In their study, a CX3CR1 knockout mouse model of excisional skin wound healing resulted in the amelioration of a pro‐fibrotic macrophage phenotype, leading to a decreased expression of αSMA and collagen deposition. This renders the chemokine receptor CX3CR1 a potential immune target for HTS.

IL‐1α and PDGF derived from Langerhans cells, macrophages, and platelets have been identified as some of the molecular targets in fibrotic conditions such as wound healing. Another study 114 showed low expression of IL‐1α and increased expression of PDGF in biopsies in both the epidermis and dermis of individuals who developed HTS after undergoing breast reduction surgery. 114

3.2. Keloid disease

Keloid disease (KD) is a complex fibroproliferative scarring disorder that even though it is non‐malignant, behaves in an aggressive, irreversible and progressive quasi‐neoplastic manner, and many studies have indicated that the immune system may play a role in keloid formation. 133 , 134 KD is characterized by an exaggerated exophytic growth and excessive deposition of collagen in areas of previous trauma or cutaneous injury. 27 , 135 KD is unique to humans since no animal model exists. 136 , 137 , 138 , 139 Keloid is thought to develop after aberrant wound healing due to alterations in the stages of normal wound healing as outlined in the sections above, and it is known to have a high recurrence rate upon resection. 25 Wound healing process after trauma or cutaneous injury could become keloidal, possibly due to a reduction in the apoptosis of fibroblasts or an imbalance between collagen production and degradation. 140 The development of keloids has been linked to the downregulation of apoptosis‐related genes including p53, 140 TGF‐β1/Mothers Against Decapentaplegic Homolog (Smad) signal transduction mediated by mitogen‐activated protein kinases (MAPKs), 141 the extracellular signal‐regulated protein kinase (ERK), c‐Jun N‐terminal kinase (JNK), and p38 pathways. 141 , 142 These might be considered specific targets of drug therapy for keloids. It is not clear why keloids develop in some individuals and not in others, and consensus on a definitive therapy is yet to be achieved.

The primary treatment target would lead to a decrease in collagen deposition and activation of fibroblasts. When keloid‐derived fibroblasts, which are the major cell type in KD, are compared with fibroblasts derived from normal skin or HTS, the keloid‐derived fibroblasts present several abnormal changes such as excessive extracellular matrix production and proliferation, altered apoptosis, growth factor response, and cytokine production. 140 Currently, there is no consensus on a definitive therapeutic target and therapy. Several studies have been carried out providing evidence of the effective use of corticosteroids including Dexamethasone, and Triamcinolone post‐surgical excision. 35 , 36 , 37 , 38 , 39 , 40 , 41 , 42 , 43 , 44 , 45 , 46 , 47 , 48 However, the problem of recurrence of keloids after excision is of primary concern and therefore, there is an urgent need for more research on alternative therapeutic options. There is also a need to identify potential biomarkers and targets that would aid in the diagnosis and treatment of dermal scars.

It is important to note that several study models have been investigated for functional testing of therapeutic anti‐scarring drug targets in keloids by employing key assays and techniques. 30 , 31 Our group has looked at multidimensional models of KD, 31 and are currently developing new models for functional testing of therapeutic anti‐scarring drug targets in keloids. 30 Going forward, understanding immune and associated molecular target candidate regulators would be significantly helpful. In addition, immune‐associated molecular targets are vital, as they may contribute either by regulating or are regulated in inflammatory molecular events involved in fibroblast activation leading to fibrosis. 143 These may consist of alterations in fibroblast and keratinocyte involvement in raised dermal scarring conditions, as well as alterations in growth factor expression. Moreover, it is vital to understand the role of bioenergetic changes that may be involved in regulating immunometabolism in skin fibrotic disorders.

In this section, we will look at Immune and associated immune targets involved in KD. Several immune cells and molecules that mediate their activity either directly or indirectly in KD have been reported (Figure 3). Here, we will discuss the various cellular as well as molecular targets associated with KD, which could be important to guide and inform future studies on possible immune‐diagnostic, −therapeutic, and ‐theranostic strategies against keloid tissue formation. Some important potential immune‐associated therapeutic targets in hypertrophic and keloid scars discussed in this review have been summarized in Figure 4. 144 , 145 , 146 , 147 , 148 , 149 , 150 , 151 , 152 , 153 , 154 , 155 , 156 , 157 , 158 , 159 , 160 , 161 , 162 , 163 , 164 , 165 , 166 , 167 , 168 , 169 , 170 , 171 , 172 , 173 , 174 , 175 Also, we will have a comprehensive look at the involvement of keloid‐associated lymphoid tissues on the pathophysiology of keloid tissue formation. 133

FIGURE 4.

FIGURE 4

Immune‐associated therapeutic targets in hypertrophic scars and keloids. Potential targets for raised dermal scarring therapeutics include Immune (KALT, neutrophils, macrophages, Langerhans cells, FDCs, T‐cells, B‐cells, PCs, MCs) and non‐immune (fibroblasts and keratinocytes) cells, and their associated matrices such as collagens, fibrin, fibronectin, elastin, HA, laminins, and tenascin. Several genes and molecules secreted by immune cells activate fibroblasts, myofibroblasts, keratinocytes, and enhanced by EMT for matrix production. Matrix production is mediated through several pathways that could be potential targets as therapeutic candidates. FDC, follicular dendritic cells; T‐cells, T lymphocytes; B‐cells, B lymphocytes; PCs, plasma cells; MCs, mast cells; HA, hyaluronidase; KALT, keloid associated lymphoid tissues; TGF‐β, transforming growth factor beta; CTGF, connective tissue growth factor; HIF‐1α, hypoxia‐inducible factor 1‐alpha; VEGF, vascular endothelial growth factor; HSP27, heat shock protein 27; α2β1, collagen‐binding Integrin α2β1; PAI‐1/2, plasminogen activator inhibitor‐1/2; CD138, cluster of differentiation 138 (syndecan‐1); SiRNAs, small interfering RNAs; p53, p53 gene; NEDD4, neuronal precursor cell‐expressed developmentally downregulated 4; CCL2, C–C motif chemokine ligand 2; IL6, interleukin 6; S100A, S100 calcium‐binding protein A1; IGF, insulin‐like growth factor‐1; CD3, T lymphocytes; CD4, helper T cells; CD8, cytotoxic T cells; CD45RO, memory T cells; CD20, B‐lymphocyte antigen; CD79a, B‐cell antigen receptor complex‐associated protein alpha chain and MB‐1 membrane glycoprotein; CD68, transmembrane glycoprotein; HLA‐DR, human leukocyte antigen – DR isotype; CD36, platelet glycoprotein 4, fatty acid translocase; ICAM‐1, intercellular adhesion molecule 1; CD31, platelet endothelial cell adhesion molecule; OX40L/CD134, tumor necrosis factor (ligand) superfamily, member 4; OCT4, octamer‐binding transcription factor 4; SOX2, sex determining region Y‐box 2; NANONG, homeobox protein NANOG; INF‐γ, interferon gamma; CX3CR1, CX3C chemokine receptor 1; IGF‐1, insulin‐like growth factor 1; FGF‐1, fibroblast growth factor 1; EGF, epidermal growth factor; PDGF, platelet‐derived growth factor; TNF‐α, tumor necrosis factor alpha; JAK, janus kinase; STAT, signal transducers and activators of transcription; SMAD‐3, Mothers against decapentaplegic homologue 3; MAPKs, mitogen‐activated protein kinases; ERK, extracellular signal‐regulated kinase; PI3K, phosphatidylinositol 3‐kinase.

3.2.1. Keloid‐associated lymphoid tissue (KALT)

Studies have investigated the role of immune cells in keloid tissue and have reported a range of immune markers. One of the key studies by Bagabir et al. showed that in intralesional and perilesional keloid tissue samples, there was a presence of distinctive lymphoid aggregates (LA) in the dermis, such as those observed in tertiary lymphoid tissues (TLT) 176 , 177 and mucosa‐associated lymphoid tissue (MALT), 178 which they termed keloid‐associated lymphoid tissue (KALT). 133 In addition, Bagabir et al. demonstrated that degranulated MCs and mature MCs (co‐expressing OX40L) were significantly increased in keloid lesional sites compared to the extra‐lesional, normal scar, and normal skin tissue. 133 Also, M2 macrophages a component of KALT, were found to be present with fibroblasts and activated T‐cells [T helper type 2 (Th2) and Th17] which further promote wound healing and fibrogenesis. 179 , 180 Grant et al. in a recent report showed that embryonic stem cell (ESC) markers were expressed in KALT including octamer‐binding transcription factor 4 (OCT4), SRY homology‐box 2 (SOX2), phospho‐signal transducer and activator of transcription 3 (pSTAT3), and homeobox protein North American Network Operators' Group (NANOG), and are located at the endothelium of the microvessels within KALT. 181 They may be responsible for the production of aberrant fibroblasts and myofibroblasts through a mesenchymal stem cell (MSC) intermediate in an endothelial‐to‐mesenchymal transition (Endo‐MT) process induced by cytokines such as TGF‐β1 thereby promoting keloid tissue formation. 181 , 182 , 183

The presence of KALT in organized TLT structures in KD is indicative of a chronic inflammatory milieu which may be important for KD fibrogenesis and may be exploited as a target for immunotherapy. Since there is a milieu of immune cells within KALT, it provides the opportunity for further research to tease out the various cells and networks to identify the possible options for immune‐targeted therapy.

3.2.2. Mast cells

Mast cells have been shown to be activated and increased in the dermis of the keloid lesional site. 133 , 184 A study by Bagabir et al. demonstrated a significant increase in the frequency of MCs degranulation compared to non‐degranulated MCs, and mature MCs in KD than in control normal skin tissue, indicating that the number and degranulation status of MCs are important factors in KD development. 133 Moreover, they found a significantly increased frequency of c‐kit+ MCs in the KD dermis and mature β‐tryptase MCs co‐expressing OX40L in intralesional and perilesional KD sites compared to normal skin control samples. 133 Their findings are in line with an earlier study that performed a comparative proteomic analysis between normal skin and keloid scar and found that MCs β‐tryptase protein was significantly upregulated in KD compared to normal skin. 185 Other studies have reported that MCs enhance scar formation and may mediate the transition from scarless to fibrotic healing. 186

There is evidence that MCs promote ECM production in in vitro co‐culture of KD fibroblasts and MCs, 187 which is reported to occur through their direct and immediate cell‐to‐cell contact with fibroblasts a phenomenon termed “cell talk.” 187 , 188 Cell talk has been shown to facilitate expression of VEGF and hypoxia‐inducible factor‐1α (HIF‐1α) which promote aberrant fibrogenesis in KD. 187 , 188 The involvement of the extracellular signal‐regulated kinase 1 and 2 (ERK1/2) cascade and phosphatidylinositol‐3‐kinase (PI3K)/Akt signalling pathways have been reported to promote the accumulation of HIF‐1α in co‐culture of keloid fibroblasts and MCs. 187 There are reports associating hypoxia environment with keloid fibrogenesis 187 , 188 , 189 , 190 which could be beneficial for the survival of keloid lesions. Keloid fibroblasts have been shown to exhibit the Warburg effect which is the preference of glycolysis and dependence on glucose for their metabolism, 191 thought to be enhanced by glycolytic enzymes activated by HIF‐1α. 192 , 193 , 194

3.2.3. Macrophages

Macrophages have been implicated in many fibrotic disorders, and are thought to be a contributor to keloid fibrogenesis. 188 , 195 Bagabir et al. 133 showed that the expression of major histocompatibility complex (MHC) II in both M1 and M2 macrophages was altered in KD compared to normal tissue, 133 which confirms findings of previous studies that reported low expression of MHC II by M2 and tumor associated macrophages (TAM) in patients with atherosclerosis a fibrotic disease as well as in cancers such as pancreatic cancer. 196 , 197 , 198 In addition, Bagabir et al. reported a significantly higher frequency of M2 macrophages at intralesional and perilesional sites in keloid tissue compared to normal control skin. 133 This finding has been supported by other studies that have reported the presence of macrophages in KD. 195 , 199 , 200 Li et al. in their study investigating the status of M1 and M2 macrophages in KD, consistently demonstrated that M2 infiltration outnumbered M1 macrophages in the dermis of keloid compared with normal control tissues, a finding similar to Bagabir's. 133 , 200 Several human leucocyte antigen (HLA) could act as potential biomarkers for KD. 201 , 202 , 203 High frequency of HLA‐DRB1*15 phenotype has been reported in Caucasian KD patients compared to controls. 201 Lu et al. showed a positive association between HLA‐DQA1 and ‐DQB1 genotypes and the development of KD in a Chinese cohort. 202 Rossi and colleagues reported an association between HLA class 1 antigen (HLA‐B21) and class 2 antigens (HLA‐DR5 and HLA‐DQw3), with keloids in their Italian cohort. 203

At the cellular level in an in vitro co‐culture experiment, there is evidence that monocytes differentiate into M2 macrophages when in an early phase of wound regeneration and repair in the presence of either keloid or normal tissue, indicating that this would occur irrespective of the individual being prone to normal or keloid scar formation. 204 M2 macrophages play a critical part in fibrogenesis by secreting growth factors that play a vital role in KD pathogenesis, as elevated levels of TGF‐β, VEGF, connective tissue growth factor (CTGF), and PDGF are reportedly involved in stimulating collagen secretion, growth, and promote keloid fibroblast proliferation as well as keloid tissue expansion. 205 , 206 , 207

3.2.4. T lymphocytes mediated cytokine responses

T helper type 1 (Th1) immune responses linked with pro‐inflammation have been shown to be stimulated in normal wound healing, while Th2 responses have been shown to be stimulated in keloid formation. 179 , 180 IFN‐α, IFN‐γ, and TNF‐β which promote Th1 differentiation and with proinflammatory capacity have been shown to inhibit fibroblast proliferation in keloid cells, while IL‐4, IL‐6, IL‐10, and IL‐13 which promote Th2 differentiation have been linked to fibrogenesis in keloids. 186 , 208 , 209 , 210 There is evidence suggesting that the type of immune response in addition to the severity of inflammation is a vital factor that predisposes individuals to hypertrophic and keloid scarring. 211

In a study investigating various cytokines in PBMCs of black KD individuals by McCauley et al., IFN‐α, INF‐γ, and TNF‐β were reported to be highly suppressed in keloid patients compared to their normal control counterparts. 210 On the contrary to the expected reduction of Th1 cytokine production by keloid individuals, McCauley et al. showed that expression of IFN‐β, and TNF‐α production was markedly higher in PBMCs of KD individuals. 210 There was increased expression of IL‐6 a Th2 cytokine in PBMCs from individuals with KD compared to control PBMCs from individuals without keloids. 210 IL‐6 and its mediated response through the Janus kinase (JAK)/Stat3 transcription factor signalling pathway, 212 , 213 , 214 have been shown to promote keloid fibroblast proliferation and ECM synthesis. 208

A study by da Silva et al. reported significantly lower mRNA expressions of IFN‐γ, IFN‐γR1, and IL‐10 in keloid tissues than in normal control tissue, but no significant difference in TNF‐α expression. 215 Interestingly, they found a negative correlation between collagen type III and expression of IFN‐γ, IFN‐γR1, and IL‐10 in keloid biopsies. 215 The predominant expression of collagen type III in keloids compared to the controls, is a reflection of keloid as an immature lesion. 215 Further, increased IL‐4/IL‐13 signalling and responses have been reported in keloid lesions, with IL‐13 significantly increased in lesional and non‐lesional keloid tissue than in normal control tissue. 216 In addition, a study by Oriente et al. demonstrated that post IL‐13 stimulation of keloid fibroblasts, there was high expression of the procollagen 1α1 gene compared to normal skin fibroblasts. 217 Also, the ability of IL‐13 to induce gene expression for procollagen 3α1 in keloid fibroblasts was unique compared to IL‐4 or TGF‐β. 217

Zhang et al. in their study investigating alteration in stem cell niche in keloid, demonstrated a robust increase in IL‐6 and IL‐17 expression in keloid stem cells. 183 Employing sub‐clonal assay, flow cytometry, and multipotent differentiation analyses, they reported that keloid lesions have a new population of stem cells, named keloid‐derived precursor cells, which exhibit clonogenicity, self‐renewal, distinct embryonic and MSC surface markers, and multipotent differentiation. 183 Their findings suggested that the altered biological functions in KD are highly regulated by the inflammatory niche mediated by an autocrine/paracrine cytokine IL‐17/IL‐6 axis. 183

3.2.5. Memory T cells

There is evidence linking memory T‐cells to the regulation of inflammation in KD. Chen et al. characterized memory T‐cells expressing CD45RO in primary cutaneous cells in the tissue and PBMCs from patients with KD. 218 They found an increased frequency of CD8+CD45RO+ memory T‐cells in KD tissue compared to normal tissue. 218 Similarly, a study by Santucci et al. identified CD45RO+ memory T cells in keloid tissues from Caucasian patients compared to individuals with HTS in their Italian cohort. 219 Also, Chen et al. found no significant difference in CCR7 expression between CD8+CD45RO+ memory T cells derived from healthy and KD tissue. 218 This was further corroborated by the observation that the CD45RA‐CD45RO+ memory T cells while poorly expressing CCR7 were significantly expressing CD103 compared to control normal cells, indicating that these memory T cells were functionally not recirculating. 218 These findings by Chen et al. show abnormalities of CD45RO+ memory T cells in KD and indicate that a disruptive T‐cell response contributes to KD progression. 218

3.2.6. Regulatory T cells

Chen et al. found a reduced frequency of CD4+CD25+FOXP3+ regulatory T‐cells (Tregs) in PBMCs of patience with multiple keloids. In cells derived from KD tissue expressing CD4+FoxP3+CD45RO+ memory T‐cells, there was reduced expression of CD25 and cytotoxic T‐lymphocyte associated antigen 4 (CTLA‐4), and IL‐10 production. 218 This indicates a lack of suppression of the inflammatory machinery of memory T‐cells in KD. Tregs can suppress effector CD4+ T‐cells and modulate their responses. It has been shown by Murao et al. that the Tregs:CD4+ T‐cells ratio is lower in KD, which may be beneficial to keloid lesions. 220

3.2.7. Keratinocytes

Conventionally, keloids are considered a raised dermal scarring disorder involving mainly abnormal fibroblasts, however, recent studies have demonstrated the involvement of potentially abnormal keratinocytes and other cell types as well. 221 , 222 Keratinocytes are the main cell type of the epidermis which are tightly held together by cell junctions and can trigger immune response via secretion of various chemokines, cytokines, cytokine receptors, and various antimicrobial peptides. 223 Keratinocytes have been shown to enhance proliferation and reduce apoptosis of underlying fibroblasts. 224 Bagabir et al. suggested that epidermal keratinocytes communicate with fibroblasts, and this intercommunication was shown to play a significant role in keloid formation. 133 Gene expression profile of keloid keratinocytes have reported expression of numerous genes involved in epithelial‐mesenchymal transition (EMT), suggesting the involvement of EMT in adhesion abnormalities that may be beneficial to KD. 225 This has been supported by other studies that have shown that keloid keratinocytes exhibit an EMT‐like state similar to activated keratinocytes during wound healing. 158 Interestingly, TGF‐β1 has been shown to decrease expression of EMT‐related genes including IL‐6, vimentin and cadherin‐11, and keloid keratinocyte migration through canonical and non‐canonical signalling pathways. 158 , 226 Hence, TGF‐β1 can be explored as a potential therapy for KD.

3.3. Folliculitis keloidalis nuchae

Folliculitis keloidalis nuchae (FKN), also known as acne keloidalis nuchae, is a chronic type of folliculitis that typically involves the back of the neck and is commonly seen in young men of African descent. The lesion is neither a keloid nor is it associated with acne vulgaris. It is often pustular in its early stages and can later coalesce to form scarred keloidal plaques. 227 Advanced lesions may present purulent discharges and abscesses when secondarily infected. It is characterized histopathologically by deep scarring folliculitis, naked hair shafts, upper dermal inflammation, and occasional plaque or pustulopapular plaque lesions in the peri‐inflammation area. Aggregates of red blood cells, bacterial colonies, and neutrophils can be seen in the superficial epithelial layers. There can also be completely/partially ruptured hair follicles in the mid dermis. Partially ruptured hair follicles may contain basophilic debris, fragmented hair shafts, neutrophils, abundant keratins, and bacterial colonies. 227

The perifollicular region may be infiltrated by lymphocytes, histiocytes, plasma cells, and eosinophils. Completely ruptured hair follicles may show infiltration of lymphocytes, histiocytes, and plasma cells, as well as a neutrophilic aggregation that replaces the original follicular structures. 227 An important aetiological factor is suspected to be inflammation/infection. Inflammation in FKN could potentially be stimulated by chronic mechanical irritation/trauma and friction to the occipital, nuchal and occipital area during frequent haircuts, when wearing a helmet or from shirt collars. 227 Other speculated etiological factors that have been implicated in some patients include the use of anti‐epileptic agents, immunosuppressive agents, metabolic syndrome, and chronic infection. 227 , 228 , 229 , 230 , 231 , 232 A few other suggested inciting agents are androgens, genetics, and in‐growing hairs. 10 As in pseudo folliculitis barbae, shaving‐induced trauma may result in abnormal keratin expression in FKN. 233 Chronic low‐grade infection (e.g. Malassezia or S. Aureus), use of medication (e.g. cyclosporine, carbamazepine, and diphenylhydantoin), and autoimmunity have been implicated in a few individuals. 228 , 234

Although inflammation is canonically central to the pathogenesis of FKN, it is unclear whether it is causal or consequent. 10 Hence, FKN is described as primary cicatricial alopecia in the absence of overwhelming evidence of causative agents. Previous histological studies have alluded to possible pathogenesis from inflammation of follicular units, leading to the attraction of autogenous antigenic targets by host inflammatory cells and the destruction of follicular or pilosebaceous units. 235 , 236 One of the questions that still needs to be resolved is whether the sebaceous gland is a direct target of the inflammatory process or gets destroyed in the process of inflammation. 237 Although the reasons for nuchal localization of FKN are unclear, abundant MCs and dermal papillary dilatation in this region have been speculated as responsible. 238 Further, obesity and friction in the scalp skin fold in this region may be responsible. In fact, a study has alluded to FKN being a cutaneous feature of a metabolic syndrome, albeit with very little proof. 230

At the molecular level, very few studies have specifically investigated the direct immune targets of FKN. A recent case report detected the expression of IL‐6 in FKN tissue using immunohistochemistry and confocal microscopy. 239 Even though, the keloidal collagen that is typically found in keloids is absent in FKN, 240 the authors argue that molecules involved in the pathogenesis of keloids (e.g. CKLF‐1, IL‐6, IL‐24, IL‐18, TGF‐β1, STAT3, JAK1, ELK1, and RAF1), 208 , 241 may be targeted for FKN. 239

4. CONCLUDING REMARKS

We have highlighted in this review, various immunological targets identified in raised dermal scars with a focus on hypertrophic, keloid, and FKN scars. Key pathways/interactions that are perturbed during dermal fibrogenesis have been discussed in detail and essential pathological deviations that result in raised dermal fibrogenic phenotypes were also discussed. The key interplay between traditional immune cells and non‐immune cell capable of producing chemical mediators of aberrant inflammation were highlighted. Understanding the immune regulators and role of immune cells, non‐immune cells, chemical modulators, genetic alteration, and “fibroblast‐to‐myofibroblast” switch in the excessive deposition of extracellular matrix molecules that always characterize raised dermal scars is key for the development of effective targeted therapies, that are highly desired for the management of raised dermal scars.

AUTHOR CONTRIBUTIONS

Elvis Banboye Kidzeru, Jyoti Rajan Sharma, Maribanyana Lebeko, Lucia Nkengazong, enry Ademola Adeola, Hlumani Ndlovu, Nonhlanhla Khumalo, and Ardeshir Bayat participated in drafting, editing, discussion, and revising the paper, and have all read and approved the final manuscript for submission.

CONFLICT OF INTEREST

None of the authors have a conflict of interest to declare.

ACKNOWLEDGEMENTS

NPK and Hair and Skin Research Lab are funded by the National Research Foundation and AB is funded by the South African Medical Research Council.

Elvis Banboye Kidzeru was supported by the Pierre Fabre‐Hair and Skin Research Lab collaboration grant, and the University of Cape Town Faculty of Health Sciences Postdoctoral Fellowship.

Jyoti Rajan Sharma was supported by the Pierre Fabre‐Hair and Skin Research Lab collaboration grant.

Henry Ademola Adeola thanks the SAMRC for mid‐career scientist and Self‐initiated research grants; and the NRF for incentive and research development grants for rated researchers.

Maribanyana Lebeko received the NRF innovative and Services SETA Fellowships.

Kidzeru EB, Lebeko M, Sharma JR, et al. Immune cells and associated molecular markers in dermal fibrosis with focus on raised cutaneous scars. Exp Dermatol. 2023;32:570‐587. doi: 10.1111/exd.14734

DATA AVAILABILITY STATEMENT

Data sharing not applicable to this article as no datasets were generated or analysed during the current study.

REFERENCES

  • 1. Schaefer L. Decoding fibrosis: mechanisms and translational aspects. Matrix Biol. 2018;68‐69:1‐7. doi: 10.1016/j.matbio.2018.04.009 [DOI] [PubMed] [Google Scholar]
  • 2. Sebastiani G, Gkouvatsos K, Pantopoulos K. Chronic hepatitis C and liver fibrosis. World J Gastroenterol. 2014;20(32):11033‐11053. doi: 10.3748/wjg.v20.i32.11033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Dong J, Ma Q. Myofibroblasts and lung fibrosis induced by carbon nanotube exposure. Part Fibre Toxicol. 2016;13(1):1‐22. doi: 10.1186/s12989-016-0172-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Gu X, Mallipattu SK, Guo Y, et al. The loss of Krüppel‐like factor 15 in Foxd1+ stromal cells exacerbates kidney fibrosis. J Autism Dev Disord. 2017;47(3):549‐562. doi: 10.1097/CCM.0b013e31823da96d.Hydrogen [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Wynn TA. Cellular and molecular mechanisms of fibrosisCellular and molecular mechanisms of fibrosis. J Pathol. 2009;214(2):199‐210. doi: 10.1002/path.2277.Cellular [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Zhu Z, Ding J, Shankowsky HA, Tredget EE. The molecular mechanism of hypertrophic scar. J Cell Commun Signal. 2013;7(4):239‐252. doi: 10.1007/s12079-013-0195-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Mokos ZB, Jović A, Grgurević L, et al. Current therapeutic approach to hypertrophic scars. Front Med. 2017;4(83):1‐11. doi: 10.3389/fmed.2017.00083 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Sidgwick GP, Bayat A. Extracellular matrix molecules implicated in hypertrophic and keloid scarring. J Eur Acad Dermatology Venereol. 2012;26:141‐152. doi: 10.1111/j.1468-3083.2011.04200.x [DOI] [PubMed] [Google Scholar]
  • 9. Chanpimol S, Seamon B, Hernandez H, Harris‐love M, Blackman MR. Keloids: the paradigm of skin fibrosis – pathomechanisms and treatment. Matrix Biol. 2017;51:37‐46. doi: 10.1016/j.matbio.2016.01.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Ogunbiyi A. Acne keloidalis nuchae: prevalence, impact, and management challenges. Clin Cosmet Investig Dermatol. 2016;9:483‐489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Umar S, David CV, Castillo JR, Queller J, Sandhu S. Innovative surgical approaches and selection criteria of large Acne Keloidalis Nuchae lesions. Plast Reconstr Surg ‐ Glob Open. 2019;7(5):e2215. doi: 10.1097/gox.0000000000002215 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Abergel RP, Pizzurro D, Meeker CA, et al. Biochemical composition of the connective tissue in keloids and analysis of collagen metabolism in keloid fibroblast cultures. J Invest Dermatol. 1985;84(5):384‐390. Accessed November 16, 2018. http://www.ncbi.nlm.nih.gov/pubmed/3998489 [DOI] [PubMed] [Google Scholar]
  • 13. Alaish SM, Yager DR, Diegelmann RF, Cohen IK. Hyaluronic acid metabolism in keloid fibroblasts. J Pediatr Surg. 1995;30(7):949‐952. Accessed November 16, 2018. http://www.ncbi.nlm.nih.gov/pubmed/7472951 [DOI] [PubMed] [Google Scholar]
  • 14. Mutsaers SE, Bishop JE, McGrouther G, Laurent GJ. Mechanisms of tissue repair: from wound healing to fibrosis. Int J Biochem Cell Biol. 1997;29(1):5‐17. doi: 10.1016/S1357-2725(96)00115-X [DOI] [PubMed] [Google Scholar]
  • 15. Ghatak S, Hascall VC, Rodriguez RM, Markwald RR, Misra S. Inflammation, Wound healing, and fibrosis. Wound Healing: Stem Cells Repair and Restorations, Basic and Clinical Aspects. Wiley Blackwell; 2018:195‐209. doi: 10.1002/9781119282518.ch15 [DOI] [Google Scholar]
  • 16. Berman B, Bieley HC. Keloids. J Am Acad Dermatol. 1995;33(1):117‐123. Accessed November 16, 2018. http://www.ncbi.nlm.nih.gov/pubmed/7601928 [DOI] [PubMed] [Google Scholar]
  • 17. Rockwell WB, Cohen IK, Ehrlich HP. Keloids and hypertrophic scars: a comprehensive review. Plast Reconstr Surg. 1989;84(5):827‐837. Accessed November 16, 2018. http://www.ncbi.nlm.nih.gov/pubmed/2682703 [DOI] [PubMed] [Google Scholar]
  • 18. Shaheen A, Khaddam J, Kesh F. Risk factors of keloids in Syrians. BMC Dermatol. 2016;16(1):13. doi: 10.1186/s12895-016-0050-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Sun L‐M, Wang K‐H, Lee Y‐CG. Keloid incidence in Asian people and its comorbidity with other fibrosis‐related diseases: a nationwide population‐based study. Arch Dermatol Res. 2014;306(9):803‐808. doi: 10.1007/s00403-014-1491-5 [DOI] [PubMed] [Google Scholar]
  • 20. Olaitan P, Olabanji J, Oladele A, Oseni G. Symptomatology of keloids In Africans. Vol 5. Wissenschaftsrat; 2013. Accessed March 9, 2019. https://www.ajol.info/index.php/sljbr/article/view/98001
  • 21. Bayat A, Arscott G, Ollier WER, Mc Grouther DA, Ferguson MWJ. Keloid disease: clinical relevance of single versus multiple site scars. Br J Plast Surg. 2005;58(1):28‐37. doi: 10.1016/j.bjps.2004.04.024 [DOI] [PubMed] [Google Scholar]
  • 22. Kouotou EA, Nansseu JR, Omona Guissana E, et al. Epidemiology and clinical features of keloids in black Africans: a nested case–control study from Yaoundé Cameroon. Int J Dermatol. 2019;58(10):1135‐1140. doi: 10.1111/ijd.14610 [DOI] [PubMed] [Google Scholar]
  • 23. Louw L. Keloids in rural black south Africans. Part 1: general overview and essential fatty acid hypotheses for keloid formation and prevention. PLEFA. 2000;63(5):237‐245. doi: 10.1054/plef.2000.0207 [DOI] [PubMed] [Google Scholar]
  • 24. Louw L. Keloids in rural black south Africans. Part 3: a lipid model for the prevention and treatment of keloid formations. PLEFA. 2000;63(5):255‐262. doi: 10.1054/plef.2000.0209 [DOI] [PubMed] [Google Scholar]
  • 25. Ashcroft KJ, Syed F, Bayat A. Site‐specific keloid fibroblasts alter the behaviour of normal skin and normal scar fibroblasts through paracrine signalling. Tsilibary EC. PLoS One. 2013;8(12):e75600. doi: 10.1371/journal.pone.0075600 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Jumper N, Hodgkinson T, Paus R, Bayat A. Site‐specific gene expression profiling as a novel strategy for unravelling keloid disease. Pathobiology. 2017;12:0172955. doi: 10.1371/journal.pone.0172955 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Syed F, Bagabir RA, Paus R, Bayat A. Ex vivo evaluation of antifibrotic compounds in skin scarring: EGCG and silencing of PAI‐1 independently inhibit growth and induce keloid shrinkage. Lab Invest. 2013;93(8):946‐960. doi: 10.1038/labinvest.2013.82 [DOI] [PubMed] [Google Scholar]
  • 28. Srivastava S, Patil A, Prakash C, Kumari H. Comparison of Intralesional triamcinolone Acetonide, 5‐fluorouracil, and their combination in treatment of keloids. World J Plast Surg. 2018;7(2):212‐219. [PMC free article] [PubMed] [Google Scholar]
  • 29. Khalid FA, Mehrose MY, Saleem M, et al. Comparison of efficacy and safety of intralesional triamcinolone and combination of triamcinolone with 5‐fluorouracil in the treatment of keloids and hypertrophic scars: randomised control trial. Burns. 2018;45:1‐7. doi: 10.1016/j.burns.2018.08.011 [DOI] [PubMed] [Google Scholar]
  • 30. Sharma JR, Lebeko M, Kidzeru EB, Khumalo NP, Bayat A. In vitro and ex vivo models for functional testing of therapeutic anti‐scarring drug targets in keloids. Adv Wound Care. 2019;8(12):655‐670. doi: 10.1089/wound.2019.1040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Lebeko M, Khumalo NP, Bayat A. Multi‐dimensional models for functional testing of keloid scars: In silico, in vitro, organoid, organotypic, ex‐vivo organ culture and in vivo models. Wound Repair Regen. 2019;27(4):298‐308. doi: 10.1111/wrr.12705 [DOI] [PubMed] [Google Scholar]
  • 32. Aslam F, Khalid U, Saleem M, et al. The efficacy of excision followed by intralesional 5‐fluorouracil and triamcinolone acetonide versus excision followed by radiotherapy in the treatment of ear keloids: a randomized control trial. Burns. 2018;44(6):1489‐1495. doi: 10.1016/j.burns.2018.02.017 [DOI] [PubMed] [Google Scholar]
  • 33. Barara M, Mendiratta V, Chander R. Cryotherapy in treatment of keloids: evaluation of factors affecting treatment outcome. J Cutan Aesthet Surg. 2012;5(3):185‐189. doi: 10.4103/0974-2077.101376 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Bran GM, Brom J, Hörmann K, Stuck BA. Auricular keloids: combined therapy with a new pressure device. Arch Facial Plast Surg. 2012;14(1):20‐26. doi: 10.1001/archfacial.2011.57 [DOI] [PubMed] [Google Scholar]
  • 35. Syed F, Bayat A. Superior effect of combination vs. single steroid therapy in keloid disease: a comparative in vitro analysis of glucocorticoids. Wound Repair Regen. 2013;21(1):88‐102. doi: 10.1111/j.1524-475X.2012.00862.x [DOI] [PubMed] [Google Scholar]
  • 36. Li J, Fu R, Li L, et al. Co‐delivery of dexamethasone and green tea polyphenols using electrospun ultrafine fibers for effective treatment of keloid. Pharm Res. 2014;31(7):1632‐1643. doi: 10.1007/s11095-013-1266-2 [DOI] [PubMed] [Google Scholar]
  • 37. Rojewska A, Karewicz A, Baster M, et al. Dexamethasone‐containing bioactive dressing for possible application in post‐operative keloid therapy. Cellul. 2019;26(3):1895‐1908. doi: 10.1007/s10570-018-2182-8 [DOI] [Google Scholar]
  • 38. Saki N, Mokhtari R, Nozari F. Comparing the efficacy of intralesional triamcinolone acetonide with verapamil in treatment of keloids: a randomized controlled trial. Dermatol Pract Concept. 2019;9(1):4‐9. doi: 10.5826/dpc.0901a02 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Abedini R, Sasani P, Mahmoudi HR, Nasimi M, Teymourpour A, Shadlou Z. Comparison of intralesional verapamil versus intralesional corticosteroids in treatment of keloids and hypertrophic scars: a randomized controlled trial. Burns. 2018;44(6):1482‐1488. doi: 10.1016/j.burns.2018.05.005 [DOI] [PubMed] [Google Scholar]
  • 40. Tel‐Aviv Sourasky Medical Center . Comparison of the Efficacy of Different Steroids in the Treatment of Abnormal Scars (Keloids, Hypertrophic Scars) ‐ Full Text View ‐ ClinicalTrials.gov. NIH ClinicalTrials.gov. Accessed June 9, 2022. https://clinicaltrials.gov/ct2/show/NCT04593706
  • 41. Wang R, Danielsen PL, Ågren MS, et al. Corticosteroid injection alone or combined with surgical excision of keloids versus other therapies including Ionising radiotherapy: a systematic review and meta‐analysis of randomised controlled trials. Eur Burn J. 2021;2(2):41‐54. doi: 10.3390/ebj2020004 [DOI] [Google Scholar]
  • 42. Wu WS, Wang FS, Yang KD, Huang CC, Kuo YR. Dexamethasone induction of keloid regression through effective suppression of VEGF expression and keloid fibroblast proliferation. J Invest Dermatol. 2006;126(6):1264‐1271. doi: 10.1038/sj.jid.5700274 [DOI] [PubMed] [Google Scholar]
  • 43. Xu S, Li D, Teng J, Zhang Z, Chen D, Bao W. Effects of steroids, interferon‐2B, or interluekin 1B on apoptosis of fibroblasts from keloid, hypertrophic scars, and normal skin and related signal pathway. Eur J Plast Surg. 2007;30(4):159‐167. doi: 10.1007/s00238-007-0165-3 [DOI] [Google Scholar]
  • 44. Bagabir R, Syed F, Paus R, Bayat A. Long‐term organ culture of keloid disease tissue. Exp Dermatol. 2012;21(5):376‐381. doi: 10.1111/j.1600-0625.2012.01476.x [DOI] [PubMed] [Google Scholar]
  • 45. Ojeh N, Bharatha A, Gaur U, Forde AL. Keloids: current and emerging therapies. Scars, Burn Heal. 2020;6:205951312094049. doi: 10.1177/2059513120940499 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Abdulhadi J, Ali A. Management of Keloid Scars: surgical versus medical therapy. J Dermatology Res Ther. 2018;4(2):1‐5. doi: 10.23937/2469-5750/1510059 [DOI] [Google Scholar]
  • 47. Wong TS, Li JZH, Chen S, Chan JYW, Gao W. The efficacy of triamcinolone acetonide in keloid treatment: a systematic review and meta‐analysis. Front Med. 2016;3(71):1‐8. doi: 10.3389/fmed.2016.00071 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Salzillo R. Triamcinolone acetonide intralesional injection for the treatment of keloid scars: patient selection and perspectives. Clin Cosmet Investig Dermatol. 2018;11:387‐396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Schultz GS, Davidson JM, Kirsner RS, Bornstein P, Herman IM. Dynamic reciprocity in the wound microenvironment. Wound Repair Regen. 2011;19(2):134‐148. doi: 10.1111/j.1524-475X.2011.00673.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature. 2008;453(7193):314‐321. doi: 10.1038/nature07039 [DOI] [PubMed] [Google Scholar]
  • 51. Boniakowski AE, Kimball AS, Jacobs BN, Kunkel SL, Gallagher KA. Macrophage‐mediated inflammation in Normal and diabetic wound healing. J Immunol. 2017;199(1):17‐24. doi: 10.4049/jimmunol.1700223 [DOI] [PubMed] [Google Scholar]
  • 52. Macleod AS, Mansbridge JN. The innate immune system in acute and chronic wounds. Adv Wound Care. 2014;5(2):65‐78. doi: 10.1089/wound.2014.0608 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Lucas T, Waisman A, Ranjan R, et al. Differential roles of macrophages in diverse phases of skin repair. J Immunol. 2010;184(7):3964‐3977. doi: 10.4049/jimmunol.0903356 [DOI] [PubMed] [Google Scholar]
  • 54. Boniakowski AE, Kimball AS, Joshi A, et al. Murine macrophage chemokine receptor CCR2 plays a crucial role in macrophage recruitment and regulated inflammation in wound healing. Eur J Immunol. 2018;48(9):1445‐1455. doi: 10.1002/eji.201747400 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Clover AJP, Kumar AHS, Caplice NM. Deficiency of CX3CR1 delays burn wound healing and is associated with reduced myeloid cell recruitment and decreased sub‐dermal angiogenesis. Burns. 2011;37(8):1386‐1393. doi: 10.1016/j.burns.2011.08.001 [DOI] [PubMed] [Google Scholar]
  • 56. Ishida Y, Gao J‐L, Murphy PM. Chemokine receptor CX3CR1 mediates skin wound healing by promoting macrophage and fibroblast accumulation and function. J Immunol. 2008;180(1):569‐579. doi: 10.4049/jimmunol.180.1.569 [DOI] [PubMed] [Google Scholar]
  • 57. Sica A, Erreni M, Allavena P, Porta C. Macrophage polarization in pathology. Cell Mol Life Sci. 2015;72(21):4111‐4126. doi: 10.1007/s00018-015-1995-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Martin P. Wound healing‐‐aiming for perfect skin regeneration. Science. 1997;276(5309):75‐81. doi: 10.1126/SCIENCE.276.5309.75 [DOI] [PubMed] [Google Scholar]
  • 59. Duffield JS, Lupher M, Thannickal VJ, Wynn TA. Host responses in tissue repair and fibrosis. Annu Rev Pathol. 2013;8:241‐276. doi: 10.1146/annurev-pathol-020712-163930 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Jenkins SJ, Ruckerl D, Cook PC, et al. Local macrophage proliferation, rather than recruitment from the blood, is a signature of TH2 inflammation. Science. 2011;332(6035):1284‐1288. doi: 10.1126/science.1204351 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Jenkins SJ, Ruckerl D, Thomas GD, et al. IL‐4 directly signals tissue‐resident macrophages to proliferate beyond homeostatic levels controlled by CSF‐1. J Exp Med. 2013;210(11):2477‐2491. doi: 10.1084/JEM.20121999 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Dal‐Secco D, Wang J, Zeng Z, et al. A dynamic spectrum of monocytes arising from the in situ reprogramming of CCR2 + monocytes at a site of sterile injury. J Exp Med. 2015;212(4):447‐456. doi: 10.1084/jem.20141539 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Gordon S. Alternative activation of macrophages. Nat Rev Immunol. 2003;3(1):23‐35. doi: 10.1038/nri978 [DOI] [PubMed] [Google Scholar]
  • 64. Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5(12):953‐964. doi: 10.1038/nri1733 [DOI] [PubMed] [Google Scholar]
  • 65. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A, Mantovani A. Macrophage polarization: tumor‐associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002;23(11):549‐555. [DOI] [PubMed] [Google Scholar]
  • 66. Engelhardt E, Toksoy A, Goebeler M, Debus S, Bröcker EB, Gillitzer R. Chemokines IL‐8, GROalpha, MCP‐1, IP‐10, and Mig are sequentially and differentially expressed during phase‐specific infiltration of leukocyte subsets in human wound healing. Am J Pathol. 1998;153(6):1849‐1860. doi: 10.1016/s0002-9440(10)65699-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. de Oliveira S, Rosowski EE, Huttenlocher A. Neutrophil migration in infection and wound repair: going forward in reverse. Nat Rev Immunol. 2016;16(6):378‐391. doi: 10.1038/nri.2016.49 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Baggiolini M, Loetscher P, Moser B. Interleukin‐8 and the chemokine family. Int J Immunopharmacol. 1995;17(2):103‐108. doi: 10.1016/0192-0561(94)00088-6 [DOI] [PubMed] [Google Scholar]
  • 69. Futosi K, Fodor S, Mócsai A. Neutrophil cell surface receptors and their intracellular signal transduction pathways. Int Immunopharmacol. 2013;17(3):638‐650. doi: 10.1016/J.INTIMP.2013.06.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Pittman K, Kubes P. Damage‐associated molecular patterns control neutrophil recruitment. J Innate Immun. 2013;5(4):315‐323. doi: 10.1159/000347132 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. de Oliveira S, López‐Muñoz A, Candel S, Pelegrín P, Calado Â, Mulero V. ATP modulates acute inflammation in vivo through dual oxidase 1‐derived H2O2 production and NF‐κB activation. J Immunol. 2014;192(12):5710‐5719. doi: 10.4049/jimmunol.1302902 [DOI] [PubMed] [Google Scholar]
  • 72. Gillitzer R, Goebeler M. Chemokines in cutaneous wound healing. J Leukoc Biol. 2001;69(4):513‐521. [PubMed] [Google Scholar]
  • 73. Simpson DM, Ross R. The neutrophilic leukocyte in wound repair a study with antineutrophil serum. J Clin Invest. 1972;51(8):2009‐2023. doi: 10.1172/JCI107007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Eming SA, Martin P, Tomic‐Canic M, et al. Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Med. 2014;6(265):3009337. doi: 10.1126/scitranslmed.3009337 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Theilgaard‐Mönch K, Knudsen S, Follin P, Borregaard N. The transcriptional activation program of human neutrophils in skin lesions supports their important role in wound healing. J Immunol. 2004;172(12):7684‐7693. doi: 10.4049/jimmunol.172.12.7684 [DOI] [PubMed] [Google Scholar]
  • 76. Ng MF. The role of mast cells in wound healing. Int Wound J. 2010;7(1):55‐61. doi: 10.1111/j.1742-481X.2009.00651.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Oskeritzian CA. Mast cells and wound healing. Adv Wound Care. 2012;1(1):23‐28. doi: 10.1089/wound.2011.0357 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Komi DEA, Khomtchouk K, Santa Maria PL. A review of the contribution of mast cells in wound healing: involved molecular and cellular mechanisms. Clin Rev Allergy Immunol. 2019;7:298‐312. doi: 10.1007/s12016-019-08729-w [DOI] [PubMed] [Google Scholar]
  • 79. Younan GJ, Heit YI, Dastouri P, et al. Mast cells are required in the proliferation and remodeling phases of microdeformational wound therapy. Plast Reconstr Surg. 2011;128(6):649 e‐58 e. doi: 10.1097/PRS.0b013e318230c55d [DOI] [PubMed] [Google Scholar]
  • 80. Weller K, Foitzik K, Paus R, Syska W, Maurer M. Mast cells are required for normal healing of skin wounds in mice. FASEB J. 2006;20(13):2366‐2368. doi: 10.1096/fj.06-5837fje [DOI] [PubMed] [Google Scholar]
  • 81. Egozi EI, Ferreira AM, Burns AL, Gamelli RL, Dipietro LA. Mast cells modulate the inflammatory but not the proliferative response in healing wounds. Wound Repair Regen. 2003;11(1):46‐54. [DOI] [PubMed] [Google Scholar]
  • 82. Iba Y, Shibata A, Kato M, Masukawa T. Possible involvement of mast cells in collagen remodeling in the late phase of cutaneous wound healing in mice. Int Immunopharmacol. 2004;4(14):1873‐1880. doi: 10.1016/J.INTIMP.2004.08.009 [DOI] [PubMed] [Google Scholar]
  • 83. Shiota N, Nishikori Y, Kakizoe E, et al. Pathophysiological role of skin mast cells in wound healing after scald injury: study with mast cell‐deficient W/W(V) mice. Int Arch Allergy Immunol. 2010;151(1):80‐88. doi: 10.1159/000232573 [DOI] [PubMed] [Google Scholar]
  • 84. Puxeddu I, Piliponsky AM, Bachelet I, Levi‐Schaffer F. Mast cells in allergy and beyond. Int J Biochem Cell Biol. 2003;35(12):1601‐1607. doi: 10.1016/s1357-2725(03)00208-5 [DOI] [PubMed] [Google Scholar]
  • 85. Maltby S, Khazaie K, McNagny KM. Mast cells in tumor growth: angiogenesis, tissue remodelling and immune‐modulation. Biochim Biophys Acta. 2009;1796(1):19‐26. doi: 10.1016/J.BBCAN.2009.02.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Artis D, Spits H. The biology of innate lymphoid cells. Nature. 2015;517(7534):293‐301. doi: 10.1038/nature14189 [DOI] [PubMed] [Google Scholar]
  • 87. Spits H, Cupedo T. Innate lymphoid cells: emerging insights in development, lineage relationships, and function. Annu Rev Immunol. 2012;30(1):647‐675. doi: 10.1146/annurev-immunol-020711-075053 [DOI] [PubMed] [Google Scholar]
  • 88. Björkström NK, Kekäläinen E, Mjösberg J. Tissue‐specific effector functions of innate lymphoid cells. Immunology. 2013;139(4):416‐427. doi: 10.1111/imm.12098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Klose CSN, Flach M, Möhle L, et al. Differentiation of type 1 ILCs from a common progenitor to all helper‐like innate lymphoid cell lineages. Cell. 2014;157(2):340‐356. doi: 10.1016/j.cell.2014.03.030 [DOI] [PubMed] [Google Scholar]
  • 90. Spits H, Artis D, Colonna M, et al. Innate lymphoid cells — a proposal for uniform nomenclature. Nat Rev Immunol. 2013;13(2):145‐149. doi: 10.1038/nri3365 [DOI] [PubMed] [Google Scholar]
  • 91. Kim BS, Siracusa MC, Saenz SA, et al. TSLP elicits IL‐33‐independent innate lymphoid cell responses to promote skin inflammation. Sci Transl Med. 2013;5(170):3005374. doi: 10.1126/scitranslmed.3005374 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Roediger B, Kyle R, Yip KH, et al. Cutaneous immunosurveillance and regulation of inflammation by group 2 innate lymphoid cells. Nat Immunol. 2013;14(6):564‐573. doi: 10.1038/ni.2584 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Salimi M, Barlow JL, Saunders SP, et al. A role for IL‐25 and IL‐33–driven type‐2 innate lymphoid cells in atopic dermatitis. J Exp Med. 2013;210(13):2939‐2950. doi: 10.1084/jem.20130351 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Yin H, Li X, Hu S, et al. IL‐33 accelerates cutaneous wound healing involved in upregulation of alternatively activated macrophages. Mol Immunol. 2013;56(4):347‐353. doi: 10.1016/j.molimm.2013.05.225 [DOI] [PubMed] [Google Scholar]
  • 95. Yin H, Li X, Hu S, et al. IL‐33 promotes Staphylococcus aureus‐infected wound healing in mice. Int Immunopharmacol. 2013;17(2):432‐438. doi: 10.1016/j.intimp.2013.07.008 [DOI] [PubMed] [Google Scholar]
  • 96. Rak GD, Osborne LC, Siracusa MC, et al. IL‐33‐dependent group 2 innate lymphoid cells promote cutaneous wound healing. J Invest Dermatol. 2016;136(2):487‐496. doi: 10.1038/JID.2015.406 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Sanchez Rodriguez R, Pauli ML, Neuhaus IM, et al. Memory regulatory T cells reside in human skin. J Clin Invest. 2014;124(3):1027‐1036. doi: 10.1172/JCI72932 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Gratz IK, Truong H‐A, Yang SH‐Y, et al. Cutting edge: memory regulatory T cells require IL‐7 and not IL‐2 for their maintenance in peripheral tissues. J Immunol. 2013;190(9):4483‐4487. doi: 10.4049/jimmunol.1300212 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Nosbaum A, Prevel N, Truong H‐A, et al. Cutting edge: regulatory T cells facilitate cutaneous wound healing. J Immunol. 2016;196(5):2010‐2014. doi: 10.4049/jimmunol.1502139 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Barros JF, Waclawiak I, Pecli C, et al. Role of chemokine receptor CCR4 and regulatory T cells in wound healing of diabetic mice. J Invest Dermatol. 2019;139(5):1161‐1170. doi: 10.1016/j.jid.2018.10.039 [DOI] [PubMed] [Google Scholar]
  • 101. Rapala K, Laato M, Niinikoski J, et al. Tumor necrosis factor alpha inhibits wound healing in the rat. Eur Surg Res. 1991;23(5–6):261‐268. doi: 10.1159/000129163 [DOI] [PubMed] [Google Scholar]
  • 102. Mori R, Kondo T, Ohshima T, Ishida Y, Mukaida N. Accelerated wound healing in tumor necrosis factor receptor p55‐deficient mice with reduced leukocyte infiltration. FASEB J. 2002;16(9):963‐974. doi: 10.1096/fj.01-0776com [DOI] [PubMed] [Google Scholar]
  • 103. Korn T, Bettelli E, Oukka M, Kuchroo VK. IL‐17 and Th17 cells. Annu Rev Immunol. 2009;27(1):485‐517. doi: 10.1146/annurev.immunol.021908.132710 [DOI] [PubMed] [Google Scholar]
  • 104. Miossec P, Kolls JK. Targeting IL‐17 and T H 17 cells in chronic inflammation. Nat Rev Drug Discov. 2012;11(10):763‐776. doi: 10.1038/nrd3794 [DOI] [PubMed] [Google Scholar]
  • 105. Wilson MS, Madala SK, Ramalingam TR, et al. Bleomycin and IL‐1β‐mediated pulmonary fibrosis is IL‐17A dependent. J Exp Med. 2010;207(3):535‐552. doi: 10.1084/jem.20092121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Okamoto Y, Hasegawa M, Matsushita T, et al. Potential roles of interleukin‐17A in the development of skin fibrosis in mice. Arthritis Rheum. 2012;64(11):3726‐3735. doi: 10.1002/art.34643 [DOI] [PubMed] [Google Scholar]
  • 107. Meng F, Wang K, Aoyama T, et al. Interleukin‐17 signaling in inflammatory, Kupffer cells, and hepatic stellate cells exacerbates liver fibrosis in mice. Gastroenterology. 2012;143(3):765‐776. doi: 10.1053/j.gastro.2012.05.049 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. MacLeod AS, Hemmers S, Garijo O, et al. Dendritic epidermal T cells regulate skin antimicrobial barrier function. J Clin Invest. 2013;123(10):4364‐4374. doi: 10.1172/JCI70064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Lai Y, Li D, Li C, et al. The antimicrobial protein REG3A regulates keratinocyte proliferation and differentiation after skin injury. Immunity. 2012;37(1):74‐84. doi: 10.1016/j.immuni.2012.04.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Larouche J, Sheoran S, Maruyama K, Martino MM. Immune regulation of skin wound healing: mechanisms and novel therapeutic targets. Adv Wound Care. 2018;7(7):209‐231. doi: 10.1089/wound.2017.0761 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Landén NX, Li D, Ståhle M. Transition from inflammation to proliferation: a critical step during wound healing. Cell Mol Life Sci. 2016;73(20):3861‐3885. doi: 10.1007/s00018-016-2268-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Nguyen AV, Soulika AM. The dynamics of the skin's immune system. Int J Mol Sci. 2019;20(8):1‐53. doi: 10.3390/ijms20081811 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Nagaraja S, Chen L, DiPietro LA, Reifman J, Mitrophanov AY. Predictive approach identifies molecular targets and interventions to restore angiogenesis in wounds with delayed healing. Front Physiol. 2019;10:00636. doi: 10.3389/fphys.2019.00636 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Niessen FB, Andriessen MP, Schalkwijk J, Visser L, Timens W. Keratinocyte‐derived growth factors play a role in the formation of hypertrophic scars. J Pathol. 2001;194(2):207‐216. doi: 10.1002/path.853 [DOI] [PubMed] [Google Scholar]
  • 115. Armour A, Scott PG, Tredget EE. Cellular and molecular pathology of HTS: basis for treatment. Wound Repair Regen. 2007;15(S1):S6‐S17. doi: 10.1111/j.1524-475X.2007.00219.x [DOI] [PubMed] [Google Scholar]
  • 116. Marshall CD, Hu MS, Leavitt T, Barnes LA, Lorenz HP, Longaker MT. Cutaneous scarring: basic science, current treatments, and future directions. Adv Wound Care. 2018;7(2):29‐45. doi: 10.1089/wound.2016.0696 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Benson A, Dickson WA, Boyce DE. ABC of wound healing: Burns. Br Med J. 2006;332(7542):649‐652. doi: 10.1136/sbmj.0609324 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Ogawa R. Keloid and hypertrophic scars are the result of chronic inflammation in the reticular dermis. Int J Mol Sci. 2017;18(606):1‐10. doi: 10.3390/ijms18030606 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Ogawa R. Recent Advances in Scar Biology. Int J Mol Sci. 2018;19:19061749. doi: 10.3390/ijms19061749 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Qian LW, Fourcaudot AB, Yamane K, You T, Chan RK, Leung KP. Exacerbated and prolonged inflammation impairs wound healing and increases scarring. Wound Repair Regen. 2016;24(1):26‐34. doi: 10.1111/wrr.12381 [DOI] [PubMed] [Google Scholar]
  • 121. Krotzsch‐Gomez FE, Furuzawa‐Carballeda J, Reyes‐Marquez R, Quiroz‐ Hernandez E, Diaz de Leon L. Cytokine expression is downregulated by collagen‐polyvinylpyrrolidone in hypertrophic scars. J Invest Dermatol. 1998;111(5):828‐834. doi: 10.1046/j.1523-1747.1998.00329.x [DOI] [PubMed] [Google Scholar]
  • 122. Butzelaar L, Schooneman DPM, Soykan EA, et al. Inhibited early immunologic response is associated with hypertrophic scarring. Exp Dermatol. 2016;25(10):797‐804. doi: 10.1111/exd.13100 [DOI] [PubMed] [Google Scholar]
  • 123. Wang JF, Jiao H, Stewart TL, Shankowsky HA, Scott PG, Tredget EE. Increased TGF‐β‐producing CD4+ T lymphocytes in postburn patients and their potential interaction with dermal fibroblasts in hypertrophic scarring. Wound Repair Regen. 2007;15(4):530‐539. doi: 10.1111/j.1524-475X.2007.00261.x [DOI] [PubMed] [Google Scholar]
  • 124. Zhou J, Zhao Y, Simonenko V, et al. Simultaneous silencing of TGF‐β1 and COX‐2 reduces human skin hypertrophic scar through activation of fibroblast apoptosis. Oncotarget. 2017;8(46):80651‐80665. doi: 10.18632/oncotarget.20869 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Tredget EE, Yang L, Delehanty M, Shankowsky H, Scott PG. Polarized Th2 cytokine production in patients with hypertrophic scar following thermal injury. J Interferon Cytokine Res. 2006;26(3):179‐189. doi: 10.1089/jir.2006.26.179 [DOI] [PubMed] [Google Scholar]
  • 126. Wynn TA. FIBROTIC DISEASE AND THE TH 1/TH 2 PARADIGM Thomas. Nat Rev Immunol. 2004;4(8):583‐594. doi: 10.1038/nri1412.FIBROTIC [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Kilani RT, Delehanty M, Shankowsky HA, Ghahary A, Scott P, Tredget EE. Fluorescent‐activated cell‐sorting analysis of intracellular interferon‐γ and interleukin‐4 in fresh and frozen human peripheral blood T‐helper cells. Wound Repair Regen. 2005;13(4):441‐449. doi: 10.1111/j.1067-1927.2005.130412.x [DOI] [PubMed] [Google Scholar]
  • 128. Wang R, Ghahary A, Shen YJ, Scott PG, Tredget EE. Human dermal fibroblasts produce nitric oxide and express both constitutive and inducible nitric oxide synthase isoforms. J Invest Dermatol. 1996;106(3):419‐427. doi: 10.1111/1523-1747.ep12343428 [DOI] [PubMed] [Google Scholar]
  • 129. Daley JM, Brancato SK, Thomay AA, Reichner JS, Albina JE. The phenotype of murine wound macrophages. J Leukoc Biol. 2010;87(1):59‐67. doi: 10.1189/jlb.0409236 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Radhika M, Devalaraja RM, Nanney LB, et al. Delayed wound healing in CXCR2 knockout mice. J Invest Dermatol. 2000;115(2):234‐244. doi: 10.1161/CIRCULATIONAHA.110.956839 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Dovi JV, He L‐K, DiPietro LA. Accelerated wound closure in neutrophil‐depleted mice. J Leukoc Biol. 2003;73(4):448‐455. doi: 10.1189/jlb.0802406 [DOI] [PubMed] [Google Scholar]
  • 132. Hesketh M, Sahin KB, West ZE, Murray RZ. Macrophage phenotypes regulate scar formation and chronic wound healing. Int J Mol Sci. 2017;18(7):1‐10. doi: 10.3390/ijms18071545 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Bagabir R, Byers RJ, Chaudhry IH, Muller W, Paus R, Bayat A. Site‐specific immunophenotyping of keloid disease demonstrates immune upregulation and the presence of lymphoid aggregates. Br J Dermatol. 2012;167(5):1053‐1066. doi: 10.1111/j.1365-2133.2012.11190.x [DOI] [PubMed] [Google Scholar]
  • 134. Sziksz E, Pap D, Lippai R, et al. Fibrosis related inflammatory mediators: role of the IL‐10 cytokine family. Mediators Inflamm. 2015;2015:764641. doi: 10.1155/2015/764641 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Marneros AG, Krieg T. Keloids ‐ clinical diagnosis, pathogenesis, and treatment options. JDDG. 2004;2(11):905‐913. doi: 10.1046/j.1439-0353.2004.04077.x [DOI] [PubMed] [Google Scholar]
  • 136. Butler PD, Ly DP, Longaker MT, Yang GP. Use of organotypic coculture to study keloid biology. Am J Surg. 2008;195(2):144‐148. doi: 10.1016/j.amjsurg.2007.10.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Harrison CA, MacNeil S. The mechanism of skin graft contraction: an update on current research and potential future therapies. Burns. 2008;34(2):153‐163. doi: 10.1016/J.BURNS.2007.08.011 [DOI] [PubMed] [Google Scholar]
  • 138. van den Broek LJ, Limandjaja GC, Niessen FB, Gibbs S. Human hypertrophic and keloid scar models: principles, limitations and future challenges from a tissue engineering perspective. Exp Dermatol. 2014;23(6):382‐386. doi: 10.1111/exd.12419 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Seok J, Warren HS, Cuenca AG, et al. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc Natl Acad Sci. 2013;110(9):3507‐3512. doi: 10.1073/pnas.1222878110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Shih B, Garside E, McGrouther DA, Bayat A. Molecular dissection of abnormal wound healing processes resulting in keloid disease. Wound Repair Regen. 2010;18(2):139‐153. doi: 10.1111/j.1524-475X.2009.00553.x [DOI] [PubMed] [Google Scholar]
  • 141. He S, Liu X, Yang Y, et al. Mechanisms of transforming growth factor β 1 /Smad signalling mediated by mitogen‐activated protein kinase pathways in keloid fibroblasts. Br J Dermatol. 2010;162(3):538‐546. doi: 10.1111/j.1365-2133.2009.09511.x [DOI] [PubMed] [Google Scholar]
  • 142. Muthusubramaniam L, Zaitseva T, Paukshto M, Martin G, Desai T. Effect of collagen Nanotopography on keloid fibroblast proliferation and matrix synthesis: implications for dermal wound healing. Tissue Eng Part A. 2014;20(19–20):2728‐2736. doi: 10.1089/ten.tea.2013.0539 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Mack M. Inflammation and fibrosis. Matrix Biol. 2018;68‐69:106‐121. doi: 10.1016/j.matbio.2017.11.010 [DOI] [PubMed] [Google Scholar]
  • 144. Xie JL, Qi SH, Pan S, et al. Expression of smad protein by normal skin fibroblasts and hypertrophic scar fibroblasts in response to transforming growth factor β1. Dermatologic Surg. 2008;34(9):1216‐1225. doi: 10.1111/j.1524-4725.2008.34261.x [DOI] [PubMed] [Google Scholar]
  • 145. He K, Am L, Morris C, Ej W, Ub H, Wj C. Investigation of the expression of the extracellular matrix glycoproteins tenascin and fibronectin during acne vulgaris. Br J Dermatol. 1994;130(5):576‐582. doi: 10.1111/j.1365-2133.1994.tb13102.x [DOI] [PubMed] [Google Scholar]
  • 146. Chaudet KM, Goyal A, Veprauskas KR, Nazarian RM. Wnt signaling pathway proteins in scar, hypertrophic scar, and keloid: evidence for a continuum? Am J Dermatopathol. 2020;42(11):842‐847. doi: 10.1097/DAD.0000000000001661 [DOI] [PubMed] [Google Scholar]
  • 147. Sato M. Upregulation of the Wnt/β‐catenin pathway induced by transforming growth factor‐β in hypertrophic scars and keloids. Acta Derm Venereol. 2006;86(4):300‐307. doi: 10.2340/00015555-0101 [DOI] [PubMed] [Google Scholar]
  • 148. Bertheim U, Hellström S. The distribution of hyaluronan in human skin and mature, hypertrophic and keloid scars. Br J Plast Surg. 1994;47(7):483‐489. doi: 10.1016/0007-1226(94)90031-0 [DOI] [PubMed] [Google Scholar]
  • 149. Yin L, Zhao X, Ji S, et al. The use of gene activated matrix to mediate effective SMAD2 gene silencing against hypertrophic scar. Biomaterials. 2014;35(8):2488‐2498. doi: 10.1016/j.biomaterials.2013.12.015 [DOI] [PubMed] [Google Scholar]
  • 150. Chun Q, Zhiyong W, Fei S, Xiqiao W. Dynamic biological changes in fibroblasts during hypertrophic scar formation and regression. Int Wound J. 2016;13(2):257‐262. doi: 10.1111/iwj.12283 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Čoma M, Fröhlichová L, Urban L, et al. Molecular changes underlying hypertrophic scarring following burns involve specific deregulations at allwound healing stages (inflammation, proliferation and maturation). Int J Mol Sci. 2021;22(2):1‐20. doi: 10.3390/ijms22020897 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Short WD, Wang X, Keswani SG. The role of T lymphocytes in cutaneous scarring. Adv Wound Care. 2022;11(3):121‐131. doi: 10.1089/wound.2021.0059 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Amadeu TP, Braune AS, Porto LC, Desmoulière A, Costa AMA. Fibrillin‐1 and elastin are differentially expressed in hypertrophic scars and keloids. Wound Repair Regen. 2004;12(2):169‐174. doi: 10.1111/j.1067-1927.2004.012209.x [DOI] [PubMed] [Google Scholar]
  • 154. Mukhopadhyay A, Sui YC, Lim IJ, Phillips DJ, Phan TT. The role of the activin system in keloid pathogenesis. Am J Physiol Cell Physiol. 2007;292(4):1331‐1338. doi: 10.1152/ajpcell.00373.2006 [DOI] [PubMed] [Google Scholar]
  • 155. Sible JC, Rettig WJ, Eriksson E, Smith SP, Oliver N. Gene expression of tenascin is altered in normal scars and keloids. Wound Repair Regen. 1995;3(1):37‐48. doi: 10.1046/j.1524-475X.1995.30109.x [DOI] [PubMed] [Google Scholar]
  • 156. Babu M, Diegelmann R, Oliver N. Fibronectin is overproduced by keloid fibroblasts during abnormal wound healing. Mol Cell Biol. 1989;9(4):1642‐1650. doi: 10.1128/mcb.9.4.1642 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Igota S, Tosa M, Murakami M, et al. Identification and characterization of Wnt signaling pathway in keloid pathogenesis. Int J Med Sci. 2013;10(4):344‐354. doi: 10.7150/ijms.5349 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Hahn JM, McFarland KL, Combs KA, Supp DM. Partial epithelial‐mesenchymal transition in keloid scars: regulation of keloid keratinocyte gene expression by transforming growth factor‐β1. BurnS Trauma. 2016;4(1):30. doi: 10.1186/s41038-016-0055-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Wang ZC, Zhao WY, Cao Y, et al. The roles of inflammation in keloid and hypertrophic scars. Front Immunol. 2020;11:1‐10. doi: 10.3389/fimmu.2020.603187 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Yu D, Shang Y, Yuan J, Ding S, Luo S, Hao L. Wnt/β‐catenin signaling exacerbates keloid cell proliferation by regulating telomerase. Cell Physiol Biochem. 2016;39(5):2001‐2013. doi: 10.1159/000447896 [DOI] [PubMed] [Google Scholar]
  • 161. Tan KT, McGrouther DA, Day AJ, Milner CM, Bayat A. Characterization of hyaluronan and TSG‐6 in skin scarring: differential distribution in keloid scars, normal scars and unscarred skin. J Eur Acad Dermatology Venereol. 2011;25(3):317‐327. doi: 10.1111/j.1468-3083.2010.03792.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Dalkowski A, Schuppan D, Orfanos CE, Zouboulis CC. Increased expression of tenascin C by keloids in vivo and in vitro. Br J Dermatol. 1999;141(1):50‐56. doi: 10.1046/j.1365-2133.1999.02920.x [DOI] [PubMed] [Google Scholar]
  • 163. Chua AWC, Gan SU, Ting Y, et al. Keloid fibroblasts are more sensitive to Wnt3a treatment in terms of elevated cellular growth and fibronectin expression. J Dermatol Sci. 2011;64(3):199‐209. doi: 10.1016/j.jdermsci.2011.09.008 [DOI] [PubMed] [Google Scholar]
  • 164. Chen ZY, Yu XF, Huang JQ, Li DL. The mechanisms of β‐catenin on keloid fibroblast cells proliferation and apoptosis. Eur Rev Med Pharmacol Sci. 2018;22(4):888‐895. [DOI] [PubMed] [Google Scholar]
  • 165. Lim IJ, Phan TT, Tan EK, et al. Synchronous activation of ERK and phosphatidylinositol 3‐kinase pathways is required for collagen and extracellular matrix production in keloids. J Biol Chem. 2003;278(42):40851‐40858. doi: 10.1074/jbc.M305759200 [DOI] [PubMed] [Google Scholar]
  • 166. Werner S, Krieg T, Smola H. Keratinocyte‐fibroblast interactions in wound healing. J Invest Dermatol. 2007;127(5):998‐1008. doi: 10.1038/sj.jid.5700786 [DOI] [PubMed] [Google Scholar]
  • 167. Liu CJ, Tahara S, Gao S. Phosphorylation of extracellular signal‐regulated protein kinase in cultured keloid fibroblasts when stimulated by platelet‐derived growth factor BB. Scand J Plast Reconstr Surg Hand Surg. 2003;37(6):321‐324. doi: 10.1080/02844310310004677 [DOI] [PubMed] [Google Scholar]
  • 168. Cheng B, Fu X, Sun T, Sun X, Sheng Z. Expression of epidermal growth factor receptor and related phosphorylation proteins in hypertrophic scars and normal skin. Chin Med J (Engl). 2002;115(10):1525‐1528. [PubMed] [Google Scholar]
  • 169. Hakvoort TE, Altun V, Van Zuijlen PPM, De Boer WI, Van Schadewijk WAAM, Van der Kwast TH. Transforming growth factor‐beta1, −beta2, −beta3, basic fibroblast growth factor and vascular endothelial growth factor expression in keratinocytes of burn scars. Eur Cytokine Netw. 2020;11(2):233‐239. https://research.vumc.nl/en/publications/transforming‐growth‐factor‐tgf‐βsub1sub‐βsub2sub‐βsub3sub‐basic‐f [PubMed] [Google Scholar]
  • 170. Lu L, Saulis AS, Liu WR, et al. The temporal effects of anti‐TGF‐β1, 2, and 3 monoclonal antibody on wound healing and hypertrophic scar formation. J Am Coll Surg. 2005;201(3):391‐397. doi: 10.1016/j.jamcollsurg.2005.03.032 [DOI] [PubMed] [Google Scholar]
  • 171. Niessen FB, Andriessen MP, Schalkwijk J, Visser L, Timens W. Keratinocyte‐derived growth factors play a role in the formation of hypertrophic scars. J Pathol. 2001;194(2):207‐216. doi: 10.1002/path.853 [DOI] [PubMed] [Google Scholar]
  • 172. Hahn JM, Glaser K, McFarland KL, Aronow BJ, Boyce ST, Supp DM. Keloid‐derived keratinocytes exhibit an abnormal gene expression profile consistent with a distinct causal role in keloid pathology. Wound Repair Regen. 2013;21(4):530‐544. doi: 10.1111/wrr.12060 [DOI] [PubMed] [Google Scholar]
  • 173. Xue M, Jackson CJ. Extracellular matrix reorganization during wound healing and its impact on abnormal scarring. Adv Wound Care. 2015;4(3):119‐136. doi: 10.1089/wound.2013.0485 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Sidgwick GP, Bayat A. Extracellular matrix molecules implicated in hypertrophic and keloid scarring _ enhanced reader. J Eur Acad Dermatol Venereol. 2011;26(2):141‐152. doi: 10.1111/j.1468-3083.2011.04200.x [DOI] [PubMed] [Google Scholar]
  • 175. Kaan Kaltalioglu SC‐C . A bioactive molecule in a complex wound healing process_ platelet‐derived growth factor _ enhanced reader. Int J Dermatol. 2015;54(8):972‐977. doi: 10.1111/ijd.12731 [DOI] [PubMed] [Google Scholar]
  • 176. Drayton DL, Liao S, Mounzer RH, Ruddle NH. Lymphoid organ development: from ontogeny to neogenesis. Nat Immunol. 2006;7(4):344‐353. doi: 10.1038/ni1330 [DOI] [PubMed] [Google Scholar]
  • 177. Cupedo T, Jansen W, Kraal G, Mebius RE. Induction of secondary and tertiary lymphoid structures in the skin. Immunity. 2004;21(5):655‐667. doi: 10.1016/j.immuni.2004.09.006 [DOI] [PubMed] [Google Scholar]
  • 178. Rangel‐Moreno J, Hartson L, Navarro C, Gaxiola M, Selman M, Randall TD. Inducible bronchus‐associated lymphoid tissue (iBALT) in patients with pulmonary complications of rheumatoid arthritis. J Clin Invest. 2006;116(12):3183‐3194. doi: 10.1172/JCI28756 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Rodero MP, Khosrotehrani K. Skin wound healing modulation by macrophages. Int J Clin Exp Pathol. 2010;3(7):643‐653. Accessed September 16, 2019. http://www.ncbi.nlm.nih.gov/pubmed/20830235 [PMC free article] [PubMed] [Google Scholar]
  • 180. Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 2011;11(11):723‐737. doi: 10.1038/nri3073 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Grant C, Chudakova DA, Itinteang T, et al. Expression of embryonic stem cell markers in keloid‐associated lymphoid tissue. J Clin Pathol. 2016;69(7):643‐646. doi: 10.1136/jclinpath-2015-203483 [DOI] [PubMed] [Google Scholar]
  • 182. Lim KH, Itinteang T, Davis PF, Tan ST. Stem cells in keloid lesions: A Review. Plast Reconstr Surg ‐ Glob Open. 2019;7(5):e2228. doi: 10.1097/gox.0000000000002228 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Zhang Q, Yamaza T, Kelly AP, et al. Tumor‐like stem cells derived from human keloid are governed by the inflammatory niche driven by IL‐17/IL‐6 axis. PLoS One. 2009;4(11):1‐16. doi: 10.1371/journal.pone.0007798 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Smith CJ, Smith JC, Finn MC. The possible role of mast cells (allergy) in the production of keloid and hypertrophic scarring. J Burn Care Rehabil. 1987;8(2):126‐131. doi: 10.1097/00004630-198703000-00008 [DOI] [PubMed] [Google Scholar]
  • 185. Ong CT, Khoo YT, Mukhopadhyay A, et al. Comparative proteomic analysis between normal skin and keloid scar. Br J Dermatol. 2010;162(6):1302‐1315. doi: 10.1111/j.1365-2133.2010.09660.x [DOI] [PubMed] [Google Scholar]
  • 186. Hunasgi S, Koneru A, Vanishree M, Shamala R. Keloid: a case report and review of pathophysiology and differences between keloid and hypertrophic scars. J Oral Maxillofac Pathol. 2013;17(1):116‐120. doi: 10.4103/0973-029X.110701 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Zhang Q, Oh CK, Messadi DV, et al. Hypoxia‐induced HIF‐1 α accumulation is augmented in a co‐culture of keloid fibroblasts and human mast cells: involvement of ERK1/2 and PI‐3K/Akt. Exp Cell Res. 2006;312(2):145‐155. doi: 10.1016/j.yexcr.2005.10.006 [DOI] [PubMed] [Google Scholar]
  • 188. Shaker SA, Ayuob NN, Hajrah NH. Cell talk: a phenomenon observed in the keloid scar by immunohistochemical study. Appl Immunohistochem Mol Morphol. 2011;19(2):153‐159. doi: 10.1097/PAI.0b013e3181efa2ef [DOI] [PubMed] [Google Scholar]
  • 189. Ueda K, Yasuda Y, Furuya E, Oba S. Inadequate blood supply persists in keloids. Scand J Plast Reconstr Surg Hand Surg. 2004;38(5):267‐271. doi: 10.1080/02844310410029552 [DOI] [PubMed] [Google Scholar]
  • 190. Steinbrech DS, Mehrara BJ, Chau D, et al. Hypoxia upregulates VEGF production in keloid febroblasts. Ann Plast Surg. 1999;42(5):514‐520. doi: 10.1097/00000637-199905000-00009 [DOI] [PubMed] [Google Scholar]
  • 191. Li Q, Qin Z, Nie F, et al. Metabolic reprogramming in keloid fibroblasts: aerobic glycolysis and a novel therapeutic strategy. Biochem Biophys Res Commun. 2018;496(2):641‐647. doi: 10.1016/j.bbrc.2018.01.068 [DOI] [PubMed] [Google Scholar]
  • 192. Semenza GL. Regulation of mammalian O2 homeostasis by hypoxia‐inducible factor 1. Annu Rev Cell Dev Biol. 1999;15:551‐578. www.annualreviews.org [DOI] [PubMed] [Google Scholar]
  • 193. Zhang Q, Wu Y, Ann DK, et al. Mechanisms of hypoxic regulation of plasminogen activator Inhibitor‐1 gene expression in keloid fibroblasts. J Invest Dermatol. 2003;121(5):1005‐1012. doi: 10.1046/j.1523-1747.2003.12564.x [DOI] [PubMed] [Google Scholar]
  • 194. Wu Y, Zhang Q, Ann DK, et al. Increased vascular endothelial growth factor may account for elevated level of plasminogen activator inhibitor‐1 via activating ERK1/2 in keloid fibroblasts. Am J Physiol Physiol. 2004;286:C905‐C912. doi: 10.1152/ajpcell.00200.2003.-Ke [DOI] [PubMed] [Google Scholar]
  • 195. Boyce DE, Ciampolini J, Ruge F, Harding KG, Murison MMSC. Inflammatory cell subpopulations in keloid scars. Br J Plast Surg. 2001;54(6):511‐516. doi: 10.1054/bjps.2001.3638 [DOI] [PubMed] [Google Scholar]
  • 196. Movahedi K, Laoui D, Gysemans C, et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 2010;70(14):5728‐5739. doi: 10.1158/0008-5472.CAN-09-4672 [DOI] [PubMed] [Google Scholar]
  • 197. Boyle JJ, Harrington HA, Piper E, et al. Coronary intraplaque hemorrhage evokes a novel atheroprotective macrophage phenotype. Am J Pathol. 2009;174(3):1097‐1108. doi: 10.2353/ajpath.2009.080431 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Chang YC, Chen TC, Lee CT, et al. Epigenetic control of MHC class II expression in tumor‐associated macrophages by decoy receptor 3. Blood. 2008;111(10):5054‐5063. doi: 10.1182/blood-2007-12-130609 [DOI] [PubMed] [Google Scholar]
  • 199. Jiao H, Fan J, Cai J, et al. Analysis of characteristics similar to autoimmune disease in keloid patients. Aesthetic Plast Surg. 2015;39(5):818‐825. doi: 10.1007/s00266-015-0542-4 [DOI] [PubMed] [Google Scholar]
  • 200. Li X, Wang Y, Yuan B, Yang H, Qiao L. Status of M1 and M2 type macrophages in keloid. Int J Clin Exp Pathol. 2017;10(11):11098‐11105. [PMC free article] [PubMed] [Google Scholar]
  • 201. Brown JJ, WERO, Thomson W, Bayat A. Positive association of HLA‐DRB1 * 15 with keloid disease in Caucasians. Int J Immunogenet. 2008;35:303‐307. doi: 10.1111/j.1744-313X.2008.00780.x [DOI] [PubMed] [Google Scholar]
  • 202. Lu W‐S, Wang J‐F, Yang S, et al. Association of HLA‐DQA1 and DQB1 alleles with keloids in Chinese Hans. J Dermatol Sci. 2008;52(2):108‐117. doi: 10.1016/j.jdermsci.2008.04.010 [DOI] [PubMed] [Google Scholar]
  • 203. Rossi A, Bozzi M. HLA and keloids: antigenic frequency and therapeutic response. G Ital Dermatol Venereol. 2019; Accessed October 1, 2019;124(7–8):341‐344. http://www.ncbi.nlm.nih.gov/pubmed/2628278 [PubMed] [Google Scholar]
  • 204. Limandjaja GC, Waaijman T, Roffel S, Niessen FB, Gibbs S. Monocytes co‐cultured with reconstructed keloid and normal skin models skew towards M2 macrophage phenotype. Arch Dermatol Res. 2019;311:615‐627. doi: 10.1007/s00403-019-01942-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. Li J, Cao J, Li M, et al. Collagen triple helix repeat containing‐1 inhibits transforming growth factor‐β1‐induced collagen type I expression in keloid. Br J Dermatol. 2011;164(5):1030‐1036. doi: 10.1111/j.1365-2133.2011.10215.x [DOI] [PubMed] [Google Scholar]
  • 206. Shang QX, Yuan R, Wang W. Expression of platelet derived growth factor receptor‐beta in fibroblasts of keloid. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi. 2000;14(5):278‐282. Accessed March 9, 2019. http://www.ncbi.nlm.nih.gov/pubmed/12516475 [PubMed] [Google Scholar]
  • 207. Hu Z‐C, Tang B, Guo D, et al. Expression of insulin‐like growth factor‐1 receptor in keloid and hypertrophic scar. Clin Exp Dermatol. 2014;39(7):822‐828. doi: 10.1111/ced.12407 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Ghazizadeh M. Essential role of IL‐6 signaling pathway in keloid pathogenesis. J Nippon Med Sch. 2007;74(1):11‐22. http://www.ncbi.nlm.nih.gov/pubmed/17384473 [DOI] [PubMed] [Google Scholar]
  • 209. Gauglitz G, Korting H. Hypertrophic scarring and keloids: Pathomechanisms and current and emerging treatment strategies. Mol Med. 2011;17(1–2):113‐125. doi: 10.2119/molmed.2009.00153 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210. McCauley RL, Chopra V, Li YY, Herndon DN, Robson MC. Altered cytokine production in black patients with keloids. J Clin Immunol. 1992;12(4):300‐308. doi: 10.1007/BF00918154 [DOI] [PubMed] [Google Scholar]
  • 211. Brown JJ, Bayat A. Genetic susceptibility to raised dermal scarring. Br J Dermatol. 2009;161:8‐18. doi: 10.1111/j.1365-2133.2009.09258.x [DOI] [PubMed] [Google Scholar]
  • 212. Ghazizadeh M, Tosa M, Shimizu H, Hyakusoku H, Kawanami O. Functional implications of the IL‐6 signaling pathway in keloid pathogenesis. J Invest Dermatol. 2007;127(1):98‐105. doi: 10.1038/sj.jid.5700564 [DOI] [PubMed] [Google Scholar]
  • 213. Chang Q, Bournazou E, Sansone P, et al. The IL‐6/JAK/Stat3 feed‐forward loop drives tumorigenesis and metastasis. Neoplasia (United States). 2013;15(7):848‐862. doi: 10.1593/neo.13706 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Lim CP, Phan T‐T, Lim IJ, Cao X. Stat3 contributes to keloid pathogenesis via promoting collagen production, cell proliferation and migration. Oncogene. 2006;25(39):5416‐5425. doi: 10.1038/sj.onc.1209531 [DOI] [PubMed] [Google Scholar]
  • 215. da Silva IR, da Tiveron LCRC, da Silva MV, et al. In situ cytokine expression and morphometric evaluation of total collagen and collagens type I and type III in keloid scars. Mediators Inflamm. 2017;2017:6573802. doi: 10.1155/2017/6573802 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216. Diaz A, Tan K, He H, et al. Keloid lesions show increased IL‐4/IL‐13 signaling and respond to Th2‐targeting dupilumab therapy. J Eur Acad Dermatol Venereol. 2019;34(4):e161‐e164. doi: 10.1111/jdv.16097 [DOI] [PubMed] [Google Scholar]
  • 217. Oriente A, Fedarko NS, Pacocha SE, Huang SK, Lichtenstein LM, Essayan DM. Interleukin‐13 modulates collagen homeostasis in human skin and keloid fibroblasts. J Pharmacol Exp Ther. 2000;292(3):988‐994. [PubMed] [Google Scholar]
  • 218. Chen Z, Zhou L, Won T, Gao Z, Wu X, Lu L. Characterization of CD45RO + memory T lymphocytes in keloid disease. Br J Dermatol. 2018;178(4):940‐950. doi: 10.1111/bjd.16173 [DOI] [PubMed] [Google Scholar]
  • 219. Santucci M, Borgognoni L, Reali UM, Gabbiani G. Keloids and hypertrophic scars of Caucasians show distinctive morphologic and immunophenotypic profiles. Virchows Arch. 2001;438(5):457‐463. Accessed September 16, 2019. http://www.ncbi.nlm.nih.gov/pubmed/11407473 [DOI] [PubMed] [Google Scholar]
  • 220. Murao N, Kichiro S, Hayashi T, et al. Treg‐enriched CD4+ T cells attenuate collagen synthesis in keloid fibroblasts. Exp Dermatol. 2014;23(4):266‐271. doi: 10.1111/exd.12368 [DOI] [PubMed] [Google Scholar]
  • 221. Meyer LJ, Russell SB, Russell JD, et al. Reduced hyaluronan in keloid tissue and cultured keloid fibroblasts. J Invest Dermatol. 2000;114(5):953‐959. doi: 10.1046/j.1523-1747.2000.00950.x [DOI] [PubMed] [Google Scholar]
  • 222. Lim IJ, Phan T‐T, Bay B‐H, et al. Fibroblasts cocultured with keloid keratinocytes: normal fibroblasts secrete collagen in a keloidlike manner. Am J Physiol Physiol. 2002;283(1):C212‐C222. doi: 10.1152/ajpcell.00555.2001 [DOI] [PubMed] [Google Scholar]
  • 223. Gröne A. Keratinocytes and cytokines. Vet Immunol Immunopathol. 2002;88(1–2):1‐12. doi: 10.1016/S0165-2427(02)00136-8 [DOI] [PubMed] [Google Scholar]
  • 224. Funayama E, Chodon T, Oyama A, Sugihara T. Keratinocytes promote proliferation and inhibit apoptosis of the underlying fibroblasts: an important role in the pathogenesis of keloid. J Invest Dermatol. 2003;121(6):1326‐1331. doi: 10.1111/j.1523-1747.2003.12572.x [DOI] [PubMed] [Google Scholar]
  • 225. Hahn JM, Glaser K, McFarland KL, Aronow BJ, Boyce ST, Supp DM. Keloid‐derived keratinocytes exhibit an abnormal gene expression profile consistent with a distinct causal role in keloid pathology. Wound Repair Regen. 2013;21(4):530‐544. doi: 10.1111/wrr.12060 [DOI] [PubMed] [Google Scholar]
  • 226. Satish L, Evdokiou A, Geletu E, Hahn JM, Supp DM. Pirfenidone inhibits epithelial–mesenchymal transition in keloid keratinocytes. Burn Trauma. 2020;8:1‐11. doi: 10.1093/burnst/tkz007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. Na K, Oh SH, Kim SK. Acne keloidalis nuchae in Asian: a single institutional experience. PLoS One. 2017;12(12):1‐8. doi: 10.1371/journal.pone.0189790 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228. Grunwald MH, Ben‐Dor D, Livni E, Halevy S. Acne Keloidalis‐like lesions on the scalp associated with antiepileptic drugs. Int J Dermatol. 1990;29(8):559‐561. doi: 10.1111/j.1365-4362.1990.tb03468.x [DOI] [PubMed] [Google Scholar]
  • 229. Piaserico S, Fortina AB, Cavallini F, Alaibac M. Acne keloidalis of the scalp in a renal transplant patient treated with cyclosporine. Acta Derm Venereol. 2009;89(3):312‐313. doi: 10.2340/00015555-0611 [DOI] [PubMed] [Google Scholar]
  • 230. Verma SB, Wollina U. Acne keloidalis nuchae: another cutaneous symptom of metabolic syndrome, truncal obesity, and impending/overt diabetes mellitus? Am J Clin Dermatol. 2010;11(6):433‐436. doi: 10.2165/11537000-000000000-00000 [DOI] [PubMed] [Google Scholar]
  • 231. Bellavista S, D'Antuono A, Gaspari V, Banzola N, Patrizi A. Acne keloidalis nuchae on herpes zoster scar in an HIV patient: isotopic response or not? G Ital Dermatol Venereol. 2012;147(2):223‐226. [PubMed] [Google Scholar]
  • 232. Pirmez R, Price VH, Cuzzi T, Trope BM. Acne keloidalis nuchae in renal transplant patients receiving tacrolimus and sirolimus. Australas J Dermatol. 2016;57(2):156‐157. doi: 10.1111/ajd.12271 [DOI] [PubMed] [Google Scholar]
  • 233. Winter H, Schissel D, Parry DAD, et al. An unusual Ala12Thr polymorphism in the 1A α‐helical segment of the companion layer‐specific keratin K6hf: evidence for a risk factor in the etiology of the common hair disorder pseudofolliculitis barbae. J Invest Dermatol. 2004;122(3):652‐657. doi: 10.1111/j.0022-202X.2004.22309.x [DOI] [PubMed] [Google Scholar]
  • 234. Otberg N, Wu W‐Y, McElwee KJ, Shapiro J. Diagnosis and management of primary cicatricial alopecia: part I. Skinmed. 2008;7(1):19‐26. Accessed October 18, 2019. http://www.ncbi.nlm.nih.gov/pubmed/18174797 [DOI] [PubMed] [Google Scholar]
  • 235. Sperling LC, Homoky C, Pratt L, Sau P. Acne keloidalis is a form of primary scarring alopecia. Arch Dermatol. 2000;136(4):479‐484. doi: 10.1001/archderm.136.4.479 [DOI] [PubMed] [Google Scholar]
  • 236. Herzberg AJ, Dinehart SM, Kerns BJ, Pollack SV, Keloidalis A. Transverse microscopy, immunohistochemistry, and electron microscopy. Am J Dermatopathol. 1990;12(2):109‐121. doi: 10.1097/00000372-199004000-00001 [DOI] [PubMed] [Google Scholar]
  • 237. Al‐Zaid T, Vanderweil S, Zembowicz A, Lyle S. Sebaceous gland loss and inflammation in scarring alopecia: a potential role in pathogenesis. J Am Acad Dermatol. 2011;65(3):597‐603. doi: 10.1016/j.jaad.2010.09.774 [DOI] [PubMed] [Google Scholar]
  • 238. Ao G, Ao A, Eu N, Jb O, Odusan O. Clinical, BIOCHEMICAL and morphologic features of acne KELOIDALIS IN a black population. Int J Dermatol. 1993;32(10):714‐716. doi: 10.1111/j.1365-4362.1993.tb02739.x [DOI] [PubMed] [Google Scholar]
  • 239. Togo S, Sugawara K, Tsuruta D. Acne keloidalis in an Asian female patient. Clin Case Reports. 2019;7(7):1412‐1414. doi: 10.1002/ccr3.2170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Silverberg NB, Durán‐McKinster C, Tay Y‐K. Pediatric Skin of Color | Nanette B. Silverberg | Springer; 2015. Accessed October 18, 2019 https://www.springer.com/gp/book/9781461466536 [Google Scholar]
  • 241. Uitto J. IL‐6 signaling pathway in keloids: a target for pharmacologic intervention? J Invest Dermatol. 2007;127(1):6‐8. doi: 10.1038/sj.jid.5700604 [DOI] [PubMed] [Google Scholar]

Associated Data

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

Data Availability Statement

Data sharing not applicable to this article as no datasets were generated or analysed during the current study.


Articles from Experimental Dermatology are provided here courtesy of Wiley

RESOURCES