Skip to main content
International Wound Journal logoLink to International Wound Journal
. 2019 Dec 20;17(2):405–418. doi: 10.1111/iwj.13286

Biological approaches for hypertrophic scars

Zhong Lingzhi 1, Li Meirong 1,2,, Fu Xiaobing 1,
PMCID: PMC7948781  PMID: 31860941

Abstract

Scar formation is usually the pathological consequence of skin trauma. And hypertrophic scars (HSs) frequently occur in people after being injured deeply. HSs are unusually considered as the result of tissue contraction and excessive extracellular matrix component deposition. Myofibroblasts, as the effector cells, mainly differentiated from fibroblasts, play the crucial role in the pathophysiology of HSs. A number of growth factors, inflammatory cytokines involved in the process of HS occurrence. Currently, with in‐depth exploration and clinical research of HSs, various creative and effective treatments budded. In here, we summarize the progress in the molecular mechanism of HSs, and review the available biotherapeutic methods for their pathophysiological characteristics. Additionally, we further prospected that the comprehensive therapy may be more suitable for HS treatment.

Keywords: wound healing, hypertrophic scar, prevention, biological strategies, MSC

1. INTRODUCTION

As the largest organ in the human body, skin senses the stress and tension outside and protects the systems inside as a barrier. When injured, it is necessary to repair fast to maintain the cutaneous integrity and to recover its function. The wound healing process is complicated, and deep injury often leaves a scar in the repaired site.1 Hypertrophic scars (HSs) as a kind of scar occur in 30% to 90% of patients suffered wound,2 such as surgeries, burns, traumas, and so on. And it mainly results from aberrations of wound healing process.

HS protrudes the normal skin surface with irregular shapes, uneven surface, congestive appearance, and solid and firm textures.3 Patients suffering from HSs often feel paresthetic including itchy and painful, which was more severe after increased ambient temperature, emotional arousal, or eating spicy food.4 Therefore, many patients bear the double burden of psychology and physiology. In the clinical, the origin of HSs was mainly from the excessive proliferation of dermis. It develops within 1 to 3 months after deep skin injury.5 During the normal healing process, the myofibroblasts undergo apoptosis in the remodelling stage and leave a rather acellular scar in the healed sites. While in HSs, the myofibroblasts persist and result in6, 7, 8 excessive extracellular matrix (ECM) component deposition and tissue contraction which can lead to severe disfigured and malfunction. A great number of studies have been proved that a variety of pro‐fibrotic cytokines and chemokines, such as TGF‐β1, TGF‐β2, VEGF, FGF, and CTGF, are associated with the anomaly in HSs.9, 10, 11, 12

Nevertheless, the exact mechanism and possible regulatory network still need to be exploration. Presently, several classical treatment methods have been developed. Because some treatment strategies have the feature with easy recurrence after treatment, or significant side effects, or poor efficacy, there is still a lack of effective treatment strategies.13, 14 Therefore, to better understand and study HS, we summarized the existing treatments based on the pathophysiological mechanism of HS and prospected the possible treatment principles of HS.

2. PATHOPHYSIOLOGY OF HYPERTROPHIC SCARS

The wound healing process involves the following phases, inflammation phase (the first 2 to 3 days after injury), proliferation phase (4 days to 2 weeks including ECM formation, angiogenesis, and reepithelialisation), and maturation phase (several months even lasting to one year).15 During proliferation phase, fibroblasts differentiate to myofibroblasts and express alpha‐smooth muscle actin (α‐SMA) which enables the wound to contract. HSs occurred within 1 month or so after wound injury,16 and turned into flatter scar within 1 to 2 years. The pathological features of HS are that collagen‐III bundles are oriented parallel to the epidermal surface with abundant nodules containing myofibroblasts expressing α‐SMA and extracellular collagen filaments.17 Besides, the occurrence of HSs is very complicated and often accompanied by the excessive inflammatory response, abnormal fibroblast‐myofibroblast transformation, non‐apoptosis of fibroblasts,8 aberrant keratinocyte‐fibroblast crosstalk, and disorder of ECM deposition.18 Because of the complexity of the pathophysiology of HS, it may be difficult to get a good curative effect on a ‘single point’ treatment. The strategy of ‘multi‐point’ combined comprehensive treatment may be a better strategy for the HS treatment.

3. CURRENT TREATMENT FOR HSs

The clinic, effective therapy should be set individually by clinicians after talking with patients on their concerns, needs, expectations, alternative treatments, and their outcomes. Also, the goals of treatment were to reduce scar volume, minimize subjective symptoms, such as pain and pruritus, and improve aesthetic and functional appearance. Mature clinical measures include first‐line therapy (silicone gel sheeting and topical onion extracts for small HSs) and second‐line therapy.19 The latter refers mainly to laser therapy as well as surgical excision in combination with intralesional corticosteroid injections postoperatively.20

4. THE CLASSICAL THERAPY OF LASER AND SURGERY WITH CORTICOSTEROIDS

The prevention and treatment of scarring after trauma represent a daunting challenge for a dermatologist. Presently, clinicians accustomed to using the following modalities to treat HSs after injury. Silicone gel is commonly used in the prevention or treatment of HSs.21 A recent study shows that the silicone gel reduces the expression of TGF‐β1and platelet‐derived growth factor 4 months after surgery for surgical scar,22 although the use of silicone gel remains conflicting on account of the upregulation of the basic fibroblast growth factor (b‐FGF) in the dermis of silicone gel sheet‐treated scars.23 Also, laser therapy is another common clinical mean for HSs treatment. It has been proved that long‐pulsed Nd: YAG laser and pulsed dye laser play therapeutic roles in shallowing the colour, reducing the thickness and tension of HSs, and relieving symptoms such as pain and pruritus.24 Surgical excision is also a highly recommended measure for surgeons. Surgery always combined with corticosteroid injection or radiotherapy in the clinic. Intralesional corticosteroids have shown to be useful in vivo by reducing the inflammatory process, decreasing collagen deposition, and fibroblasts proliferation.25 It was demonstrated that triamcinolone acetonide alone reduced at least half of scar occurrences with a recurrence rate of 9% to 53%.26 Also, there are other ways that have used in the prevention and treatment of HSs including 5‐Fluorouracil, bleomycin, interferon, imiquimod, and methotrexate.27 In spite of remitting the pathological scarring more or less in the short term by surgery above, it is easy to relapse as to the secondary injury to the wound. Besides, laser therapy combined with corticosteroids is at risk of leading to atrophy of the normal surrounding skin, fat and muscle, and even side effects such as osteoporosis and pain at the injection site.26 Therefore, these methods could not lay claim to an ideal standard.

5. TRADITIONAL PLANT‐BASED MEDICINE AND PRODUCTS

In recent years, traditional Chinese medicine (TCM) has been received more and more attention in the treatment of diseases. Some TCM, including extracts, plays a non‐negligible role in promoting skin regeneration and resisting skin aging. Presently, many plants have been proved effective as the potential therapy for HSs treatment in vivo and/or in vitro.

Quercetin, a flavonoid, has been demonstrated to reduce the HSs formation. It plays various biological functions including anti‐inflammatory, anti‐oxidant, and anti‐bacterial properties.28, 29 In vitro, quercetin has been shown to inhibit the collagen synthesis of myofibroblasts derived from keloids and HSs mainly through reducing their proliferation.30, 31 It was found that the Mederma, a derivative of quercetin improved and ordered collagen organization in the rabbit model, suggesting the potential effect on the treatment of HSs.32 Quercetin is derived from vegetables like onions, and studies show that the onion extract has the properties of anti‐inflammation and anti‐proliferation on several cancer cells such as glioma and oophoroma A‐2780 cells in vitro and vivo.33, 34, 35 In addition, quercetin and onion extract have both been shown to up‐regulate the expression of matrix metalloproteinase‐1 (MMP‐1) which plays an important role in ECM remodelling.36 These results imply its effective prevention in fibrosis formation.

Onion extract, the main ingredient of onion. It could reduce the scar height and improve the appearance of the scar after Pfannenstiel's incision in the study of caesarean for Asian women.37 In another study of some children undergoing a median sternotomy, the incidences of HSs were significantly decreased by the application of onion extract.38 Furthermore, the combination of silicone derivative and onion extract relieved the itch and pain effectively more than the former alone, and the Vancouver Scar score was improved obviously.39

Resveratrol, a natural plant polyphenol and phytoestrogen derived from grape and peanuts,40, 41 is proved to have many beneficial functions, including anti‐inflammatory and anti‐oxidant properties.42, 43, 44 In fibroblasts derived from HSs, resveratrol reduced their proliferation through cell cycle arrest at G1 and induce their apoptosis.45 Besides, resveratrol has been shown to decrease the expression of collagen I and III, and reduce hydroxyproline levels. Meanwhile, resveratrol inhibited the proliferation and induced apoptosis of keloid derived fibroblasts. In an animal model, it was demonstrated that resveratrol decreased the expression of TGF‐β and α‐SMA in fibroblasts derived from scar, but has no effect on fibroblasts under physiological conditions.46

Epigallocatechin gallate (EGCG), as major catechin in green tea, has many biological properties in preventing fibrosis in various organs.47 Connective tissue growth factor (CTGF) is an early response gene and belongs to a new family of cysteine‐rich growth factors. It could promote the proliferation and migration of fibroblasts and plays a key role in tissue fibrosis.48

It has been shown that EGCG reduced the expression of CTGF and pro‐fibrotic molecules such as TGF‐β1.49 Inhibition of TGF‐β1 by EGCG could result in a reduction of ECM synthesis and HSs formation.50 And EGCG relieved cardiac fibrosis through low expression of CTGF, suggesting the therapeutic potential on the prevention of multiple fibrotic diseases including scaring.51

Oleanolic acid (OA) is the compound of triterpenoid which has the properties of anti‐inflammatory and anti‐tumour effects.52, 53 It was found that OA reduced the expression of TGF‐β1, induced the apoptosis of HS fibroblasts, decreased collagen synthesis, and alleviated deposition of collagen‐I/III.54, 55 MMP‐1 plays a crucial role in the degradation of collagen‐I and III in scaring56 and MMP‐2 can degrade collagen‐I.57 Studies show that after OA treatment, the level of MMP‐1 and MMP‐2 was increased in the rabbit HSs model and the HS was suppressed significantly.58, 59 Additionally, compared with the higher collagen‐III in normal skin, there is a high ratio of collagen‐I to III in HSs, which could be reversed by OA.

Curcumin, as the antioxidant, has been proved to promote wound closure and promote wound healing in the rat model. After treating with curcumin, the wound shows increased maturation fibrin and collagen, thus to improve all the phases of wound repair.60 Besides, it was reported that curcumin could induce apoptosis of several cells including fibroblasts after wound healing.61 Furthermore, a high dose of curcumin could reduce and prevent scarring through reactive oxygen species‐mediated heme oxygenase pathway.62

The TCM compound therapies including the above were commonly used in clinical, and the patients have relieved more or less to some extent. However, as the classical treatment, it still has the shortcomings of short curative effect, side effects, and recurrence of HS. Therefore, the development of other promising treatments with better effects and fewer side effects is urgent. Biotherapy has been favoured by clinicians and researchers in recent years. And biotherapy is used to treat various diseases by the way of biological macromolecules,63 antibodies,64 and even cellular methods.65 Meanwhile, the strategy of biotherapy is also validated in the prevention and treating in HSs in vivo and in vitro.66, 67, 68 Based on the unique physiological characteristics of HS, many targeted biological therapies have emerged.

6. BIOTHERAPY FOR INFLAMMATION IN HSs

Wounds in early mammalian embryos evolved into scar‐free wound healing compared with scar‐forming wound healing in adults.69 The development of HSs is associated with an excessive inflammatory response,7, 70 while the foetal wounds usually show fewer inflammatory cells, a shorter period of inflammation and less inflammatory factors.71, 72 Therefore, attempts to interfere with inflammation specific immune cells, or to block HS‐related critical inflammatory signalling pathways, may effectively prevent the occurrence of HS.

Neutrophils are the most abundant inflammatory cells in the early stage of wound healing, which mainly play an anti‐infection role in local sites, and the number of neutrophils has no significant influence on the wound healing rate.73 Besides, neutrophils also play the role in recruiting other inflammatory cells, such as mononuclear derived macrophages which phagocytizes fibrin clots to remove tissue debris and dead cells.74, 75 Macrophages have been considered the indispensable immunological cells in wound repair, who involve in phagocytosis, antigen‐presentation, and the secretion of cytokines and chemokines in wound sites.76, 77 Macrophages can be divided into M1 (classically activated) and M2 (alternatively activated) populations in the process of wound healing, and exhibit different functions.78 This characteristic determines that macrophages play an important role not only in the inflammatory phase but also in the proliferation phase. Generally, macrophages with M1 phenotype have antimicrobial properties, and with the M2 phenotype possess the properties of stimulating the production of collagen, angiogenesis, reepithelialisation, regeneration of wounds, and the anti‐fibrosis.79, 80 In the inflammation stage, M1 is required to defend against pathogens and clear senescent cells, and after that cells appear apoptosis. Minor macrophages undergo a transition from M1 to M2, modulating the wound healing. Experimental results showed that mice lacking macrophages showed delayed epithelial regeneration, collagen deposition and reduced angiogenesis.76, 81 However, M2 is an important source of TGF‐β1 which plays multiple roles in different phases of wound healing. M2 persisted activation lead to excessive TGF‐β1 production, which would further induce myofibroblast proliferation, abundant ECM deposition and the occurrence of fibrosis.82 Therefore, the balance of M1 and M2 macrophage population is an important impact on the development of a scar. However, there are no studies aimed at mediating the different phenotypes of macrophages to achieve the anti‐scar repair. Studies have shown that promoting M1 to M2 phenotype transition can promote chronic wound healing.83

Another strategy is targeting the regulation of inflammatory mediators. Therapies targeting inflammatory cytokines are used to lighten the HSs. Increased Th2 and Th3 response cytokines including interleukin‐2 (IL‐2), IL‐4, and IL‐10 have been found in circulating lymphocytes of fibrotic conditions.84 Also, IL‐4 and IL‐13 have been proved to activate myofibroblasts through the IL‐4R pathway.85, 86 And inhibitors of IL‐4 could reduce dermal fibrosis in mice with scleroderma.87 Besides, inhibition of IL4 and IL‐10 simultaneously mediated pathway reduced fibrosis of multi‐tissues.88 Of course, the biological effects of different inflammatory factors are different. For example, proinflammatory cytokine IL‐6 and IL‐8 enhanced scarring and the anti‐inflammatory cytokine IL‐10 has the opposite effect.89 Therefore, upregulating anti‐inflammatory cytokine and reducing pro‐inflammatory cytokine could inhibit HSs formation, as found in some TCM, such as bupleurum90, 91 and pirfenidone ointment.92 Recently, a breakthrough study showed that adipocytes are regenerated from myofibroblasts by activating the adipocyte transcription factors during wound healing. The consequential effect was triggered by BMP signalling from the actively growing hair follicles, and the findings fortify the importance of BMPs in the trans‐differentiation of myofibroblasts and adipocytes.93 From another perspective, we should further pay attention to the temporal and spatial dynamic changes of BMP in the process of myofibroblasts‐adipocytes transformation, to better regulate the number of myofibroblasts by BMP to reduce fibrosis.

Compared with acute inflammatory reactions, the pathological fibrosis representatively results from the chronic inflammatory reactions. Under chronic inflammatory condition, the injured area presents a complex traumatic microenvironment such as inflammation, tissue dissolution, and tissue repair.86 It was reported that mitochondrial reactive oxygen species (ROS) participated in inflammatory reaction of wound healing. And fibroblast dysfunction is linked to the overproduction of free ROS.94, 95 Application of antioxidant or drugs with antioxidant properties could relieve the microenvironment of chronic inflammation, accelerate wound healing and alleviate scar formation on wounds.96, 97 Similarly, classical traditional medicine, arsenic trioxide, could effectively inhibit the formation of rabbit ear scars through the overexpression of antioxidant genes.98

7. BIOTHERAPY TARGETING FIBROBLASTS AND MYOFIBROBLASTS IN HSs

In the proliferation stage, a great number of fibroblasts differentiate into myofibroblasts, which is an important process in wound repair. Meanwhile, there are heterogeneous population cells of derived fibroblasts.99 In the haemostatic phase of wound healing, collagen ECM replaced the provisional fibrin clot, and this change requires for fibroblastic cells with sufficient ability of migration from adjacent tissues and circulation to deposition of the ECM.100 Firstly, the fibroblastic cells transform into a ‘proto‐myofibroblasts’ phenotype101 with low contractile capacity and high migration ability. Then, the proto‐myofibroblasts evolve into mature myofibroblasts with contractile features and contribute to producing mechano‐resistant scars. The two phenotypes of myofibroblasts not only enhance wound contraction101 but also promote the formation of abnormal scarring.102, 103

In the case of HSs, excessive fibroblasts are transformed into myofibroblasts in the proliferation phase. Massive and persistent myofibroblasts will lead to tissue deformation by contracture manifesting as HSs104 and scleroderma105 in the skin. Fibrillar collagen plays a crucial role in the elasticity and strength of the skin.106 While in HSs, there is excess deposition of collagen‐I and collagen‐III,107 especially the collagen‐I which increases in both early and final remodelling stages of wound healing.108 After repaired and regained tissue homeostasis, the myofibroblasts undergo apoptosis, leaving a rather acellular scar.109 However, in HSs, myofibroblasts persist even in the final remodelling stage, resulting in bulky, contracted scar.110 Therefore, myofibroblast has been considered the main mediator of non‐healing wounds and excessive repair (HSs). Insufficient myofibroblasts are associated with chronic wounds, while excessive myofibroblasts are associated with scarring. Therefore, there are two available approaches to control the number of myofibroblasts in wound sits.

One way is to properly control the myofibroblast formation. Reducing the main derived fibroblast is an important way. Inhibitors of histone deacetylase have been reported play roles in regulation of fibrotic gene expression.111 Suppressing histone deacetylase limits the proliferation of lung fibroblasts and inhibits fibrosis‐related gene transcription, leading to the anti‐fibrotic ending.112 Trichostatin A, as the potent molecule, it could inhibit the proliferation of fibroblasts in vivo or in vitro.113 And also, it could decrease the deposition of ECM in bleomycin‐induced skin fibrosis of the mouse model.114 TGF‐β1 signalling pathway is the most important regulatory signal in fibroblast differentiation. It was found that TGF‐β1 treatment could repress the expression of peroxisome proliferator‐activated receptors in scleroderma, indicating the anti‐fibrosis effect of TGF‐β1 inhibitor.115 Recently, the substance extracted from Chinese herb Arnebiae shikonin has been shown to attenuate the TGF‐β1 expression and suppress myofibroblasts formation through modulating SMAD/ERK pathway.116 The other way is to enhance the apoptosis of myofibroblasts or promote myofibroblast‐to‐non‐fibroblastic cell conversion. As aspect to be insensitive to apoptotic signalling and expressing antiapoptotic‐related molecules in myofibroblasts,117 two kinases have been suggested to control myofibroblasts apoptosis, including phosphatidylinositol 3‐kinase (PI3K)‐AKT and focal adhesion kinase signalling.118, 119 Small molecule inhibitors targeting these protein kinases were prove to be effective anti‐fibrotic therapeutic strategies in pulmonary fibrosis,120 suggesting another promising strategy in skin fibrosis treatment.

8. BIOTHERAPY TARGETING EPITHELIAL‐MESENCHYMAL TRANSITION PROCESS IN HSs

Epithelial‐mesenchymal transition (EMT) is involved in both embryonic development and wound repair. Response to skin injury, epidermal keratinocytes undergo EMT and became to motile cells with the mesenchymal phenotype to migrate across to wound bed.121 The mesenchymal cells, especially contractile myofibroblast is necessary to restore tissue integrity in normal wound healing.122 And in process of remodelling, myofibroblasts disappear once reepithelialisation completes.68, 123 But under pathological conditions, myofibroblasts persisted instead of undergoing dedifferentiation. Indeed, EMT is essential for proper reepithelialisation and ECM deposition, but the uncontrolled sustaining transition from epithelial cells to myofibroblasts might result in HSs. Therefore, targeting both fibroblasts and keratinocytes populations were the novel therapies for scarring.124 It was shown that extracorporeal shock wave therapy was responsible for the anti‐scarring by suppressing EMT in the post‐burn scars.125 As such, focus on the dysregulation of injury‐triggered EMT is proved to contribute to HSs treatment. Keratin and vimentin are characteristic markers of epithelial cells and mesenchymal cells, respectively, and can serve as indicative markers of EMT processes.126 Several studies proved that down‐expression of vimentin and over‐expression of Keratin can inhibit the process of EMT,127, 128, 129 prompting another possible way to suppress scars.

Uncontrolled persist EMT and excessive proliferation of (myo)fibroblasts cause the deposition of ECM. ECM plays a critical role in wound healing and scar formation which was produced mainly by fibroblasts and myofibroblasts. The breakdown of fibrillar collagen type I, II, and III were mediated by MMP‐1. Fibroblasts of HSs appear to have a low MMP‐1 (collagenase) activity.130 It was demonstrated that MMP‐2 effect denatured collagen in the late stage of wound healing, while MMP‐9 was involved in the early stage of wound healing degrading collagen type IV and V, fibronectin, and elastin.131, 132 MMP transcription is induced partly by TGF‐β and interleukin‐I.133 MMP expression is high in injuring skin compared with the intact skin.134 Additionally, it was determined that myofibroblasts were resistant to breakdown by collagenase D and MMP‐2. It was further found that fibroblasts derived from HSs over‐expressed tissue transglutaminase which could inhibit the apoptosis of myofibroblasts.135 It was reported that inhibiting molecular chaperone, FK506‐binding protein 10 (FKBP10) could reduce ECM components and attenuate HS formation through TGF‐β/Smad signalling pathway.136 Y‐box‐binding protein (YB)‐1, a suppressor of Collagen‐1A1 resulted in repression of Collagen‐1 and anti‐fibrosis in cardiac, suggesting a novel therapeutic target for pathological scar point to ECM.137

9. BIOTHERAPY AIMED AT MAIN CYTOKINES AND PROTEIN INVOLVED IN HSs

The wound repair process requires a variety of signal pathways to form a regulatory network that interacts with each other. The future of HSs management may lie in targeting specific molecular pathways. A large number of studies have confirmed inflammation and the abnormality of multiple signalling pathways during the healing of pathological scars, which leads to pathological repair.138

There are three isotypes of TGF‐β, and all of them are associated with wound healing.139, 140 The HSs formation is associated with the overexpression of TGF‐β1/2, and depressing the effect of TGF‐β1/2 reduced fibrosis and scarring significantly wound model in vivo.141 However, scarless healing is associated with high levels of TGF‐β3.142, 143 In particular, scarless wound healing in foetal is always with the high ratio of TGF‐β3/TGF‐β1 which hint the ratio between different TGF‐β types determines the effect of TGF‐β signalling pathway.144 TGF‐β/Smad signalling has a pivotal role in scar healing through binding to dimeric TGF‐β receptor complexes.2, 145 Upon activation, this receptor complex phosphorylated Smad2/3 proteins, which form corresponding dimmers with Smad4 and translocate into the nucleus to initiate downstream target genes including collagen‐I and III.146 The extent of wound fibrosis is related to the activity of TGF‐β1 to a large extent.147 In transgenic animals, it has been found that overexpression of the constitutively TGF‐β1 receptor leads to spontaneous fibrosis of skin.148 Activation of TGF‐β1 assembles and TGF‐β‐receptor complex activated the Smad2/3 and JNK signalling in fibroblasts and exhibited the biological behaviour of myofibroblasts.149 On the contrary, silencing TGF‐β1 resulted in lower collagen synthesis and alleviative scarring which suggested it was a potential therapeutic target for limiting scar formation.150 High levels of TGF‐β1 may stimulate the activation of detrimental myofibroblasts, but blocking TGF‐β1 completely shows spontaneous skin inflammation and defective vasculogenesis in TGF‐β1 knockout mouse,151 which indicated that maintaining a certain level of TGF‐β1 in wounds was essential. What we need to do now is to pay attention to the expression level of TGF‐β1 in scar‐free repair wounds, and to the temporal and spatial dynamics of TGF‐β1 to control the homeostasis of reepithelialisation, vascularization, and inflammation.152, 153 In fact, there are various means of regulating the TGF‐β1 signalling pathway, including antibodies, antagonists, and even miRNA means.154, 155, 156, 157, 158 However, using antagonists of TGF‐β1 receptors or TGF‐β1 specific antibodies could merely reach clinical request due to the safety accidents.159, 160 Hence, targeting to the modulation of TGF‐β1 or its downstream pathway may be more feasible.

As to the downregulation of TGF‐β signalling, blocking the downstream molecule, Smad3, has been attempted.114 Smad3 shows a different effect on wound healing.141 It was reported that Smad3 protein‐induced HSs formation accordingly through activating the WNT pathway.145 Studies showed that counteracting Smad3 signalling improved wound healing with inhibition of scarring. The rate of reepithelialisation is accelerated, but the area of the wounds and the number of myofibroblasts are significantly reduced in Smad3‐null mice compared with wild‐type mice after the exposure of irradiation.141 It is thought to prevent Smad2/3‐receptor interaction and phosphorylation after inhibiting fibrosis and preventing HSs formation.161 A great body of studies has shown that the inhibitors of Smad3, for instance, halofuginone, quercetin, trichostatin A, and paclitaxel, could suppress the fibrosis of skin through inhibiting the phosphorylation of Smad2/3 and the formation of the Smad2/3/4 complex.30, 162, 163

Another way referring to the upregulation of the TGF‐β family inhibitor, such as Smad7, has been tried.164 Smad7, as a member of Smad protein, downregulated in HSs.165 Studies have indicated that overexpression of Smad7 prevents the collagen gel contraction and inhibits collagen‐I and α‐SMA expression in fibroblasts derived from normal skin and scar.166 Asiaticoside was isolated from the leaves of Centella Asiatic possessed inhibition properties to reduce scar formation through promoting the expression of Smad 7 instead of other members of the Smad family.167

The sequential transition of three post‐traumatic repair phases mentioned above is pivotal in the HSs formation. In the fibrotic and antifibrotic events of wound healing, the imbalance between profibrotic growth factors and antifibrotic factors results in abnormal deposition of ECM showing HSs.168 To balance the two processes, an agent which binded to several targets is more refined. One typical example is decorin, a small leucine‐rich proteoglycan,169 which exists in the interstitial matrix of the dermis and combines with collagen fibrils preferentially,170 and then setting their assembly.171 And the normal scar is replaced by HSs with increased production of collagen‐I/III, fibronectin, laminin, and decreased expression of hyaluronic acid and decorin.15, 172 Decorin, as a proteoglycan in the dermal ECM, regulates TGF‐β1 to influence collagen fibrillogenesis and diminish scarring.173 Studies showed decorin inhibited TGF‐β induced contraction in HS‐derived fibroblast‐populated collagen lattice.174 Therefore, balance is a very important point to adjust the outcome of wound healing, especially the balance between the pro‐ and anti‐fibrotic processes in time and space.

10. CONCLUSION AND PROSPECT

We summarized the feasibility of traditional laser, small molecule of TCM, and biotherapy in the clinical treatment of HSs. Systematically, biotherapy has its own characteristics compared with traditional therapy. We can conclude that biotherapy is based on one entry point, maybe a cell (inflammatory cell or fibroblasts), a factor (inflammatory factors or cytokines), or a response (trans‐differentiation and dedifferentiation), and only one step change can significantly alter the formation and prognosis of HSs. So as to biotherapy, it is relatively more efficient than traditional treatments above.

About various schemes above, most treatments are carried out merely after scar formation has already taken place, targeting reversal fibrosis phenotype and restore normal ECM composition and structure. However, adjusting inflammatory response and proliferative phase in fibrosis pathological changes are relatively early. It prefers to prevention and early treatment, and is more attractive in anti‐fibrosis strategies. So as far as current techniques are concerned, we could infer that early diagnosis and advanced prevention may be more advisable.

As we all know, prevention is the most significant section of the HSs therapy, including following Langer's lines that correspond to the direction of collagen fibres and paralleling to the orientation of underlying muscle fibres in elective surgery.175 Generally speaking, scarring and other fibrotic diseases have a long process which is often ignored. Unfortunately, it was only when the fibrosis progresses to obvious clinical symptoms, such as function abnormalities, that would attract the attention of the patients.176 And at this stage, fibrosis may be serious and current treatment could merely delay its process. Therefore, for most or large areas of scarring patients, the significance of prevention is greater than that of the treatment itself. Studies in vitro and in vivo have shown that there are many precautionary strategies against scarring.177, 178 Furthermore, the variation and outcome of the same measures in patients undergoing clinical trials are huge, and this leads to comparing problems. However, the same treatments are highly variable in the results obtained in different patients, which leads to comparing problems. Additionally, there remains no gold standard in the prevention and treatment of pathological scars due to the limitation of effective assess for anti‐scarring therapeutics, HSs prevention, and scar models.179 Because of this, there is a large amount of clinical data, and it is still difficult to compare the obtained data horizontally. Therefore, the prevention and clinical treatment of HSs still have a long way to go.

Additionally, although numerous scars are caused by local trauma,180 many patients also manifest the formation of multiple scars, implying the systemic disease of HSs. In this case, we could hardly achieve desired outcome by altering separate genes. Starting from one entry point, and then covering whole body like a net to achieve earlier and extensive therapeutic effect needs to be further considered. Currently, HSs have been considered as an autoimmune disease.181, 182 Master cells (MCs), as an important of immune cells, participated in innate/acquired immunity and blood coagulation, which contributed to all three stages of wound healing.164, 165 In MC‐deficient mice, it showed less scarring with more follicles than MC‐sufficient mice after scald burns, and the significant difference was due to the activities of mouse mast cell proteases 4/5 (MCP‐4/5).166 In foetal mice, the wound was healed without scars at embryonic day 15 (E15), compared scar healing at embryonic day 18 (E18).183, 184 It was shown that there were few MCs in the dermis of mice on E15 detected with immunohistochemistry. Injection of the lysate of MCs into mice at E15 resulted in scar formation that was similar to which was observed in E18 embryos. On the contrary, wounds of MC‐deficient mice revealed less scarring than MC‐deficient embryos at the same time.167 Therefore, it was suggested that inhibition of the mediators derived from activated MCs could prevent the formation of pathologic scarring. Regarding immune cells, it was reported that specialized dendritic cells, such as Langerhans cells (LCs), reside in epidermis play the key role in the forming of HSs.185 In aberrant wound healing, LCs stimulated naive T cell responses, and identify the triggering receptor expressed on myeloid (TREM)‐1, a member of the Ig immunoregulatory receptor family, infiltrating hypoxic areas of active HSs, pointing to the pathogenic role in wound repair disorders.186 Besides, it has been found that there is a correlation of NK cell, T‐cell, and scarring.187 For example, NK cells inhibit the progression of liver fibrosis through targeting and clearing the hepatic astrocytes directly which secrete ECM, suggesting an important role in antifibrosis.188 Also, we mentioned in the previous section that macrophages of different phenotypes play different roles in the scar formation. These evidences suggest that immunomodulatory therapy is essential for HS.

In recent years, mesenchymal stem cells (MSCs) have been considered to possess the immunomodulatory capacity and play a therapeutic role in a variety of immune diseases.189, 190 In wound healing, stem cells could not only promote skin repair but also reduce scar formation and resist pathological scarring.191 Several studies demonstrated that MSCs suppressed the formation of HSs in several ways. The first is its immunomodulatory effect.192 MSCs prevented the formation of scar by regulated inflammation cells and pro‐inflammatory mediators. The mechanism of modulating inflammatory reaction of MSC was mainly through reducing mast cell degranulation, suppressing T‐cell proliferation, reducing NK cell function, activating macrophages, recruiting neutrophils, and antibacterial actions.193, 194, 195, 196 In atopic dermatitis, hMSCs were demonstrated the therapeutic functions in response to Th2 cytokines to mitigate dermatitis through suppressing MCs degranulation.197 In vitro, MSC was able to diminish human keratocyte differentiation into α‐SMA+ limbal myofibroblast, and reduce the release of neutrophil extracellular traps, thus to promote corneal wound healing in an anti‐inflammatory and anti‐fibrotic way.198

Second, MSC can act on myofibroblasts, the main effector cells in HS formation. It was proved that bone marrow‐derived MSCs inhibit myofibroblasts proliferation, migration, ECM synthesis, and scar formation through paracrine signalling.199 Moreover, not only stem cells themselves, the conditioned medium of MSCs and exosomes can be used as the treatment of anti‐scarring. The exosomes secreted by human adipose mesenchymal stem cells could benefit wound healing and promote scarless cutaneous repair by regulating ECM remodelling.200 At the same time, MSC inhibited the bioactivities of fibroblasts in the HSs,201 inhibited the proliferation and the differentiation of fibroblasts to myofibroblasts, and further suppressed the scar formation. Finally, as the multipotent cells, MSC could migrate to local trauma to prevent excessive matrix deposition by fibroblasts, forming new microenvironment to promote healing and alleviate scar.202 More importantly, as a promising therapy, stem cells can be modified. And we could use physical chemistry and even genes to transform the stem cells into the therapeutic missiles we need to treat HSs. Therefore, MSC treatment may become a new generation of HS treatment strategies.

In sum, the occurrence of HSs is complex, and it is still a ‘black box’ that needs to be further explored, which further obstacle the development of effective therapeutic strategies. With a better understanding of pathophysiological mechanisms of HSs and the development of the high‐throughput screening technologies, future studies in HSs will overcome current hurdles and develop promising treatments. And this may be solved with the joint efforts by multidisciplinary experts, including pathophysiologists, immunologists, pharmacologists, and so on. However, at present, due to the complexity of the development of HSs, a single treatment strategy is difficult to achieve the desired clinical effect and the simultaneous or sequential use of multiple methods, that is, the comprehensive treatment method may be preferable.

CONFLICT OF INTEREST

The authors declare that there is no conflict of interest regarding the publication of this paper.

Lingzhi Z, Meirong L, Xiaobing F. Biological approaches for hypertrophic scars. Int Wound J. 2020;17:405–418. 10.1111/iwj.13286

Funding information Natinal Key Research Development Plan, Grant/Award Numbers: 2017YFC1103300, 2017YFC1104701; National Nature Science Foundation of China, Grant/Award Numbers: 81721092, 81830064, 81901973, 81971841; the General Hospital of PLA Medical Big Data R&D Project, Grant/Award Number: MBD2018030; the Military Logistics Research Key Project, Grant/Award Number: AWS17J005; the National S&T Resource Sharing service platform Project of China, Grant/Award Number: YCZYPT[2018]07

Contributor Information

Li Meirong, Email: meirong811225@126.com.

Fu Xiaobing, Email: fuxiaobing@vip.sina.com.

REFERENCES

  • 1. Coentro JQ, Pugliese E, Hanley G, Raghunath M, Zeugolis DI. Current and upcoming therapies to modulate skin scarring and fibrosis. Adv Drug Deliv Rev. 2019;146:37‐59. [DOI] [PubMed] [Google Scholar]
  • 2. Arno AI, Gauglitz GG, Barret JP, Jeschke MG. Up‐to‐date approach to manage keloids and hypertrophic scars: a useful guide. Burns. 2014;40(7):1255‐1266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Miletta N, Siwy K, Hivnor C, et al. Fractional ablative laser therapy is an effective treatment for hypertrophic burn scars: a prospective study of objective and subjective outcomes. Ann Surg. 2019. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 4. Bijlard E, Kouwenberg CA, Timman R, Hovius SE, Busschbach JJ, Mureau MA. Burden of keloid disease: a cross‐sectional health‐related quality of life assessment. Acta Derm Venereol. 2017;97(2):225‐229. [DOI] [PubMed] [Google Scholar]
  • 5. Wolfram D, Tzankov A, Pulzl P, Piza‐Katzer H. Hypertrophic scars and keloids—a review of their pathophysiology, risk factors, and therapeutic management. Dermatol Surg. 2009;35(2):171‐181. [DOI] [PubMed] [Google Scholar]
  • 6. Nedelec B, Shankowsky H, Scott PG, Ghahary A, Tredget EE. Myofibroblasts and apoptosis in human hypertrophic scars: the effect of interferon‐alpha2b. Surgery. 2001;130(5):798‐808. [DOI] [PubMed] [Google Scholar]
  • 7. Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature. 2008;453(7193):314‐321. [DOI] [PubMed] [Google Scholar]
  • 8. Cheon SS, Cheah AY, Turley S, et al. beta‐Catenin stabilization dysregulates mesenchymal cell proliferation, motility, and invasiveness and causes aggressive fibromatosis and hyperplastic cutaneous wounds. Proc Natl Acad Sci USA. 2002;99(10):6973‐6978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Samuel CS, Royce SG, Hewitson TD, Denton KM, Cooney TE, Bennett RG. Anti‐fibrotic actions of relaxin. Br J Pharmacol. 2017;174(10):962‐976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Simonetti O, Lucarini G, Orlando F, et al. Role of daptomycin on burn wound healing in an animal methicillin‐resistant staphylococcus aureus infection model. Antimicrob Agents Chemother. 2017;61(9):606‐617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Hwang J, Chang C, Kim JH, et al. Development of cell‐penetrating asymmetric interfering rna targeting connective tissue growth factor. J Invest Dermatol. 2016;136(11):2305‐2313. [DOI] [PubMed] [Google Scholar]
  • 12. Wang L, Yang J, Ran B, et al. Small molecular tgf‐beta1‐inhibitor‐loaded electrospun fibrous scaffolds for preventing hypertrophic scars. ACS Appl Mater Interfaces. 2017;9(38):32545‐32553. [DOI] [PubMed] [Google Scholar]
  • 13. Bu W, Fang F, Zhang M, Chen J. Combination of 5‐ALA photodynamic therapy, surgery and superficial X‐ray for the treatment of keloid. Photodermatol Photoimmunol Photomed. 2019. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 14. Sommer S, Sheehan‐Dare RA. Atrophie blanche‐like scarring after pulsed dye laser treatment. J Am Acad Dermatol. 1999;41(1):100‐102. [DOI] [PubMed] [Google Scholar]
  • 15. Xue M, Jackson CJ. extracellular matrix reorganization during wound healing and its impact on abnormal scarring. Adv Wound Care (New Rochelle). 2015;4(3):119‐136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Tziotzios C, Profyris C, Sterling J. Cutaneous scarring: pathophysiology, molecular mechanisms, and scar reduction therapeutics Part II. Strategies to reduce scar formation after dermatologic procedures. J Am Acad Dermatol. 2012;66(1):13‐24. quiz 5‐6. [DOI] [PubMed] [Google Scholar]
  • 17. Martin P, Nunan R. Cellular and molecular mechanisms of repair in acute and chronic wound healing. Br J Dermatol. 2015;173(2):370‐378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Hong KM, Belperio JA, Keane MP, Burdick MD, Strieter RM. Differentiation of human circulating fibrocytes as mediated by transforming growth factor‐beta and peroxisome proliferator‐activated receptor gamma. J Biol Chem. 2007;282(31):22910‐22920. [DOI] [PubMed] [Google Scholar]
  • 19. Kelemen O, Kollar L, Menyhei G. A comparative clinical study of the treatment of hypertrophic scars with either intralaesional steroid or silicone gel sheeting. Magy Seb. 2007;60(6):297‐300. [DOI] [PubMed] [Google Scholar]
  • 20. Atiyeh BS. Nonsurgical management of hypertrophic scars: evidence‐based therapies, standard practices, and emerging methods. Aesthet Plast Surg. 2007;31(5):468‐492. discussion 93‐4. [DOI] [PubMed] [Google Scholar]
  • 21. Reno F, Grazianetti P, Stella M, Magliacani G, Pezzuto C, Cannas M. Release and activation of matrix metalloproteinase‐9 during in vitro mechanical compression in hypertrophic scars. Arch Dermatol. 2002;138(4):475‐478. [DOI] [PubMed] [Google Scholar]
  • 22. Choi J, Lee EH, Park SW, Chang H. Regulation of transforming growth factor beta1, platelet‐derived growth factor, and basic fibroblast growth factor by silicone gel sheeting in early‐stage scarring. Arch Plast Surg. 2015;42(1):20‐27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. O'Brien L, Jones DJ. Silicone gel sheeting for preventing and treating hypertrophic and keloid scars. Cochrane Database Syst Rev. 2013;9:CD003826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Amini‐Nik S, Yousuf Y, Jeschke MG. Scar management in burn injuries using drug delivery and molecular signaling: current treatments and future directions. Adv Drug Deliv Rev. 2018;123:135‐154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Roques C, Teot L. The use of corticosteroids to treat keloids: a review. Int J Low Extrem Wounds. 2008;7(3):137‐145. [DOI] [PubMed] [Google Scholar]
  • 26. Gauglitz GG. Management of keloids and hypertrophic scars: current and emerging options. Clin Cosmet Investig Dermatol. 2013;6:103‐114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Rabello FB, Souza CD, Farina Junior JA. Update on hypertrophic scar treatment. Clinics (Sao Paulo). 2014;69(8):565‐573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Williams RJ, Spencer JP, Rice‐Evans C. Flavonoids: antioxidants or signalling molecules? Free Radic Biol Med. 2004;36(7):838‐849. [DOI] [PubMed] [Google Scholar]
  • 29. Prior RL. Fruits and vegetables in the prevention of cellular oxidative damage. Am J Clin Nutr. 2003;78(3 Suppl):570S‐578S. [DOI] [PubMed] [Google Scholar]
  • 30. Phan TT, Lim IJ, Chan SY, Tan EK, Lee ST, Longaker MT. Suppression of transforming growth factor beta/smad signaling in keloid‐derived fibroblasts by quercetin: implications for the treatment of excessive scars. J Trauma. 2004;57(5):1032‐1037. [DOI] [PubMed] [Google Scholar]
  • 31. Long X, Zeng X, Zhang FQ, Wang XJ. Influence of quercetin and x‐ray on collagen synthesis of cultured human keloid‐derived fibroblasts. Chin Med Sci J. 2006;21(3):179‐183. [PubMed] [Google Scholar]
  • 32. Saulis AS, Mogford JH, Mustoe TA. Effect of Mederma on hypertrophic scarring in the rabbit ear model. Plast Reconstr Surg. 2002;110(1):177‐183. discussion 84‐6. [DOI] [PubMed] [Google Scholar]
  • 33. Li J, Tang C, Li L, Li R, Fan Y. Quercetin sensitizes glioblastoma to t‐AUCB by dual inhibition of Hsp27 and COX‐2 in vitro and in vivo. J Exp Clin Cancer Res. 2016;35:61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Niedzwiecki A, Roomi MW, Kalinovsky T, Rath M. Anticancer efficacy of polyphenols and their combinations. Nutrients. 2016;8(9):552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Augusti KT. Therapeutic values of onion (Allium cepa L.) and garlic (Allium sativum L.). Indian J Exp Biol. 1996;34(7):634‐640. [PubMed] [Google Scholar]
  • 36. Cho JW, Cho SY, Lee SR, Lee KS. Onion extract and quercetin induce matrix metalloproteinase‐1 in vitro and in vivo. Int J Mol Med. 2010;25(3):347‐352. [PubMed] [Google Scholar]
  • 37. Chanprapaph K, Tanrattanakorn S, Wattanakrai P, Wongkitisophon P, Vachiramon V. Effectiveness of onion extract gel on surgical scars in asians. Dermatol Res Prac. 2012;2012:212945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Wananukul S, Chatpreodprai S, Peongsujarit D, Lertsapcharoen P. A prospective placebo‐controlled study on the efficacy of onion extract in silicone derivative gel for the prevention of hypertrophic scar and keloid in median sternotomy wound in pediatric patients. J Med Assoc Thail. 2013;96(11):1428‐1433. [PubMed] [Google Scholar]
  • 39. Jenwitheesuk K, Surakunprapha P, Kuptarnond C, Prathanee S, Intanoo W. Role of silicone derivative plus onion extract gel in presternal hypertrophic scar protection: a prospective randomized, double blinded, controlled trial. Int Wound J. 2012;9(4):397‐402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. George J, Singh M, Srivastava AK, et al. Resveratrol and black tea polyphenol combination synergistically suppress mouse skin tumors growth by inhibition of activated MAPKs and p53. PLoS One. 2011;6(8):e23395. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 41. Hiroto Y, Tadokoro K, Tsuda T, et al. Resveratrol, a phytoestrogen found in red wine, down‐regulates protein S expression in HepG2 cells. Thromb Res. 2011;127(1):e1‐e7. [DOI] [PubMed] [Google Scholar]
  • 42. Olson ER, Naugle JE, Zhang X, Bomser JA, Meszaros JG. Inhibition of cardiac fibroblast proliferation and myofibroblast differentiation by resveratrol. Am J Physiol Heart Circ Physiol. 2005;288(3):H1131‐H1138. [DOI] [PubMed] [Google Scholar]
  • 43. Glehr M, Fritsch‐Breisach M, Lohberger B, et al. Influence of resveratrol on rheumatoid fibroblast‐like synoviocytes analysed with gene chip transcription. Phytomedicine. 2013;20(3‐4):310‐318. [DOI] [PubMed] [Google Scholar]
  • 44. Yao J, Wang JY, Liu L, et al. Anti‐oxidant effects of resveratrol on mice with DSS‐induced ulcerative colitis. Arch Med Res. 2010;41(4):288‐294. [DOI] [PubMed] [Google Scholar]
  • 45. Zeng G, Zhong F, Li J, Luo S, Zhang P. Resveratrol‐mediated reduction of collagen by inhibiting proliferation and producing apoptosis in human hypertrophic scar fibroblasts. Biosci Biotechnol Biochem. 2013;77(12):2389‐2396. [DOI] [PubMed] [Google Scholar]
  • 46. Ikeda K, Torigoe T, Matsumoto Y, Fujita T, Sato N, Yotsuyanagi T. Resveratrol inhibits fibrogenesis and induces apoptosis in keloid fibroblasts. Wound Repair Regen. 2013;21(4):616‐623. [DOI] [PubMed] [Google Scholar]
  • 47. Mitrica R, Dumitru I, Ruta LL, Ofiteru AM, Farcasanu IC. The dual action of epigallocatechin gallate (EGCG), the main constituent of green tea, against the deleterious effects of visible light and singlet oxygen‐generating conditions as seen in yeast cells. Molecules. 2012;17(9):10355‐10369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Wang X, Cui H, Wu S. CTGF: A potential therapeutic target for bronchopulmonary dysplasia. Eur J Pharmacol. 2019;860:172588. [DOI] [PubMed] [Google Scholar]
  • 49. Wang CY, Deng YT, Huang SY, Liu CM, Chang HH, Wong MY. Epigallocatechin‐3‐gallate inhibits lysophosphatidic acid‐stimulated connective tissue growth factor via JNK and Smad3 suppression in human gingival fibroblasts. J Formosan Med Assoc. 2014;113(1):50‐55. [DOI] [PubMed] [Google Scholar]
  • 50. Klass BR, Branford OA, Grobbelaar AO, Rolfe KJ. The effect of epigallocatechin‐3‐gallate, a constituent of green tea, on transforming growth factor‐beta1‐stimulated wound contraction. Wound Repair Regen: Off Publ Wound Healing Soc [and] Eur Tissue Repair Soc. 2010;18(1):80‐88. [DOI] [PubMed] [Google Scholar]
  • 51. Cai Y, Yu SS, Chen TT, et al. EGCG inhibits CTGF expression via blocking NF‐kappaB activation in cardiac fibroblast. Phytomed: Int J Phytother Phytopharmacol. 2013;20(2):106‐113. [DOI] [PubMed] [Google Scholar]
  • 52. Yang EJ, Lee W, Ku SK, Song KS, Bae JS. Anti‐inflammatory activities of oleanolic acid on HMGB1 activated HUVECs. Food Chem Toxicol. 2012;50(5):1288‐1294. [DOI] [PubMed] [Google Scholar]
  • 53. Chakravarti B, Maurya R, Siddiqui JA, et al. In vitro anti‐breast cancer activity of ethanolic extract of Wrightia tomentosa: role of pro‐apoptotic effects of oleanolic acid and urosolic acid. J Ethnopharmacol. 2012;142(1):72‐79. [DOI] [PubMed] [Google Scholar]
  • 54. Beldon P. Abnormal scar formation in wound healing. Nurs Times. 2000;96(10):44‐45. [PubMed] [Google Scholar]
  • 55. Clark JA, Leung KS, Cheng JC, Leung PC. The hypertrophic scar and microcirculation properties. Burns. 1996;22(6):447‐450. [DOI] [PubMed] [Google Scholar]
  • 56. Dasu MR, Hawkins HK, Barrow RE, Xue H, Herndon DN. Gene expression profiles from hypertrophic scar fibroblasts before and after IL‐6 stimulation. J Pathol. 2004;202(4):476‐485. [DOI] [PubMed] [Google Scholar]
  • 57. Manase K, Endo T, Chida M, et al. Coordinated elevation of membrane type 1‐matrix metalloproteinase and matrix metalloproteinase‐2 expression in rat uterus during postpartum involution. Reprod Biol Endocrinol: RB&E. 2006;4:32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Wei YJ, Yan XQ, Ma L, Wu JG, Zhang H, Qin LP. Oleanolic acid inhibits hypertrophic scarring in the rabbit ear model. Clin Exp Dermatol. 2011;36(5):528‐533. [DOI] [PubMed] [Google Scholar]
  • 59. Zhang H, Zhang Y, Jiang YP, et al. Curative effects of oleanolic Acid on formed hypertrophic scars in the rabbit ear model. Evid Based Complement Alternat Med. 2012;2012:837581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Panchatcharam M, Miriyala S, Gayathri VS, Suguna L. Curcumin improves wound healing by modulating collagen and decreasing reactive oxygen species. Mol Cell Biochem. 2006;290(1‐2):87‐96. [DOI] [PubMed] [Google Scholar]
  • 61. Dujic J, Kippenberger S, Hoffmann S, et al. Low concentrations of curcumin induce growth arrest and apoptosis in skin keratinocytes only in combination with UVA or visible light. J Invest Dermatol. 2007;127(8):1992‐2000. [DOI] [PubMed] [Google Scholar]
  • 62. Scharstuhl A, Mutsaers HA, Pennings SW, Szarek WA, Russel FG, Wagener FA. Curcumin‐induced fibroblast apoptosis and in vitro wound contraction are regulated by antioxidants and heme oxygenase: implications for scar formation. J Cell Mol Med. 2009;13(4):712‐725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Szejk‐Arendt M, Czubak‐Prowizor K, Macieja A, et al. Polyphenolic‐polysaccharide conjugates from medicinal plants of Rosaceae/Asteraceae family protect human lymphocytes but not myeloid leukemia K562 cells against radiation‐induced death. Int J Biol Macromol. 2019. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 64. Garfall AL, June CH. Trispecific antibodies offer a third way forward for anticancer immunotherapy. Nature. 2019;575(7783):450‐451. [DOI] [PubMed] [Google Scholar]
  • 65. Chai CY, Song J, Tan Z, Tai IC, Zhang C, Sun S. Adipose tissue‐derived stem cells inhibit hypertrophic scar (HS) fibrosis via p38/MAPK pathway. J Cell Biochem. 2019;120(3):4057‐4064. [DOI] [PubMed] [Google Scholar]
  • 66. Bai XZ, Liu JQ, Yang LL, et al. Identification of sirtuin 1 as a promising therapeutic target for hypertrophic scars. Br J Pharmacol. 2016;173(10):1589‐1601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Zhao J, Shu B, Chen L, et al. Prostaglandin E2 inhibits collagen synthesis in dermal fibroblasts and prevents hypertrophic scar formation in vivo. Exp Dermatol. 2016;25(8):604‐610. [DOI] [PubMed] [Google Scholar]
  • 68. Gabbiani G. The myofibroblast in wound healing and fibrocontractive diseases. J Pathol. 2003;200(4):500‐503. [DOI] [PubMed] [Google Scholar]
  • 69. Ding J, Ma Z, Shankowsky HA, Medina A, Tredget EE. Deep dermal fibroblast profibrotic characteristics are enhanced by bone marrow‐derived mesenchymal stem cells. Wound Repair Regen. 2013;21(3):448‐455. [DOI] [PubMed] [Google Scholar]
  • 70. van der Veer WM, Bloemen MC, Ulrich MM, et al. Potential cellular and molecular causes of hypertrophic scar formation. Burns. 2009;35(1):15‐29. [DOI] [PubMed] [Google Scholar]
  • 71. Satish L, Kathju S. Cellular and molecular characteristics of scarless versus fibrotic wound healing. Dermatol Res Prac. 2010;2010:790234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Ferguson MW, O'Kane S. Scar‐free healing: from embryonic mechanisms to adult therapeutic intervention. Philos Trans R Soc Lond Ser B Biol Sci. 2004;359(1445):839‐850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol. 2008;214(2):199‐210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Dovi JV, He LK, DiPietro LA. Accelerated wound closure in neutrophil‐depleted mice. J Leukoc Biol. 2003;73(4):448‐455. [DOI] [PubMed] [Google Scholar]
  • 75. Devalaraja RM, Nanney LB, Du J, et al. Delayed wound healing in CXCR2 knockout mice. J Invest Dermatol. 2000;115(2):234‐244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Mirza R, DiPietro LA, Koh TJ. Selective and specific macrophage ablation is detrimental to wound healing in mice. Am J Pathol. 2009;175(6):2454‐2462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. 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] [PubMed] [Google Scholar]
  • 78. Liao X, Sharma N, Kapadia F, et al. Kruppel‐like factor 4 regulates macrophage polarization. J Clin Invest. 2011;121(7):2736‐2749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Mahdavian Delavary B, van der Veer WM, van Egmond M, Niessen FB, Beelen RH. Macrophages in skin injury and repair. Immunobiology. 2011;216(7):753‐762. [DOI] [PubMed] [Google Scholar]
  • 80. Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest. 2012;122(3):787‐795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Zhu Z, Ding J, Ma Z, Iwashina T, Tredget EE. Systemic depletion of macrophages in the subacute phase of wound healing reduces hypertrophic scar formation. Wound Repair Regen: Off Publ Wound Healing Soc [and] Eur Tissue Repair Soc. 2016;24(4):644‐656. [DOI] [PubMed] [Google Scholar]
  • 82. Braga TT, Agudelo JS, Camara NO. Macrophages during the fibrotic process: M2 as friend and foe. Front Immunol. 2015;6:602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Krzyszczyk P, Schloss R, Palmer A, Berthiaume F. The role of macrophages in acute and chronic wound healing and interventions to promote pro‐wound healing phenotypes. Front Physiol. 2018;9:419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Armour A, Scott PG, Tredget EE. Cellular and molecular pathology of HTS: basis for treatment. Wound Repair Regen. 2007;15(Suppl 1):S6‐S17. [DOI] [PubMed] [Google Scholar]
  • 85. Lafyatis R, Farina A. New insights into the mechanisms of innate immune receptor signalling in fibrosis. Open Rheumatol J. 2012;6:72‐79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med. 2012;18(7):1028‐1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Ong C, Wong C, Roberts CR, Teh HS, Jirik FR. Anti‐IL‐4 treatment prevents dermal collagen deposition in the tight‐skin mouse model of scleroderma. Eur J Immunol. 1998;28(9):2619‐2629. [DOI] [PubMed] [Google Scholar]
  • 88. Aliprantis AO, Wang J, Fathman JW, et al. Transcription factor T‐bet regulates skin sclerosis through its function in innate immunity and via IL‐13. Proc Natl Acad Sci USA. 2007;104(8):2827‐2830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Profyris C, Tziotzios C, Do VI. Cutaneous scarring: pathophysiology, molecular mechanisms, and scar reduction therapeutics Part I. The molecular basis of scar formation. J Am Acad Dermatol. 2012;66(1):1‐10. [DOI] [PubMed] [Google Scholar]
  • 90. Peranteau WH, Zhang L, Muvarak N, et al. IL‐10 overexpression decreases inflammatory mediators and promotes regenerative healing in an adult model of scar formation. J Invest Dermatol. 2008;128(7):1852‐1860. [DOI] [PubMed] [Google Scholar]
  • 91. Huang MQ, Cao XY, Chen XY, et al. Saikosaponin a increases interleukin‐10 expression and inhibits scar formation after sciatic nerve injury. Neural Regen Res. 2018;13(9):1650‐1656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Dorati R, Medina JL, DeLuca PP, Leung KP. Development of a topical 48‐H release formulation as an anti‐scarring treatment for deep partial‐thickness burns. AAPS Pharm Sci Tech. 2018;19(5):2264‐2275. [DOI] [PubMed] [Google Scholar]
  • 93. Plikus MV, Guerrero‐Juarez CF, Ito M, et al. Regeneration of fat cells from myofibroblasts during wound healing. Science. 2017;355(6326):748‐752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Melov S, Ravenscroft J, Malik S, et al. Extension of life‐span with superoxide dismutase/catalase mimetics. Science. 2000;289(5484):1567‐1569. [DOI] [PubMed] [Google Scholar]
  • 95. Fujiwara T, Dohi T, Maan ZN, et al. Age‐associated intracellular superoxide dismutase deficiency potentiates dermal fibroblast dysfunction during wound healing. Exp Dermatol. 2017;28(4):485‐492. [DOI] [PubMed] [Google Scholar]
  • 96. Zhao P, Sui BD, Liu N, et al. Anti‐aging pharmacology in cutaneous wound healing: effects of metformin, resveratrol, and rapamycin by local application. Aging Cell. 2017;16(5):1083‐1093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Demyanenko IA, Popova EN, Zakharova VV, et al. Mitochondria‐targeted antioxidant SkQ1 improves impaired dermal wound healing in old mice. Aging (Albany NY). 2015;7(7):475‐485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Zhong L, Hao H, Chen D, et al. Arsenic trioxide inhibits the differentiation of fibroblasts to myofibroblasts through nuclear factor erythroid 2‐like 2 (NFE2L2) protein and the Smad2/3 pathway. J Cell Physiol. 2018;234(3):2606‐2617. [DOI] [PubMed] [Google Scholar]
  • 99. Hinz B. Myofibroblasts. Exp Eye Res. 2016;142:56‐70. [DOI] [PubMed] [Google Scholar]
  • 100. Petrie RJ, Yamada KM. Fibroblasts lead the way: a unified view of 3D cell motility. Trends Cell Biol. 2015;25(11):666‐674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myofibroblasts and mechano‐regulation of connective tissue remodelling. Nat Rev Mol Cell Biol. 2002;3(5):349‐363. [DOI] [PubMed] [Google Scholar]
  • 102. Talele NP, Fradette J, Davies JE, Kapus A, Hinz B. Expression of alpha‐smooth muscle actin determines the fate of mesenchymal stromal cells. Stem Cell Rep. 2015;4(6):1016‐1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Tondeleir D, Lambrechts A, Muller M, et al. Cells lacking beta‐actin are genetically reprogrammed and maintain conditional migratory capacity. Mol Cell Proteomics. 2012;11(8):255‐271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Gauglitz GG, Korting HC, Pavicic T, Ruzicka T, Jeschke MG. Hypertrophic scarring and keloids: pathomechanisms and current and emerging treatment strategies. Mol Med. 2011;17(1‐2):113‐125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Castelino FV, Varga J. Emerging cellular and molecular targets in fibrosis: implications for scleroderma pathogenesis and targeted therapy. Curr Opin Rheumatol. 2014;26(6):607‐614. [DOI] [PubMed] [Google Scholar]
  • 106. Kadler KE, Holmes DF, Trotter JA, Chapman JA. Collagen fibril formation. Biochem J. 1996;316(Pt 1):1‐11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Linares HA, Kischer CW, Dobrkovsky M, Larson DL. The histiotypic organization of the hypertrophic scar in humans. J Invest Dermatol. 1972;59(4):323‐331. [DOI] [PubMed] [Google Scholar]
  • 108. Gay S, Vijanto J, Raekallio J, Penttinen R. Collagen types in early phases of wound healing in children. Acta Chir Scand. 1978;144(4):205‐211. [PubMed] [Google Scholar]
  • 109. Sarrazy V, Billet F, Micallef L, Coulomb B, Desmouliere A. Mechanisms of pathological scarring: role of myofibroblasts and current developments. Wound Repair Regen. 2011;19(Suppl 1):s10‐s15. [DOI] [PubMed] [Google Scholar]
  • 110. Hinz B. The role of myofibroblasts in wound healing. Curr Res Transl Med. 2016;64(4):171‐177. [DOI] [PubMed] [Google Scholar]
  • 111. Sangshetti JN, Sakle NS, Dehghan MH, Shinde DB. Histone deacetylases as targets for multiple diseases. Mini Rev Med Chem. 2013;13(7):1005‐1026. [DOI] [PubMed] [Google Scholar]
  • 112. Wang Z, Chen C, Finger SN, et al. Suberoylanilide hydroxamic acid: a potential epigenetic therapeutic agent for lung fibrosis? Eur Respir J. 2009;34(1):145‐155. [DOI] [PubMed] [Google Scholar]
  • 113. Chen CZ, Peng YX, Wang ZB, et al. The Scar‐in‐a‐Jar: studying potential antifibrotic compounds from the epigenetic to extracellular level in a single well. Br J Pharmacol. 2009;158(5):1196‐1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Huber LC, Distler JH, Moritz F, et al. Trichostatin A prevents the accumulation of extracellular matrix in a mouse model of bleomycin‐induced skin fibrosis. Arthritis Rheum. 2007;56(8):2755‐2764. [DOI] [PubMed] [Google Scholar]
  • 115. Wei J, Ghosh AK, Sargent JL, et al. PPARgamma downregulation by TGFss in fibroblast and impaired expression and function in systemic sclerosis: a novel mechanism for progressive fibrogenesis. PLoS One. 2010;5(11):e13778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Fan C, Dong Y, Xie Y, et al. Shikonin reduces TGF‐beta1‐induced collagen production and contraction in hypertrophic scar‐derived human skin fibroblasts. Int J Mol Med. 2015;36(4):985‐991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Moulin V, Larochelle S, Langlois C, Thibault I, Lopez‐Valle CA, Roy M. Normal skin wound and hypertrophic scar myofibroblasts have differential responses to apoptotic inductors. J Cell Physiol. 2004;198(3):350‐358. [DOI] [PubMed] [Google Scholar]
  • 118. Horowitz JC, Rogers DS, Sharma V, et al. Combinatorial activation of FAK and AKT by transforming growth factor‐beta1 confers an anoikis‐resistant phenotype to myofibroblasts. Cell Signal. 2007;19(4):761‐771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Horowitz JC, Lee DY, Waghray M, et al. Activation of the pro‐survival phosphatidylinositol 3‐kinase/AKT pathway by transforming growth factor‐beta1 in mesenchymal cells is mediated by p38 MAPK‐dependent induction of an autocrine growth factor. J Biol Chem. 2004;279(2):1359‐1367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Garneau‐Tsodikova S, Thannickal VJ. Protein kinase inhibitors in the treatment of pulmonary fibrosis. Curr Med Chem. 2008;15(25):2632‐2640. [DOI] [PubMed] [Google Scholar]
  • 121. Yan C, Grimm WA, Garner WL, et al. Epithelial to mesenchymal transition in human skin wound healing is induced by tumor necrosis factor‐alpha through bone morphogenic protein‐2. Am J Pathol. 2010;176(5):2247‐2258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Hinz B, Gabbiani G. Cell‐matrix and cell‐cell contacts of myofibroblasts: role in connective tissue remodeling. Thromb Haemost. 2003;90(6):993‐1002. [DOI] [PubMed] [Google Scholar]
  • 123. Desmouliere A, Redard M, Darby I, Gabbiani G. Apoptosis mediates the decrease in cellularity during the transition between granulation tissue and scar. Am J Pathol. 1995;146(1):56‐66. [PMC free article] [PubMed] [Google Scholar]
  • 124. 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] [PubMed] [Google Scholar]
  • 125. Cui HS, Hong AR, Kim JB, et al. Extracorporeal shock wave therapy alters the expression of fibrosis‐related molecules in fibroblast derived from human hypertrophic scar. Int J Mol Sci. 2018;19(1):124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Etienne‐Manneville S. Cytoplasmic intermediate filaments in cell biology. Annu Rev Cell Dev Biol. 2018;34:1‐28. [DOI] [PubMed] [Google Scholar]
  • 127. Polioudaki H, Agelaki S, Chiotaki R, et al. Variable expression levels of keratin and vimentin reveal differential EMT status of circulating tumor cells and correlation with clinical characteristics and outcome of patients with metastatic breast cancer. BMC Cancer. 2015;15:399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Konig K, Meder L, Kroger C, et al. Loss of the keratin cytoskeleton is not sufficient to induce epithelial mesenchymal transition in a novel KRAS driven sporadic lung cancer mouse model. PLoS One. 2013;8(3):e57996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Mendez MG, Kojima S, Goldman RD. Vimentin induces changes in cell shape, motility, and adhesion during the epithelial to mesenchymal transition. FASEB J: Off Publ Federation of Am Soc Exp Biol. 2010;24(6):1838‐1851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Eto H, Suga H, Aoi N, et al. Therapeutic potential of fibroblast growth factor‐2 for hypertrophic scars: upregulation of MMP‐1 and HGF expression. Lab Investig. 2012;92(2):214‐223. [DOI] [PubMed] [Google Scholar]
  • 131. Mauviel A. Cytokine regulation of metalloproteinase gene expression. J Cell Biochem. 1993;53(4):288‐295. [DOI] [PubMed] [Google Scholar]
  • 132. Zhang Y, McCluskey K, Fujii K, Wahl LM. Differential regulation of monocyte matrix metalloproteinase and TIMP‐1 production by TNF‐alpha, granulocyte‐macrophage CSF, and IL‐1 beta through prostaglandin‐dependent and ‐independent mechanisms. J Immunol. 1998;161(6):3071‐3076. [PubMed] [Google Scholar]
  • 133. Ghahary A, Shen YJ, Nedelec B, Wang R, Scott PG, Tredget EE. Collagenase production is lower in post‐burn hypertrophic scar fibroblasts than in normal fibroblasts and is reduced by insulin‐like growth factor‐1. J Invest Dermatol. 1996;106(3):476‐481. [DOI] [PubMed] [Google Scholar]
  • 134. Gill SE, Parks WC. Metalloproteinases and their inhibitors: regulators of wound healing. Int J Biochem Cell Biol. 2008;40(6‐7):1334‐1347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Linge C, Richardson J, Vigor C, Clayton E, Hardas B, Rolfe K. Hypertrophic scar cells fail to undergo a form of apoptosis specific to contractile collagen‐the role of tissue transglutaminase. J Invest Dermatol. 2005;125(1):72‐82. [DOI] [PubMed] [Google Scholar]
  • 136. Liang X, Chai B, Duan R, Zhou Y, Huang X, Li Q. Inhibition of FKBP10 attenuates hypertrophic scarring through suppressing fibroblast activity and extracellular matrix deposition. J Invest Dermatol. 2017;137(11):2326‐2335. [DOI] [PubMed] [Google Scholar]
  • 137. Choong OK, Chen CY, Zhang J, et al. Hypoxia‐induced H19/YB‐1 cascade modulates cardiac remodeling after infarction. Theranostics. 2019;9(22):6550‐6567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Kendall RT, Feghali‐Bostwick CA. Fibroblasts in fibrosis: novel roles and mediators. Front Pharmacol. 2014;5:123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Malmstrom J, Lindberg H, Lindberg C, et al. Transforming growth factor‐beta 1 specifically induce proteins involved in the myofibroblast contractile apparatus. Mol Cell Proteomics. 2004;3(5):466‐477. [DOI] [PubMed] [Google Scholar]
  • 140. Shah M, Foreman DM, Ferguson MW. Neutralisation of TGF‐beta 1 and TGF‐beta 2 or exogenous addition of TGF‐beta 3 to cutaneous rat wounds reduces scarring. J Cell Sci. 1995;108(Pt 3):985‐1002. [DOI] [PubMed] [Google Scholar]
  • 141. Ashcroft GS, Yang X, Glick AB, et al. Mice lacking Smad3 show accelerated wound healing and an impaired local inflammatory response. Nat Cell Biol. 1999;1(5):260‐266. [DOI] [PubMed] [Google Scholar]
  • 142. Ishida Y, Gao JL, Murphy PM. Chemokine receptor CX3CR1 mediates skin wound healing by promoting macrophage and fibroblast accumulation and function. J Immunol. 2008;180(1):569‐579. [DOI] [PubMed] [Google Scholar]
  • 143. Wang R, Ghahary A, Shen Q, Scott PG, Roy K, Tredget EE. Hypertrophic scar tissues and fibroblasts produce more transforming growth factor‐beta1 mRNA and protein than normal skin and cells. Wound Repair Regen: Off Publ Wound Healing Soc [and] Eur Tissue Repair Soc. 2000;8(2):128‐137. [DOI] [PubMed] [Google Scholar]
  • 144. Bullard KM, Longaker MT, Lorenz HP. Fetal wound healing: current biology. World J Surg. 2003;27(1):54‐61. [DOI] [PubMed] [Google Scholar]
  • 145. Sun Q, Guo S, Wang CC, et al. Cross‐talk between TGF‐beta/Smad pathway and Wnt/beta‐catenin pathway in pathological scar formation. Int J Clin Exp Pathol. 2015;8(6):7631‐7639. [PMC free article] [PubMed] [Google Scholar]
  • 146. Pakyari M, Farrokhi A, Maharlooei MK, Ghahary A. Critical role of transforming growth factor beta in different phases of wound healing. Adv Wound Care (New Rochelle). 2013;2(5):215‐224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Whitfield ML, Finlay DR, Murray JI, et al. Systemic and cell type‐specific gene expression patterns in scleroderma skin. Proc Natl Acad Sci USA. 2003;100(21):12319‐12324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Sonnylal S, Denton CP, Zheng B, et al. Postnatal induction of transforming growth factor beta signaling in fibroblasts of mice recapitulates clinical, histologic, and biochemical features of scleroderma. Arthritis Rheum. 2007;56(1):334‐344. [DOI] [PubMed] [Google Scholar]
  • 149. Massague J. TGFbeta signalling in context. Nat Rev Mol Cell Biol. 2012;13(10):616‐630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Zhou J, Zhao Y, Simonenko V, et al. Simultaneous silencing of TGF‐beta1 and COX‐2 reduces human skin hypertrophic scar through activation of fibroblast apoptosis. Oncotarget. 2017;8(46):80651‐80665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Shull MM, Ormsby I, Kier AB, et al. Targeted disruption of the mouse transforming growth factor‐beta 1 gene results in multifocal inflammatory disease. Nature. 1992;359(6397):693‐699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Henderson NC, Sheppard D. Integrin‐mediated regulation of TGFbeta in fibrosis. Biochim Biophys Acta. 2013;1832(7):891‐896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Hinz B. It has to be the alphav: myofibroblast integrins activate latent TGF‐beta1. Nat Med. 2013;19(12):1567‐1568. [DOI] [PubMed] [Google Scholar]
  • 154. Viera MH, Vivas AC, Berman B. Update on keloid management: clinical and basic science advances. Adv Wound Care. 2012;1(5):200‐206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Kobayashi M, Ota S, Terada S, et al. The combined use of losartan and muscle‐derived stem cells significantly improves the functional recovery of muscle in a young mouse model of contusion injuries. Am J Sports Med. 2016;44(12):3252‐3261. [DOI] [PubMed] [Google Scholar]
  • 156. Zhou R, Zhang Q, Zhang Y, Fu S, Wang C. Aberrant miR‐21 and miR‐200b expression and its pro‐fibrotic potential in hypertrophic scars. Exp Cell Res. 2015;339(2):360‐366. [DOI] [PubMed] [Google Scholar]
  • 157. Macconi D, Remuzzi G, Benigni A. Key fibrogenic mediators: old players. Renin‐angiotensin system. Kidney Int Suppl. 2014;4(1):58‐64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Lyu L, Zhao Y, Lu H, et al. Integrated interaction network of microrna target genes in keloid scarring. Mol Diagn Ther. 2019;23(1):53‐63. [DOI] [PubMed] [Google Scholar]
  • 159. Akhurst RJ, Hata A. Targeting the TGFbeta signalling pathway in disease. Nat Rev Drug Discov. 2012;11(10):790‐811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Anderton MJ, Mellor HR, Bell A, et al. Induction of heart valve lesions by small‐molecule ALK5 inhibitors. Toxicol Pathol. 2011;39(6):916‐924. [DOI] [PubMed] [Google Scholar]
  • 161. Li N, Kong M, Ma T, Gao W, Ma S. Uighur medicine abnormal savda munzip (ASMq) suppresses expression of collagen and TGF‐beta1 with concomitant induce Smad7 in human hypertrophic scar fibroblasts. Int J Clin Exp Med. 2015;8(6):8551‐8560. [PMC free article] [PubMed] [Google Scholar]
  • 162. Maciver AH, McCall MD, Edgar RL, et al. Sirolimus drug‐eluting, hydrogel‐impregnated polypropylene mesh reduces intra‐abdominal adhesion formation in a mouse model. Surgery. 2011;150(5):907‐915. [DOI] [PubMed] [Google Scholar]
  • 163. Santiago B, Gutierrez‐Canas I, Dotor J, et al. Topical application of a peptide inhibitor of transforming growth factor‐beta1 ameliorates bleomycin‐induced skin fibrosis. J Invest Dermatol. 2005;125(3):450‐455. [DOI] [PubMed] [Google Scholar]
  • 164. Yan X, Liu Z, Chen Y. Regulation of TGF‐beta signaling by Smad7. Acta Biochim Biophys Sin. 2009;41(4):263‐272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Iudintseva NM, Blinova MI, Pinaev GP. Characteristics of cytoskeleton organization of human normal postnatal, scar and embryonic skin fibroblasts spreading on different proteins of extracellular matrix. Tsitologiia. 2008;50(10):861‐867. [PubMed] [Google Scholar]
  • 166. Kopp J, Preis E, Said H, et al. Abrogation of transforming growth factor‐beta signaling by SMAD7 inhibits collagen gel contraction of human dermal fibroblasts. J Biol Chem. 2005;280(22):21570‐21576. [DOI] [PubMed] [Google Scholar]
  • 167. Tang B, Zhu B, Liang Y, et al. Asiaticoside suppresses collagen expression and TGF‐beta/Smad signaling through inducing Smad7 and inhibiting TGF‐betaRI and TGF‐betaRII in keloid fibroblasts. Arch Dermatol Res. 2011;303(8):563‐572. [DOI] [PubMed] [Google Scholar]
  • 168. Walton KL, Johnson KE, Harrison CA. Targeting TGF‐beta mediated SMAD signaling for the prevention of fibrosis. Front Pharmacol. 2017;8:461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Vogel KG, Paulsson M, Heinegard D. Specific inhibition of type I and type II collagen fibrillogenesis by the small proteoglycan of tendon. Biochem J. 1984;223(3):587‐597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Lee WJ, Ahn HM, Roh H, et al. Decorin‐expressing adenovirus decreases collagen synthesis and upregulates MMP expression in keloid fibroblasts and keloid spheroids. Exp Dermatol. 2015;24(8):591‐597. [DOI] [PubMed] [Google Scholar]
  • 171. Jarvelainen H, Puolakkainen P, Pakkanen S, et al. A role for decorin in cutaneous wound healing and angiogenesis. Wound Repair Regen: Off Publ Wound Healing Soc [and] European Tissue Repair Soc. 2006;14(4):443‐452. [DOI] [PubMed] [Google Scholar]
  • 172. Sidgwick GP, Bayat A. Extracellular matrix molecules implicated in hypertrophic and keloid scarring. J Eur Acad Dermatol Venereol. 2012;26(2):141‐152. [DOI] [PubMed] [Google Scholar]
  • 173. Scott PG, Dodd CM, Tredget EE, Ghahary A, Rahemtulla F. Chemical characterization and quantification of proteoglycans in human post‐burn hypertrophic and mature scars. Clin Sci (Lond). 1996;90(5):417‐425. [DOI] [PubMed] [Google Scholar]
  • 174. Zhang Z, Garron TM, Li XJ, et al. Recombinant human decorin inhibits TGF‐beta1‐induced contraction of collagen lattice by hypertrophic scar fibroblasts. Burns. 2009;35(4):527‐537. [DOI] [PubMed] [Google Scholar]
  • 175. Carmichael SW. The tangled web of Langer's lines. Clin Anat. 2014;27(2):162‐168. [DOI] [PubMed] [Google Scholar]
  • 176. Hayden T. Scarred by disease. Nat Med. 2011;17(1):18‐20. [DOI] [PubMed] [Google Scholar]
  • 177. Jones LL, Calvert M, Moiemen N, et al. Outcomes important to burns patients during scar management and how they compare to the concepts captured in burn‐specific patient reported outcome measures. Burns. 2017;43(8):1682‐1692. [DOI] [PubMed] [Google Scholar]
  • 178. Lepault E, Celeste C, Dore M, Martineau D, Theoret CL. Comparative study on microvascular occlusion and apoptosis in body and limb wounds in the horse. Wound Repair Regen: Off Publ Wound Healing Soc [and] European Tissue Repair Soc. 2005;13(5):520‐529. [DOI] [PubMed] [Google Scholar]
  • 179. Ehanire T, Vissoci JR, Slaughter K, et al. A systematic review of the psychometric properties of self‐reported scales assessing burn contractures reveals the need for a new tool to measure contracture outcomes. Wound Repair Regen: Off Publ Wound Healing Soc [and] Eur Tissue Repair Soc. 2013;21(4):520‐529. [DOI] [PubMed] [Google Scholar]
  • 180. Carleton MM, Sefton MV. Injectable and degradable methacrylic acid hydrogel alters macrophage response in skeletal muscle. Biomaterials. 2019;223:119477. [DOI] [PubMed] [Google Scholar]
  • 181. Wang J, Jiao H, Stewart TL, Shankowsky HA, Scott PG, Tredget EE. Increased TGF‐beta‐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] [PubMed] [Google Scholar]
  • 182. Tredget EE, Yang L, Delehanty M, Shankowsky H, Scott PG. Polarized Th2 cytokine production in patients with hypertrophic scar following thermal injury. J Interf Cytokine Res. 2006;26(3):179‐189. [DOI] [PubMed] [Google Scholar]
  • 183. Goldberg SR, McKinstry RP, Sykes V, Lanning DA. Rapid closure of midgestational excisional wounds in a fetal mouse model is associated with altered transforming growth factor‐beta isoform and receptor expression. J Pediatr Surg. 2007;42(6):966‐971. discussion 71‐3. [DOI] [PubMed] [Google Scholar]
  • 184. Jain K, Sykes V, Kordula T, Lanning D. Homeobox genes Hoxd3 and Hoxd8 are differentially expressed in fetal mouse excisional wounds. J Surg Res. 2008;148(1):45‐48. [DOI] [PubMed] [Google Scholar]
  • 185. Niessen FB, Schalkwijk J, Vos H, Timens W. Hypertrophic scar formation is associated with an increased number of epidermal Langerhans cells. J Pathol. 2004;202(1):121‐129. [DOI] [PubMed] [Google Scholar]
  • 186. Pierobon D, Raggi F, Cambieri I, et al. Regulation of Langerhans cell functions in a hypoxic environment. J Mol Med (Berl). 2016;94(8):943‐955. [DOI] [PubMed] [Google Scholar]
  • 187. Ramadhani AM, Derrick T, Macleod D, et al. Immunofibrogenic gene expression patterns in tanzanian children with ocular chlamydia trachomatis infection, active trachoma and scarring: baseline results of a 4‐year longitudinal study. Front Cell Infect Microbiol. 2017;7:406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Jin H, Jia Y, Yao Z, et al. Hepatic stellate cell interferes with NK cell regulation of fibrogenesis via curcumin induced senescence of hepatic stellate cell. Cell Signal. 2017;33:79‐85. [DOI] [PubMed] [Google Scholar]
  • 189. Li Y, Huang L, Cai Z, et al. A study of the immunoregulatory function of TLR3 and TLR4 on mesenchymal stem cells in ankylosing spondylitis. Stem Cells Dev. 2019;28(20):1398‐1412. [DOI] [PubMed] [Google Scholar]
  • 190. Shi X, Liu J, Chen D, et al. MSC‐triggered metabolomic alterations in liver‐resident immune cells isolated from CCl4‐induced mouse ALI model. Exp Cell Res. 2019;383(2):111511. [DOI] [PubMed] [Google Scholar]
  • 191. Caccioppo A, Franchin L, Grosso A, Angelini F, D'Ascenzo F, Brizzi MF. Ischemia reperfusion injury: mechanisms of damage/protection and novel strategies for cardiac recovery/regeneration. Int J Mol Sci. 2019;20(20):5024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Prockop DJ, Oh JY. Mesenchymal stem/stromal cells (MSCs): role as guardians of inflammation. Mol Ther. 2012;20(1):14‐20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. De Miguel MP, Fuentes‐Julian S, Blazquez‐Martinez A, et al. Immunosuppressive properties of mesenchymal stem cells: advances and applications. Curr Mol Med. 2012;12(5):574‐591. [DOI] [PubMed] [Google Scholar]
  • 194. Elizabeth E, Baranwal G, Krishnan AG, Menon D, Nair M. ZnO nanoparticle incorporated nanostructured metallic titanium for increased mesenchymal stem cell response and antibacterial activity. Nanotechnology. 2014;25(11):115101. [DOI] [PubMed] [Google Scholar]
  • 195. Nuschke A. Activity of mesenchymal stem cells in therapies for chronic skin wound healing. Organogenesis. 2014;10(1):29‐37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Kim HS, Yun JW, Shin TH, et al. Human umbilical cord blood mesenchymal stem cell‐derived PGE2 and TGF‐beta1 alleviate atopic dermatitis by reducing mast cell degranulation. Stem Cells. 2015;33(4):1254‐1266. [DOI] [PubMed] [Google Scholar]
  • 197. Shin TH, Lee BC, Choi SW, et al. Human adipose tissue‐derived mesenchymal stem cells alleviate atopic dermatitis via regulation of B lymphocyte maturation. Oncotarget. 2017;8(1):512‐522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Navas A, Magana‐Guerrero FS, Dominguez‐Lopez A, et al. Anti‐inflammatory and anti‐fibrotic effects of human amniotic membrane mesenchymal stem cells and their potential in corneal repair. Stem Cells Transl Med. 2018;7(12):906‐917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Fang F, Huang RL, Zheng Y, Liu M, Huo R. Bone marrow derived mesenchymal stem cells inhibit the proliferative and profibrotic phenotype of hypertrophic scar fibroblasts and keloid fibroblasts through paracrine signaling. J Dermatol Sci. 2016;83(2):95‐105. [DOI] [PubMed] [Google Scholar]
  • 200. Wang L, Hu L, Zhou X, et al. Exosomes secreted by human adipose mesenchymal stem cells promote scarless cutaneous repair by regulating extracellular matrix remodelling. Sci Rep. 2017;7(1):13321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Jiao Y, Wang X, Zhang J, Qi Y, Gong H, Jiang D. Inhibiting function of human fetal dermal mesenchymal stem cells on bioactivities of keloid fibroblasts. Stem Cell Res Ther. 2017;8(1):170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Yates CC, Rodrigues M, Nuschke A, et al. Multipotent stromal cells/mesenchymal stem cells and fibroblasts combine to minimize skin hypertrophic scarring. Stem Cell Res Ther. 2017;8(1):193. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from International Wound Journal are provided here courtesy of Wiley

RESOURCES