REGENERATIVE MEDICINE Scarless Wound Healing: Chasing the Holy Grail Graham G. Walmsley, B.A. Zeshaan N. Maan, M.B.B.S., M.S. Victor W. Wong, M.D. Dominik Duscher, M.D. Michael S. Hu, M.D. Elizabeth R. Zielins, M.D. Taylor Wearda, B.A. Ethan Muhonen, B.A. Adrian McArdle, M.B., B.Ch., B.A.O. Ruth Tevlin, M.B., B.Ch., B.A.O. David A. Atashroo, M.D. Kshemendra Senarath-Yapa, M.A., M.B.B.Chir. H. Peter Lorenz, M.D. Geoffrey C. Gurtner, M.D. Michael T. Longaker, M.D., M.B.A. Stanford, Calif.; and Baltimore, Md.

Summary: Over 100 million patients acquire scars in the industrialized world each year, primarily as a result of elective operations. Although undefined, the global incidence of scarring is even larger, extending to significant numbers of burn and other trauma-related wounds. Scars have the potential to exert a profound psychological and physical impact on the individual. Beyond aesthetic considerations and potential disfigurement, scarring can result in restriction of movement and reduced quality of life. The formation of a scar following skin injury is a consequence of wound healing occurring through reparative rather than regenerative mechanisms. In this article, the authors review the basic stages of wound healing; differences between adult and fetal wound healing; various mechanical, genetic, and pharmacologic strategies to reduce scarring; and the biology of skin stem/progenitor cells that may hold the key to scarless regeneration.   (Plast. Reconstr. Surg. 135: 907, 2015.)

A

dult mammalian wound healing involves a complex series of molecular and cellular events that typically culminate in a fibrotic “patch-like” repair. Wound healing can be considered in three overlapping stages. The first stage, inflammation, begins with the formation of a fibrin clot in conjunction with platelets at the site of injury. This provides a temporary extracellular matrix, and a stimulus that recruits inflammatory cells to the wound environment. The first immune cells to arrive are neutrophils, whose primary function is prevention of bacterial infection. They are found in highest concentrations 1 to 2 days after injury.1,2 Days 2 and 3 after wounding see the arrival of circulating monocytes and macrophages,3 which play a role in the transition between the inflammatory stage and subsequent proliferative stage of wound healing. Early wound macrophages From the Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, and the Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine; and the Department of Surgery, Division of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine. Received for publication May 15, 2014; accepted August 14, 2014. Copyright © 2015 by the American Society of Plastic Surgeons DOI: 10.1097/PRS.0000000000000972

serve to phagocytose apoptotic neutrophils and clear debris. In days 5 to 7 after injury, wound macrophages adopt a more antiinflammatory role in preparation for tissue rebuilding. Moreover, secretion of growth factors, such as plateletderived growth factor and transforming growth factor (TGF)-β, allows macrophages to stimulate fibroblast migration and activation.1,4 The proliferative phase of wound healing is estimated to begin within 48 hours to 10 days after wounding and is characterized by replacement of the temporary, fibrin-based extracellular matrix with granulation tissue,3 which consists of a vascularized extracellular matrix formed by fibroblasts and endothelial cells. The formation of granulation tissue facilitates reepithelialization, which occurs through the migration and proliferation of keratinocytes, beginning hours after wounding.5 The migrating fibroblasts secrete newly formed extracellular matrix, composed of glycosaminoglycans, proteoglycans, and collagen, although the proportion of type III to type I collagen is higher in early scar tissue than Disclosure: The authors wish to voluntarily declare a potential conflict of interest involving Neodyne Biosciences. Dr. Gurtner and Dr. Longaker are founders and serve on the board of directors. The other authors have no financial information to disclose.

www.PRSJournal.com

907

Plastic and Reconstructive Surgery • March 2015 in unwounded tissue or mature scar.1 In parallel, myofibroblast-mediated wound contraction reduces the original wound size by bringing wound edges closer together.1,3 Furthermore, migration of endothelial cells, mediated largely by vascular endothelial growth factor production from macrophages and fibroblasts,1,4 and subsequent angiogenesis of the newly formed tissue are responsible for the red hue of immature scars. In humans, wound closure is achieved primarily through granulation tissue formation and reepithelialization, whereas in loose-skinned animals (e.g., mice), wound closure is achieved primarily through contraction. Although collagen deposition by fibroblasts and myofibroblasts is typically at its maximal level 3 weeks after injury, at this point, wound strength is at a minimum. Subsequent remodeling of collagen, along with a shift toward a more normal proportion of type I to type III collagen, results in a mature scar that has 80 percent of the strength of normal skin after 3 to 4 months.1 Remodeling, the final phase of wound healing, begins after reepithelialization and wound closure, typically from 14 to 21 days after injury, and can last for over 1 year.3 Reorganization of extracellular matrix by matrix metalloproteinases and collagenases is accompanied by decreased cellularity and vascularity of scar tissue. Epithelial

appendages (i.e., hair follicles, sweat glands, and sebaceous glands) are not reformed, and a healed scar therefore represents a “bare area.” Thus, the final product of the body’s response to injury is a relatively acellular mass of epithelialized extracellular matrix that has a distinct life cycle of its own (Fig. 1).1,6–8 Although normal wound healing typically follows the phases and time frames outlined above, the degree of scarring occurs along a continuum. The first type of pathologic scarring (hypertrophic) typically develops following wound closure under excess tension or infection.9 Hypertrophic scarring is characterized by wide, raised scars that remain within the original borders of injury and have a rapid growth phase of 6 months with gradual regression to a flat scar over a period of 1 to 3 years.10 Another type of pathologic scarring (i.e., keloids) differs markedly from both normal and hypertrophic scars. Keloids appear as firm, bosselated tumors that extend beyond the original borders of injury, do not regress spontaneously, and can develop up to several years after cutaneous injury.11

FETAL SCARLESS WOUND HEALING Early fetal skin has unique wound healing capabilities, with an intrinsic ability to

Fig. 1. Schematic representing the timing of major clinical manifestations and their underlying biology in the life cycle of scar formation. Erythema, heat, and swelling occur from days 0 to 6 and are attributable to platelet degranulation, vasodilation, and leukocytic infiltration. Joint contractures peak at approximately 3 to 6 months, and are caused by altered matrix metalloproteinase expression, changes in extracellular matrix composition, collagen cross-linking, and increased myofibroblasts at the scar site. Finally, pruritus and pain typically peak at 9 months and are caused by small-fiber neuropathy and histamine release.

908

Volume 135, Number 3 • Scarless Wound Healing regenerate tissue rather than form a scar.12,13 Understanding the mechanisms underlying regenerative healing in the fetus may potentially inform scarless healing in adults. After wounding, fetal skin is capable of rapid reepithelialization,14–16 with minimal inflammation.17 Fetal skin undergoes many changes during development. A single layer of ectodermal cells, seen at days 7 and 8 in human gestation, changes into stratified, keratinized epithelium between 22 and 24 weeks. Keratin expression varies throughout development and differs greatly from that of adult epithelium.18 The composition of extracellular matrix is thought to contribute significantly to fetal healing. Fetal tissues display an elevated ratio of type III to type I collagen in comparison with adult tissues.19 The loose network of extracellular matrix helps to promote cellular migration, proliferation, and differentiation, thought to play an important role in wound healing.15,20 The extracellular matrix of fetal tissue is also rich in hyaluronic acid, a glycosaminoglycan that promotes a microenvironment conducive to cell motility and inhibits the proliferation of fetal fibroblasts.21,22 The key differences between fetal and adult wounds are summarized in Table 1.12–22 Although the environment within which the fetus resides has been thought to contribute to its regenerative healing,23,24 prior studies using transplantation methodologies have suggested that the ability of fetal skin to heal scarlessly is cell intrinsic. Longaker and colleagues showed that full-thickness adult sheep skin transplanted onto the dorsa of 60-day gestational fetal lambs healed with scar formation after incisional wounding.25 In a reciprocal experiment, Lorenz et al. showed that human fetal skin transplanted subcutaneously into adult athymic mice recapitulated the scarless phenotype after incisional wounding.13 Collectively, these experiments suggest that properties intrinsic in the transplanted tissues are more significant than extrinsic

environmental factors in determining the outcome of wound healing and suggest that therapies directly targeting the fibroblasts largely responsible for scar formation may be effective. In summary, understanding fetal wound healing offers a blueprint for manipulating postnatal healing to be more fetal-like.

SKIN STEM CELLS IN WOUND HEALING To date, exciting advances in genetic lineage-tracing technologies, cellular assays, and imaging techniques have revealed important stem and progenitor cell reservoirs in the interfollicular epidermis, the eccrine sweat glands, and the hair follicle.26 Given that scarring following a full-thickness excisional wound is defined as a bare area lacking sweat glands, sebaceous glands, and hair follicle regeneration, a thorough understanding of these stem and progenitor cells will likely contribute to achieving the ultimate goal of scarless wound healing following injury. Interfollicular Epidermis The epidermis is a dynamic structure tasked with protecting the body from the outside world. As such, it frequently sustains damage and must repair itself.3 This healing is accomplished through the contributions of numerous tissuespecific stem and progenitor cell populations located throughout the epidermis and its associated appendages.27 During wound healing, a population of “slow-cycling cells” is proposed to exhibit autocrine regulation28 until mobilized by a wound healing response. When wounding occurs, slow-cycling cell division frequency increases, providing excess daughter cells to help repair the damage. Given the gathering evidence for behavioral plasticity in injured epithelia, with progenitors capable of reverting to multipotent states and multipotent cells differentiating to fill unipotent roles,29,30 these data hold exciting ramifications

Table 1.  Key Differences between Adult and Fetal Wound Healing Inflammation Hyaluronic acid21,22 Collagen15,20 Keratin14,18 Mast cells15,16 TGF-β12–14 Stem cells12 17,18

Fetal Wounds

Adult Wounds

Few or no inflammatory cells High, prolonged levels, promotes cellular movement Elevated ratio of type III compared to type I K8 and K19 are present during fetal development Low numbers and less mature Low expression of TGF-β1 and TGF-β2 Higher levels of MSCs at injury sites with ­accompanying E-cadherin–positive cells

Many inflammatory cells Lower levels, inhibits cellular movement Elevated ratio of type I compared to type III Absence of K8 and K19 Higher numbers, more mature High expression of TGF-β1 and TGF-β2 Lower levels of MSCs at injury sites without accompanying E-cadherin–positive cells

MSCs, mesenchymal stem cells.

909

Plastic and Reconstructive Surgery • March 2015 for researchers hoping to shift wound healing away from scarring and toward a more regenerative phenotype. Sweat Glands Eccrine sweat glands (henceforth “sweat glands”) are widely distributed across the adult human body, and can reach a density of up to approximately 700 glands/cm2 of adult human skin.31 Loss of sweat glands in burn patients remains an unsolved problem. Recent groundbreaking studies of the biology and development of these epidermal appendages have identified gland-specific progenitors,32 and revealed the roles of these cells in development, homeostasis, and wound repair.32,33 In elucidating the developmental lineage and healing capabilities of sweat gland structures, the common heritage of all epidermal appendages is further underscored. Developmentally, nascent sweat glands form from a downfolding of multipotent, K5- and K15-positive, epidermal basal progenitors. As morphology becomes more specified, these multipotent progenitors further differentiate into unipotent luminal and myoepithelial progenitor populations. Two sweat apparatus progenitor populations are of particular interest in epidermal regeneration and in considering potential therapeutic strategies. These are the progenitor populations housed in the myoepithelium and the multipotent progenitors found in the sweat gland duct. When engrafted into murine back skin, myoepithelial progenitors have intriguingly been observed to form new epidermis,32,34 which reflects the common multipotent K14-positive basal cell origins of both the interfollicular epidermis and the sweat glands. Regarding native wound healing responses, the luminal and myoepithelial progenitors in the secretory coil have only been observed to proliferate and contribute to the repair of deep glandular wounds.32 In contrast, progenitors in the sweat gland duct have been noted to proliferate and contribute to healing scratch wounds in the interfollicular epidermis. This observation, speculated to be orchestrated by a mechanism of keratinocyte outgrowth,33 is of immense interest to translational medicine. As physicians and researchers further decipher the myriad iterations of healing strategies used throughout the epidermis, we may concomitantly open the door to a future where therapeutically selecting from among epidermal progenitors becomes reality.

910

Hair Follicles The mammalian hair follicle is a dynamic structure known to house numerous progenitor cell populations and contribute to the repair of injured skin.35–38 The rapid turnover of these cells makes lineage analysis a considerable challenge, but a multipotent progenitor population capable of regenerating basal keratinocytes has been characterized.39 Cells in the bulge are multipotent, have high proliferative potential, and are capable of giving rise to all epithelial layers in the hair follicle.40 During wound healing, cells in the bulge and upper follicle migrate and contribute to interfollicular epidermis regeneration.41 A recent study successfully generated folliculogenic human epithelial stem cells from induced pluripotent stem cells and demonstrated differentiation along all hair follicle lineages in skin reconstitution assays.42 Given that loss of dermal appendages is an unwanted feature of cutaneous scarring, the transplantation of induced pluripotent stem cell– derived folliculogenic epithelial stem cells holds promise for the regeneration of normal skin architecture following cutaneous injury.

FIBROBLAST LINEAGES Dermal fibroblasts represent a heterogeneous population of stromal cells and are responsible for scar deposition following cutaneous injury. In 1987, Cormack demonstrated that two subpopulations of fibroblasts reside within the papillary and reticular dermis, respectively.43 Later, a third subpopulation was found associated with hair follicles in the dermal papilla region of the follicle.44,45 When cultured, these fibroblast subtypes exhibit differences in growth potential, morphology, and growth factor/cytokines profiles.46–49 However, these and other studies examining fibroblast heterogeneity have largely failed to convincingly demonstrate developmental and functional diversity among fibroblasts in vivo, particularly in the case of dermal skin fibroblasts. A recent article by Driskell et al. revealed that skin connective tissue contains two distinct lineages of fibroblasts and that each lineage exhibits specific functional properties.50 The first lineage gives rise to the upper dermis and is responsible for hair follicle formation, and the second gives rise to the lower dermis and is responsible for the deposition of connective tissue following excisional wounding. The study demonstrated, for the first time, that distinct embryonic origins for functional subtypes of fibroblasts exist. Most importantly from the perspective of scarless

Volume 135, Number 3 • Scarless Wound Healing wound healing, the study showed that epidermal B-catenin activation stimulates the expansion of the upper dermal lineage and renders excisional wounds permissive to hair follicle growth. As our understanding of the heterogeneous fibroblast population continues to evolve, novel molecular and cellular targets for the modulation of both scar deposition and dermal appendage regeneration will likely emerge.

STRATEGIES TO REDUCE SCARRING Scars resulting from surgical incisions represent an enormous market totaling over 100 million cases per year in the developed world.51 Cutaneous injury initiates a complex cascade of molecular and cellular events within a dynamic extracellular matrix, features of which are common among fibrotic diseases (Fig. 2).2, 52–54 For this reason, treatment modalities capable of broadly influencing biological repair pathways hold the greatest potential for scar reduction. Strategies to minimize the appearance of these

scars and restore the integrity of normal skin will be reviewed here. Current Pharmaceutical Strategies Topical therapy and intralesional injections of corticosteroids have formed the mainstay of nonsurgical therapy for the treatment of keloids and hypertrophic scars for many years. Although this approach can be effective, with success rates of 50 to 100 percent reported in the literature,55–57 it is also associated with hypopigmentation, dermal atrophy, telangiectasia, widening of the scar, and delayed wound healing.56 Although the precise mechanisms through which steroids exert their effects in this context have not been fully elucidated, their principal role is thought to be inhibition of the inflammatory response, suppressing fibroblast growth and promoting collagen degradation.57 Triamcinolone, the most widely used agent, is able to inhibit TGF-β1 expression and induce apoptosis in fibroblasts,58 and inhibit TGFβ1, COL4A1, and COL7A1 in keratinocytes.59

Fig. 2. Schematic representing the common biology of fibrotic diseases such as idiopathic pulmonary fibrosis, renal fibrosis, scleroderma, cirrhosis, and scarring. In general, transient or chronic inflammatory states characterized by the presence of neutrophils, macrophages, and lymphocytes stimulate resident fibroblasts to produce and secrete collagen proteins, which then crosslink to form mature collagen fibers. The link between inflammation and fibrosis includes key mediators such as the mechanoresponsive focal adhesion kinase–monocyte chemoattractant protein pathway, and the TGF-β and Wnt signaling pathways.

911

Plastic and Reconstructive Surgery • March 2015 Evidence supporting the use of calcium antagonists as a standalone treatment remains scarce, although some studies have used them as an adjunct to other therapies. Intralesional verapamil used as an adjunct to surgical excision and topical silicone was found to increase the cure rate of keloids from 18 percent with the latter two treatments alone, to 54 percent in combination with verapamil.60 Although the mechanism of action remains poorly delineated, there is evidence that verapamil injection decreases collagen production.61 Verapamil has also been shown to reduce the production of interleukin-6 (which is thought to be a mitogenic signal for keratinocytes) and vascular endothelial growth factor in fibroblasts harvested from the central portions of keloid scars.62 Topical agents based on plant extracts also exist. Mederma (Merz Pharmaceuticals, Frankfurt, Germany) is a topical gel used to improve scar appearance, with the extract from the common onion as the main active ingredient. Onion extract contains a bioflavonoid with both antihistamine and antiproliferative effects.63 Although studies have shown that Mederma improves collagen organization following excisional ear wounding in rabbits,63 a 2006 clinical trial found no statistically significant difference in hypertrophic scar appearance compared with standard petroleum emollient.64 These findings illustrate a common pattern seen in treatments attempting to reduce scar formation: success in animal models is often not recapitulated in human clinical trials. Gene Targets A number of genes involved in the scarring response have been identified, typically influencing fibrosis by regulating collagen production and degradation. Molecularly targeted therapies seek to either inhibit or up-regulate the pathways controlled by these genes to decrease collagen synthesis.65–68 TGF-β is a major regulator of fibroblast physiology. TGF-β expression has been strongly implicated in the fibroblast-to-myofibroblast transition and in the production and secretion of extracellular matrix. The significant role of TGF-β has garnered it attention as a potential target for fibrosis therapy. The gene is expressed as three isoforms in humans with differential activity. TGF-β1 and TGF-β2 both promote fibrosis, whereas TGF-β3 has been shown to decrease fibrosis and scarring.69,70 Therapeutic strategies targeting TGF-β signaling involve the use of either TGF-β1 and TGF-β2 inhibitors and neutralizing antibodies or up-regulating TGF-β3 signaling.

912

Neutralizing antibodies bind directly to TGFβ1 and TGF-β2, preventing them from binding to and activating TGF-β receptors. In animal models, these antibodies have successfully reduced fibrosis across a number of organs, including a reduction in cutaneous scarring following injury.71–74 Unfortunately, the success of TGF-β antibodies in animal models has not translated across to human clinical trials. Metelimumab, the first anti–TGF-β antibody used in clinical trials for the treatment of systemic sclerosis, demonstrated no significant benefits.75 The TGF-β and platelet-derived growth factor inhibitor imatinib mesylate was also unsuccessful in phase II clinical trials targeting scleroderma.76 Up-regulating TGF-β3 has also demonstrated efficacy in animal models, reducing scarring in a number of studies.77–80 Unfortunately, unpublished data from an international phase III clinical trial has shown that TGF-β3 has no significant effect on scarring (Juvista EU; Renovo, Bristol, United Kingdom). A number of other genes involved in fibrosis have also been addressed in animal models, including modulation of Wnt signaling,65 targeting the hedgehog pathway,81 interleukin-10, which has been implicated in a variety of mechanisms leading to fibrosis,69,82–86 histone deacetylase,66 early growth response gene-1,82 and heat shock protein 90.67 Unfortunately, efficacy is limited to animal studies. Although murine models for both normal87 and hypertrophic scarring88 exist, the discrepancy between animal and clinical trial results highlights differences in signaling between the two species and suggests the need for alternative approaches to address the signaling overlap. Dermal Substitutes Dermal substitutes provide coverage to the wound site and establish a matrix that encourages engraftment and proliferation of endogenous cells and enhances the function of transplanted cells. Acellular matrices, derived from animal or human tissue through the removal of living cells, are biocompatible, remain morphologically similar to natural tissue structure, and have mechanical properties similar to host skin.89 Acellular matrices derived from human dermis currently in clinical use include AlloDerm (LifeCell Corp., Branchburg, N.J.), DermaMatrix (Synthes, West Chester, Pa.), and FlexHD (Ethicon, Inc., Somerville, N.J.). Dermal substitutes using human chorion and amnion have also been used. Among these, Epifix is composed of dehydrated human amnion/chorion membrane (MiMedx Group, Kennesaw, Ga.). Grafix (Osiris Therapeutics, Inc.,

Volume 135, Number 3 • Scarless Wound Healing Columbia, Md.) is cryopreserved placental membrane with native mesenchymal stem cells used as a covering to promote healing and tissue repair. Xenografts are skin substitutes for use in humans harvested from animals. The OASIS Wound Matrix (Cook Biotech, Inc., West Lafayette, Ind.), derived from the submucosal layers of porcine jejunum, is one of the more commonly used non–human-derived acellular matrices in clinical use. Similarly, Surgisis (Cook Biotech) is a biological graft extracted from porcine small intestinal submucosa. More recently, fetal bovine dermal matrices have becomes available, including PriMatrix (TEI Biosciences, Inc., Waltham, Mass.) and SurgiMend (TEI Biosciences). Synthetic fabricated scaffolds are typically composed of proteins normally present in extracellular matrix. Integra (Integra, Plainsboro, N.J.) is composed of bovine collagen and chondroitin6-sulfate cross-linked with glutaraldehyde with a silicone membrane as a temporary epidermis.90 Chitosan-based scaffolds have shown promise as a supportive template for fibroblast attachment.91 A biodegradable porous scaffold composed of collagen, hyaluronic acid, and gelatin accelerated wound closure by providing optimal pore size/ water absorption for human skin cell growth.92 Pullulan-collagen composite dermal hydrogels have been shown to accelerate early wound healing, potentially by modifying cell recruitment and augmenting granulation tissue formation.93 Matrices seeded with cells may also play a role in preventing scar formation. Although useful for enhancing the rate of wound healing, particularly in the context of burns and large excisional wounds, dermal substitutes have not been shown to significantly impact scarring. In the future, the seeding of purified stem cell populations and niche components onto dermal substitutes may allow for a more significant shift away from repair and toward regeneration. Progress on this front will first require a more thorough characterization of skin stem cell populations and their niches. Mechanical Offloading An alternative approach to limiting fibrosis in the setting of cutaneous injury is mechanical offloading. Mechanical tension plays a significant role in the development of fibrosis, activating a number of mechanoresponsive signaling pathways, with focal adhesion kinase being one of the best described.53,94 Plastic surgeons have long been aware of the presence of Langer lines in human skin, which correspond to the natural

orientation of collagen fibers within the dermis. Incisions made parallel to Langer lines generally heal with less scar (because of reduced tension) than those placed perpendicular.95 Paper tape96,97 and silicone sheets98–100 have been reported to reduce scarring, with a proposed mechanism related primarily to tensile reduction. In a randomized controlled trial of 39 patients, the delayed application of paper tapes to closed surgical wounds for up to 12 weeks postoperatively was shown to reduce scar size and decrease the likelihood of developing a hypertrophic scar.96 A study involving 195 patients with torso scars (63 taped and 73 controls) found an overall improvement in scar appearance in taped compared with control scars.97 Success in animal models has also been reported, with microporous paper tape application demonstrating reduced hypertrophic scarring in a rabbit ear model.101 In an attempt to further improve the action of tapes, hybrid materials such as Dynaclose (mediGroup Australia Pty Ltd., Melbourne, Victoria, Australia), a tape with a central region of silicone elastomer, have been developed.102 However, when used as an alternative to sutures, paper tape alone had no significant effect on scar size versus sutures alone in closing abdominal wounds.103 Similarly, the use of Steri-Strip S (3M, St. Paul, Minn.) tapes compared to sutures alone for the closure of median sternotomy incisions demonstrated no significant difference in scar size.104 Silicone gel sheets have been widely used for many years despite an overall lack of knowledge regarding the underlying mechanism and limited evidence supporting clinical efficacy. Although studies have shown that silicone gel sheets effectively reduce tension on the wound site,105 other mechanisms for the scar-reducing effects such as hydration of the stratum corneum,106,107 direct modification of extracellular matrix composition,108,109 and decreased TGF-β2 expression in scar fibroblasts110,111 have been proposed. Silicone gel sheeting placed over evolving hypertrophic and keloid scars led to reductions in scar size in 17 of 20 patients.98 Another study following 30 patients with scars of different types—including superficial scars, hypertrophic scars, and keloids—found that silicone gel application initiated a few days after wound closure led to improvements in scar grade after 6-month follow-up.99 Recently, more focused efforts to provide mechanical offloading during wound healing have demonstrated efficacy in both animal and human clinical trials in reducing fibrosis by simultaneously influencing multiple signaling pathways with

913

Plastic and Reconstructive Surgery • March 2015 a mechanoreceptive element. A phase I clinical trial112 and a multicenter, randomized, controlled trial113 have demonstrated that stress-shielding of half of an abdominal incision leads to significant improvements in scar appearance compared with the contralateral side. In addition to scarring following primary surgery, mechanical offloading offers an opportunity to reduce scarring following scar revision surgery. The device (Neodyne Biosciences, Inc., Menlo Park, Calif.) significantly decreased scarring following revision surgery in a randomized controlled trial of 12 patients.114 Collectively, these findings suggest that the pathways involved in fibrosis are too complex to currently target pharmacologically, and that device approaches appear highly promising for reducing scarring following primary and revision surgery.

CONCLUSIONS Our current understanding of wound healing and cell subpopulations within the skin has permitted the development of scar-reducing therapies with only marginal effectiveness. Although differences between wound healing in fetal and adult skin have been well-characterized, the cellular mechanisms governing fetal skin regeneration remain poorly understood. This is not surprising considering that we are only now beginning to precisely characterize distinct functional lineages in the skin. Achieving scarless wound healing in the adult will require not only an understanding of signaling molecules and growth factors but also a thorough understanding of lineage-specific cellular origin and function during both fetal and adult stages. Michael T. Longaker, M.D., M.B.A. Department of Surgery Division of Plastic and Reconstructive Surgery Stanford University School of Medicine 257 Campus Drive Stanford, Calif. 94305-5148 [email protected] Geoffrey C. Gurtner, M.D. Department of Surgery Division of Plastic and Reconstructive Surgery Stanford University School of Medicine 257 Campus Drive Stanford, Calif. 94305-5148 [email protected]

references 1. Baum CL, Arpey CJ. Normal cutaneous wound healing: Clinical correlation with cellular and molecular events. Dermatol Surg. 2005;31:674–686; discussion 686. 2. Singer AJ, Clark RA. Cutaneous wound healing. N Engl J Med. 1999;341:738–746.

914

3. Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature 2008;453:314–321. 4. Sindrilaru A, Scharffetter-Kochanek K. Disclosure of the culprits: Macrophages—Versatile regulators of wound healing. Adv Wound Care (New Rochelle) 2013;2:357–368. 5. Raja, Sivamani K, Garcia MS, Isseroff RR. Wound re-epithelialization: Modulating keratinocyte migration in wound healing. Front Biosci. 2007;12:2849–2868. 6. Nedelec B, Ghahary A, Scott PG, Tredget EE. Control of wound contraction: Basic and clinical features. Hand Clin. 2000;16:289–302. 7. Orgill DP, Ogawa R. Current methods of burn reconstruction. Plast Reconstr Surg. 2013;131:827e–836e. 8. Lee SS, Yosipovitch G, Chan YH, Goh CL. Pruritus, pain, and small nerve fiber function in keloids: A controlled study. J Am Acad Dermatol. 2004;51:1002–1006. 9. Murray JC, Pollack SV, Pinnell SR. Keloids and hypertrophic scars. Clin Dermatol. 1984;2:121–133. 10. Alster TS, West TB. Treatment of scars: A review. Ann Plast Surg. 1997;39:418–432. 11. Al-Attar A, Mess S, Thomassen JM, Kauffman CL, Davison SP. Keloid pathogenesis and treatment. Plast Reconstr Surg. 2006;117:286–300. 12. Lo DD, Zimmermann AS, Nauta A, Longaker MT, Lorenz HP. Scarless fetal skin wound healing update. Birth Defects Res C Embryo Today 2012;96:237–247. 13. Lorenz HP, Longaker MT, Perkocha LA, Jennings RW, Harrison MR, Adzick NS. Scarless wound repair: A human fetal skin model. Development 1992;114:253–259. 14. Hantash BM, Zhao L, Knowles JA, Lorenz HP. Adult and fetal wound healing. Front Biosci. 2008;13:51–61. 15. Larson BJ, Longaker MT, Lorenz HP. Scarless fetal wound healing: A basic science review. Plast Reconstr Surg. 2010;126:1172–1180. 16. Wilgus TA. Regenerative healing in fetal skin: A review of the literature. Ostomy Wound Manage. 2007;53:16–31; quiz 32. 17. Krummel TM, Nelson JM, Diegelmann RF, et al. Fetal response to injury in the rabbit. J Pediatr Surg. 1987;22:640–644. 18. Coolen NA, Schouten KC, Middelkoop E, Ulrich MM. Comparison between human fetal and adult skin. Arch Dermatol Res. 2010;302:47–55. 19. Merkel JR, DiPaolo BR, Hallock GG, Rice DC. Type I and type III collagen content of healing wounds in fetal and adult rats. Proc Soc Exp Biol Med. 1988;187:493–497. 20. Colwell AS, Longaker MT, Lorenz HP. Fetal wound healing. Front Biosci. 2003;8:s1240–s1248. 21. Longaker MT, Chiu ES, Adzick NS, Stern M, Harrison MR, Stern R. Studies in fetal wound healing: V. A prolonged presence of hyaluronic acid characterizes fetal wound fluid. Ann Surg. 1991;213:292–296. 22. Mast BA, Diegelmann RF, Krummel TM, Cohen IK. Hyaluronic acid modulates proliferation, collagen and protein synthesis of cultured fetal fibroblasts. Matrix 1993;13:441–446. 23. Wulff BC, Parent AE, Meleski MA, DiPietro LA, Schrementi ME, Wilgus TA. Mast cells contribute to scar formation during fetal wound healing. J Invest Dermatol. 2012;132:458–465. 24. Longaker MT, Chiu ES, Harrison MR, et al. Studies in fetal wound healing: IV. Hyaluronic acid-stimulating activity distinguishes fetal wound fluid from adult wound fluid. Ann Surg. 1989;210:667–672. 25. Longaker MT, Whitby DJ, Ferguson MW, Lorenz HP, Harrison MR, Adzick NS. Adult skin wounds in the fetal environment heal with scar formation. Ann Surg. 1994;219:65–72. 26. Alcolea MP, Jones PH. Lineage analysis of epidermal stem cells. Cold Spring Harb Perspect Med. 2014;4:a015206.

Volume 135, Number 3 • Scarless Wound Healing 27. Tadeu AM, Horsley V. Epithelial stem cells in adult skin. Curr Top Dev Biol. 2014;107:109–131. 28. Frede J, Jones PH. Development: Permission to proliferate. Science 2013;342:1183–1184. 29. Buczacki SJ, Zecchini HI, Nicholson AM, et al. Intestinal label-retaining cells are secretory precursors expressing Lgr5. Nature 2013;495:65–69. 30. Tata PR, Mou H, Pardo-Saganta A, et al. Dedifferentiation of committed epithelial cells into stem cells in vivo. Nature 2013;503:218–223. 31. Lu C, Fuchs E. Sweat gland progenitors in development, homeostasis, and wound repair. Cold Spring Harb Perspect Med. 2014;4:a015222. 32. Lu CP, Polak L, Rocha AS, et al. Identification of stem cell populations in sweat glands and ducts reveals roles in homeostasis and wound repair. Cell 2012;150:136–150. 33. Rittié L, Sachs DL, Orringer JS, Voorhees JJ, Fisher GJ. Eccrine sweat glands are major contributors to reepithelialization of human wounds. Am J Pathol. 2013;182:163–171. 34. Biedermann T, Pontiggia L, Böttcher-Haberzeth S, et al. Human eccrine sweat gland cells can reconstitute a stratified epidermis. J Invest Dermatol. 2010;130:1996–2009. 35. Ito M, Liu Y, Yang Z, et al. Stem cells in the hair follicle bulge contribute to wound repair but not to homeostasis of the epidermis. Nat Med. 2005;11:1351–1354. 36. Levy V, Lindon C, Zheng Y, Harfe BD, Morgan BA. Epidermal stem cells arise from the hair follicle after wounding. FASEB J. 2007;21:1358–1366. 37. Nowak JA, Polak L, Pasolli HA, Fuchs E. Hair follicle stem cells are specified and function in early skin morphogenesis. Cell Stem Cell 2008;3:33–43. 38. Snippert HJ, Haegebarth A, Kasper M, et al. Lgr6 marks stem cells in the hair follicle that generate all cell lineages of the skin. Science 2010;327:1385–1389. 39. Brownell I, Guevara E, Bai CB, Loomis CA, Joyner AL. Nervederived sonic hedgehog defines a niche for hair follicle stem cells capable of becoming epidermal stem cells. Cell Stem Cell 2011;8:552–565. 40. Cotsarelis G. Epithelial stem cells: A folliculocentric view. J Invest Dermatol. 2006;126:1459–1468. 41. Arwert EN, Hoste E, Watt FM. Epithelial stem cells, wound healing and cancer. Nat Rev Cancer 2012;12:170–180. 42. Yang R, Zheng Y, Burrows M, et al. Generation of folliculogenic human epithelial stem cells from induced pluripotent stem cells. Nat Commun. 2014;5:3071. 43. Sorrell JM, Caplan AI. Fibroblast heterogeneity: More than skin deep. J Cell Sci. 2004;117:667–675. 44. Jahoda CA, Reynolds AJ. Dermal-epidermal interactions: Adult follicle-derived cell populations and hair growth. Dermatol Clin. 1996;14:573–583. 45. Jahoda CA, Reynolds AJ. Hair follicle dermal sheath cells: Unsung participants in wound healing. Lancet 2001;358:1445–1448. 46. Sorrell JM, Baber MA, Caplan AI. Construction of a bilayered dermal equivalent containing human papillary and reticular dermal fibroblasts: Use of fluorescent vital dyes. Tissue Eng. 1996;2:39–49. 47. Harper RA, Grove G. Human skin fibroblasts derived from papillary and reticular dermis: Differences in growth potential in vitro. Science 1979;204:526–527. 48. Schafer IA, Pandy M, Ferguson R, Davis BR. Comparative observation of fibroblasts derived from the papillary and reticular dermis of infants and adults: Growth kinetics, packing density at confluence and surface morphology. Mech Ageing Dev. 1985;31:275–293. 49. Sorrell JM, Baber MA, Caplan AI. Site-matched papillary and reticular human dermal fibroblasts differ in their release of

specific growth factors/cytokines and in their interaction with keratinocytes. J Cell Physiol. 2004;200:134–145. 50. Driskell RR, Lichtenberger BM, Hoste E, et al. Distinct fibroblast lineages determine dermal architecture in skin development and repair. Nature 2013;504:277–281. 51. Gauglitz GG, Korting HC, Pavicic T, Ruzicka T, Jeschke MG. Hypertrophic scarring and keloids: Pathomechanisms and current and emerging treatment strategies. Mol Med. 2011;17:113–125. 52. Wong VW, Rustad KC, Akaishi S, et al. Focal adhesion kinase links mechanical force to skin fibrosis via inflammatory signaling. Nat Med. 2012;18:148–152. 53. Stephens DJ. Cell biology: Collagen secretion explained. Nature 2012;482:474–475. 54. Akhmetshina A, Palumbo K, Dees C, et al. Activation of canonical Wnt signalling is required for TGF-β-mediated fibrosis. Nat Commun. 2012;3:735. 55. Manuskiatti W, Fitzpatrick RE. Treatment response of keloidal and hypertrophic sternotomy scars: Comparison among intralesional corticosteroid, 5-fluorouracil, and 585nm flashlamp-pumped pulsed-dye laser treatments. Arch Dermatol. 2002;138:1149–1155. 56. Reish RG, Eriksson E. Scar treatments: Preclinical and clinical studies. J Am Coll Surg. 2008;206:719–730. 57. McCoy BJ, Diegelmann RF, Cohen IK. In vitro inhibition of cell growth, collagen synthesis, and prolyl hydroxylase activity by triamcinolone acetonide. Proc Soc Exp Biol Med. 1980;163:216–222. 58. Xu SJ, Teng JY, Xie J, Shen MQ, Chen DM. Comparison of the mechanisms of intralesional steroid, interferon or verapamil injection in the treatment of proliferative scars (in Chinese). Zhonghua Zheng Xing Wai Ke Za Zhi 2009;25:37–40. 59. Stojadinovic O, Lee B, Vouthounis C, et al. Novel genomic effects of glucocorticoids in epidermal keratinocytes: Inhibition of apoptosis, interferon-gamma pathway, and wound healing along with promotion of terminal differentiation. J Biol Chem. 2007;282:4021–4034. 60. D’Andrea F, Brongo S, Ferraro G, Baroni A. Prevention and treatment of keloids with intralesional verapamil. Dermatology 2002;204:60–62. 61. Berman B, Villa AM, Ramirez CC. Novel opportunities in the treatment and prevention of scarring. J Cutan Med Surg. 2004;8(Suppl 3):32–36. 62. Giugliano G, Pasquali D, Notaro A, et al. Verapamil inhibits interleukin-6 and vascular endothelial growth factor production in primary cultures of keloid fibroblasts. Br J Plast Surg. 2003;56:804–809. 63. Saulis AS, Mogford JH, Mustoe TA. Effect of Mederma on hypertrophic scarring in the rabbit ear model. Plast Reconstr Surg. 2002;110:177–183; discussion 184–186. 64. Chung VQ, Kelley L, Marra D, Jiang SB. Onion extract gel versus petrolatum emollient on new surgical scars: Prospective double-blinded study. Dermatol Surg. 2006;32:193–197. 65. Beyer C, Reichert H, Akan H, et al. Blockade of canonical Wnt signalling ameliorates experimental dermal fibrosis. Ann Rheum Dis. 2013;72:1255–1258. 66. Diao JS, Xia WS, Yi CG, et al. Histone deacetylase inhibitor reduces hypertrophic scarring in a rabbit ear model. Plast Reconstr Surg. 2013;132:61e–69e. 67. Tomcik M, Zerr P, Pitkowski J, et al. Heat shock protein 90 (Hsp90) inhibition targets canonical TGF-β signalling to prevent fibrosis. Ann Rheum Dis. 2014;73:1215–1222. 68. Yamaguchi Y, Takihara T, Chambers RA, Veraldi KL, Larregina AT, Feghali-Bostwick CA. A peptide derived from endostatin ameliorates organ fibrosis. Sci Transl Med. 2012;4:136ra71.

915

Plastic and Reconstructive Surgery • March 2015 69. Shi JH, Guan H, Shi S, et al. Protection against TGF-β1induced fibrosis effects of IL-10 on dermal fibroblasts and its potential therapeutics for the reduction of skin scarring. Arch Dermatol Res. 2013;305:341–352. 70. Samarakoon R, Overstreet JM, Higgins PJ. TGF-β signaling in tissue fibrosis: Redox controls, target genes and therapeutic opportunities. Cell Signal. 2013;25:264–268. 71. McCormick LL, Zhang Y, Tootell E, Gilliam AC. Anti-TGFbeta treatment prevents skin and lung fibrosis in murine sclerodermatous graft-versus-host disease: A model for human scleroderma. J Immunol. 1999;163:5693–5699. 72. Shah M, Foreman DM, Ferguson MW. Control of scarring in adult wounds by neutralising antibody to transforming growth factor beta. Lancet 1992;339:213–214. 73. Shah M, Foreman DM, Ferguson MW. Neutralising antibody to TGF-beta 1,2 reduces cutaneous scarring in adult rodents. J Cell Sci. 1994;107:1137–1157. 74. Liu W, Chua CH, Wu XL, et al. Inhibiting scar formation in rat cutaneous wounds by blocking TGF-beta signaling (in Chinese). Zhonghua Yi Xue Za Zhi 2003;83:31–36. 75. Denton CP, Merkel PA, Furst DE, et al.; Cat-192 Study Group; Scleroderma Clinical Trials Consortium. Recombinant human anti-transforming growth factor beta1 antibody therapy in systemic sclerosis: A multicenter, randomized, placebo-controlled phase I/II trial of CAT-192. Arthritis Rheum. 2007;56:323–333. 76. Prey S, Ezzedine K, Doussau A, et al. Imatinib mesylate in scleroderma-associated diffuse skin fibrosis: A phase II multicentre randomized double-blinded controlled trial. Br J Dermatol. 2012;167:1138–1144. 77. Chang Z, Kishimoto Y, Hasan A, Welham NV. TGF-beta3 modulates the inflammatory environment and reduces scar formation following vocal fold mucosal injury in rats. Dis Models Mech. 2014;7:83–91. 78. O’Kane S, Ferguson MW. Transforming growth factor beta s and wound healing. Int J Biochem Cell Biol. 1997;29:63–78. 79. Ohno S, Hirano S, Kanemaru S, et al. Transforming growth factor beta3 for the prevention of vocal fold scarring. Laryngoscope 2012;122:583–589. 80. Shah M, Foreman DM, Ferguson MW. Neutralisation of TGF-beta 1 and TGF-beta 2 or exogenous addition of TGFbeta 3 to cutaneous rat wounds reduces scarring. J Cell Sci. 1995;108:985–1002. 81. Horn A, Kireva T, Palumbo-Zerr K, et al. Inhibition of hedgehog signalling prevents experimental fibrosis and induces regression of established fibrosis. Ann Rheum Dis. 2012;71:785–789. 82. Nakagome K, Dohi M, Okunishi K, Tanaka R, Miyazaki J, Yamamoto K. In vivo IL-10 gene delivery attenuates bleomycin induced pulmonary fibrosis by inhibiting the production and activation of TGF-beta in the lung. Thorax 2006;61:886–894. 83. Occleston NL, O’Kane S, Goldspink N, Ferguson MW. New therapeutics for the prevention and reduction of scarring. Drug Discov Today 2008;13:973–981. 84. Reitamo S, Remitz A, Tamai K, Uitto J. Interleukin-10 modulates type I collagen and matrix metalloprotease gene expression in cultured human skin fibroblasts. J Clin Invest. 1994;94:2489–2492. 85. Yamamoto T, Eckes B, Krieg T. Effect of interleukin-10 on the gene expression of type I collagen, fibronectin, and decorin in human skin fibroblasts: Differential regulation by transforming growth factor-beta and monocyte chemoattractant protein-1. Biochem Biophys Res Commun. 2001;281:200–205. 86. Yuan W, Varga J. Transforming growth factor-beta repression of matrix metalloproteinase-1 in dermal fibroblasts involves Smad3. J Biol Chem. 2001;276:38502–38510.

916

87. Galiano RD, Michaels J V, Dobryansky M, Levine JP, Gurtner GC. Quantitative and reproducible murine model of excisional wound healing. Wound Repair Regen. 2004;12:485–492. 88. Aarabi S, Bhatt KA, Shi Y, et al. Mechanical load initiates hypertrophic scar formation through decreased cellular apoptosis. FASEB J. 2007;21:3250–3261. 89. Shridharani SM, Tufaro AP. A systematic review of acellular dermal matrices in head and neck reconstruction. Plast Reconstr Surg. 2012;130(Suppl 2):35S–43S. 90. Melendez MM, Martinez RR, Dagum AB, et al. Porcine wound healing in full-thickness skin defects using Integra with and without fibrin glue with keratinocytes. Can J Plast Surg. 2008;16:147–152. 91. Hilmi AB, Halim AS, Hassan A, Lim CK, Noorsal K, Zainol I. In vitro characterization of a chitosan skin regenerating template as a scaffold for cells cultivation. Springerplus 2013;2:79. 92. Wang HM, Chou YT, Wen ZH, et al. Novel biodegradable porous scaffold applied to skin regeneration. PLoS One 2013;8:e56330. 93. Wong VW, Rustad KC, Galvez MG, et al. Engineered pullulancollagen composite dermal hydrogels improve early cutaneous wound healing. Tissue Eng Part A 2011;17:631–644. 94. Rustad KC, Wong VW, Gurtner GC. The role of focal adhesion complexes in fibroblast mechanotransduction during scar formation. Differentiation 2013;86:87–91. 95. Silver FH, Siperko LM, Seehra GP. Mechanobiology of force transduction in dermal tissue. Skin Res Technol. 2003;9:3–23. 96. Atkinson JA, McKenna KT, Barnett AG, McGrath DJ, Rudd M. A randomized, controlled trial to determine the efficacy of paper tape in preventing hypertrophic scar formation in surgical incisions that traverse Langer’s skin tension lines. Plast Reconstr Surg. 2005;116:1648–1656; discussion 1657. 97. Rosengren H, Askew DA, Heal C, Buettner PG, Humphreys WO, Semmens LA. Does taping torso scars following dermatologic surgery improve scar appearance? Dermatol Pract Concept. 2013;3:75–83. 98. Fulton JE Jr. Silicone gel sheeting for the prevention and management of evolving hypertrophic and keloid scars. Dermatol Surg. 1995;21:947–951. 99. Puri N, Talwar A. The efficacy of silicone gel for the treatment of hypertrophic scars and keloids. J Cutan Aesthet Surg. 2009;2:104–106. 100. Sawada Y, Sone K. Hydration and occlusion treat ment for hypertrophic scars and keloids. Br J Plast Surg. 1992;45:599–603. 101. Tollefson TT, Kamangar F, Aminpour S, Lee A, DurbinJohnson B, Tinling S. Comparison of effectiveness of silicone gel sheeting with microporous paper tape in the prevention of hypertrophic scarring in a rabbit model. Arch Facial Plast Surg. 2012;14:45–51. 102. Doumit J, Vale R, Kanigsberg N. Dynaclose tape: A practical alternative to punch biopsy site closure. J Cutan Med Surg. 2013;17:62–65. 103. Chen HH, Tsai WS, Yeh CY, Wang JY, Tang R. Prospective study comparing wounds closed with tape with sutured wounds in colorectal surgery. Arch Surg. 2001;136:801–803. 104. van de Gevel DF, Hamad MA, Elenbaas TW, Ostertag JU, Schonberger JP. Is the use of Steri-StripTM S for wound closure after coronary artery bypass grafting better than intracuticular suture? Interact Cardiovasc Thorac Surg. 2010;10:561–564. 105. Akaishi S, Akimoto M, Hyakusoku H, Ogawa R. The tensile reduction effects of silicone gel sheeting. Plast Reconstr Surg. 2010;126:109e–111e. 106. Mustoe TA. Evolution of silicone therapy and mechanism of action in scar management. Aesthetic Plast Surg. 2008;32:82–92.

Volume 135, Number 3 • Scarless Wound Healing 107. Quinn KJ. Silicone gel in scar treatment. Burns Incl Therm Inj. 1987;13(Suppl):S33–S40. 108. Ahn ST, Monafo WW, Mustoe TA. Topical silicone gel: A new treatment for hypertrophic scars. Surgery 1989;106:781–786; discussion 786. 109. Chang CC, Kuo YF, Chiu HC, Lee JL, Wong TW, Jee SH. Hydration, not silicone, modulates the effects of keratinocytes on fibroblasts. J Surg Res. 1995;59:705–711. 110. Kuhn MA, Moffit MR, Smith PD, et al. Silicone sheeting decreases fibroblast activity and downregulates TGFbeta2 in hypertrophic scar model. Int J Surg Investig. 2001;2:467–474. 111. Ricketts CH, Martin L, Faria DT, Saed GM, Fivenson DP. Cytokine mRNA changes during the treatment of

FILLER-032

hypertrophic scars with silicone and nonsilicone gel dressings. Dermatol Surg. 1996;22:955–959. 112. Gurtner GC, Dauskardt RH, Wong VW, et al. Improving cutaneous scar formation by controlling the mechanical environment: Large animal and phase I studies. Ann Surg. 2011;254:217–225. 113. Longaker MT, Rohrich RJ, Greenberg L, et al. A randomized controlled trial of the embrace advanced scar therapy device to reduce incisional scar formation. Plast Reconstr Surg. 2014;134:536–546. 114. Lim AF, Weintraub J, Kaplan EN, et al. The embrace device significantly decreases scarring following scar revision surgery in a randomized controlled trial. Plast Reconstr Surg. 2014;133:398–405.

917

Scarless wound healing: chasing the holy grail.

Over 100 million patients acquire scars in the industrialized world each year, primarily as a result of elective operations. Although undefined, the g...
703KB Sizes 2 Downloads 40 Views