Skip to main content

REVIEW article

Front. Cell Dev. Biol., 06 January 2022
Sec. Stem Cell Research
This article is part of the Research Topic Tissue Stem Cells during Trauma: from Basic Biology to Translational Medicine View all 13 articles

Use of Adipose Stem Cells Against Hypertrophic Scarring or Keloid

Hongbo Chen&#x;Hongbo ChenKai Hou&#x;Kai HouYiping WuYiping WuZeming Liu
Zeming Liu*
  • Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China

Hypertrophic scars or keloid form as part of the wound healing reaction process, and its formation mechanism is complex and diverse, involving multi-stage synergistic action of multiple cells and factors. Adipose stem cells (ASCs) have become an emerging approach for the treatment of many diseases, including hypertrophic scarring or keloid, owing to their various advantages and potential. Herein, we analyzed the molecular mechanism of hypertrophic scar or keloid formation and explored the role and prospects of stem cell therapy, in the treatment of this condition.

Introduction

Hypertrophic scars are abnormal fibroproliferative wound healing reactions (Zouboulis and Orfanos, 1990; Limandjaja et al., 2020), hypertrophic scars can continue to develop into keloids, keloids are lesions that are higher than the skin surface and beyond the original injury range. The texture of the keloid becomes hard and thickened, presenting nodular or flake masses. Hypertrophic scars and keloid are characterized by excessive scar tissue formation and invasive growth beyond the original wound boundary. Hypertrophic scarring is an unpleasant, maladaptive comorbidity that affects most people around the world and imposes a heavy social and economic burden on affected parties.

Scars usually occur on specific body sites, such as the anterior chest, ear-lobe, mandibular border, and the suprapubic region (Huang and Ogawa, 2021). In addition to obvious cosmetic disfigurement, hypertrophic scars can also produce symptoms such as itching, pain, contracture, and movement restriction, resulting in serious impairment of emotional health and reduced quality of life (Bijlard et al., 2017; Balci et al., 2009). Hypertrophic scars can be acquired genetically or pathologically. Histologically, hypertrophic scars are benign hyperplastic disorder caused by excessive accumulation of extracellular matrix (ECM) and other components (Feng et al., 2017).

The incidence of hypertrophic scarring varies widely and is known to correlate with race and ethnicity. Incidence rates are reportedly as low as <1.0% in Taiwanese Chinese and Caucasians (Seifert and Mrowietz, 2009; Sun et al., 2014), and range from 4.5 to 16.0% in the black and Hispanic general population (Rockwell et al., 1989; Hunasgi et al., 2013). More recent data are available for specific subgroups, reporting that the incidence of hypertrophic scar formation increases significantly in African Americans compared to Caucasians, Asians and other groups, after head and neck surgery, and in women, following cesarean section (C-section) (Tulandi et al., 2011; Sun et al., 2014). Overcoming the complications, and social and economic burden caused by hypertrophic scarring, has become an important medical research topic.

Mechanism of Scar Formation

Scar formation is part of the abnormal wound healing response after non-severe trauma, such as caesarean section incision, chest hair folliculitis, ear piercing and vaccination. The potential mechanism of scar is complicated, involves genetic susceptibility, mechanobiology, endocrine factors, infection, excessive inflammatory response and so on (Huang and Ogawa, 2021; Yu et al., 2021, Riccio et al., 2019b). Scar formation include multiple cell factors and synergy between multiple stages of repair.

The stages of wound healing mainly comprise inflammation, proliferation, and reshaping (Mari et al., 2015; Leavitt et al., 2016; Morikawa et al., 2019; Rodrigues et al., 2019; Tan et al., 2019; De Francesco et al., 2020). When skin lesions occur, the first reaction entails the formation of a platelet plug, followed by clot formation that immediately stems the bleeding. Then, damaged tissue and activated platelets initiate an inflammatory response by recruiting immune cells such as neutrophils and monocytes to fight local infection, engulf localized debris and damaged connective tissue, and subsequently remove fibrin. After the inflammation subsides, the hyperplasia phase begins, during which new blood vessels and connective tissue appear in the wound area. Re-epithelialization, driven by keratinocyte migration, marks the beginning of proliferation. Thereafter, organizational maturity signifies the reshaping phase, which involves the degeneration of neovascularization and concomitant reconstruction of the extracellular matrix (ECM), resulting in development of organized collagen fibrils that serve as the basis of normal scarring (Wang et al., 2020).

Interference with any of these processes can lead to poor or excessive wound healing, resulting in the formation of hypertrophic scars and non-malignant dermal tumors, which share similar phenotypes, cellular bioenergetics, epigenetic methylation, and other characteristics. Platelet-derived growth factor (PDGF), transforming growth factor-β (TGF-β) and other factors can lead to fibroblast dysfunction and phenotypic changes, ECM disorder and repeated deposition, and an unbalanced angiogenesis cascade, ultimately causing excessive scarring (Tan et al., 2019; Wang et al., 2020; Putri and Prasetyono, 2021).

Genetic Variation, Epigenetic Modifications and Scar

Single nucleotide polymorphisms have also been implicated in keloid formation, and genetic studies have identified several SNPS and genes that may contribute to understanding the association with keloid development (Halim et al., 2012, Tsai and Ogawa, 2019). For example, a genome-wide association study (GWAS) by Nakashima et al. found that four SNPS (rs873549, rs1511412, rs940187 and rs8032158) in three chromosomal regions were significantly associated with keloid (Nakashima et al., 2010). In other study, Ogawa et al. also reported that rs8032158 may also influence keloid development (Halim et al., 2012).

Recent studies suggest that epigenetic inheritance may also contribute to keloid formation. For example, DNA methylation can change the structure of DNA, thus affecting cell differentiation and cell phenotype (Bataille et al., 2012). Other epigenetic changes include changes in cell phenotypes by popular non-coding RNA. Epigenetic modifications and DNA methylation caused by non-coding RNA (such as microRNAs and LncRNAs) may also play an important role by inducing sustained activation of keloid fibroblasts (Tsai and Ogawa, 2019).

Mechanical Stress and Scar

The two main anatomical layers of skin, from top to bottom, are the epidermis and dermis. The epidermis consists mainly of epithelial keratinocytes, which are forced together by actin (Harn et al., 2016). These keratinocytes form the skin’s barrier and are layered on top of the dermis, acting as building blocks against external shearing and stretching forces. In contrast, the dermis contains mostly ECM, blood vessels, fibroblasts, and other mesenchymal cells, rather than epithelial cells. Mesenchymal cells, such as fibroblasts, exhibit more cell-matrix interactions (Tracy et al., 2016; Harn et al., 2019; Watt and Fujiwara, 2011).

Mechanical forces regulate skin homeostasis and play a role in the pathogenesis of skin diseases (Fu et al., 2021). As mention, the dermis is rich in ECM—especially collagen, secreted by fibroblasts—that provides the bulk of the skin’s tension (Hsu et al., 2018). Changes in external tension and the internal mechanical properties of cells are associated with collagen, and scarring is specifically the result of excessive collagen production. To some extent, the mechanical stiffness of the skin environment determines the regenerative ability of wound healing. Strategies that alter mechanical forces or mechanical transduction signals may provide new approaches for treating skin diseases and promoting skin regeneration (Harn et al., 2019).

Immunological Aspects of Scar

Scarring and autoimmune skin disease represent fibrosis events that require a variety of growth factors and result from synergy between immune cells and the medium. These conditions share certain features, such as high infiltration of immune cells and immunoglobulin (Ig), as well as complement deposition in the scar tissue and scar fibroblasts, and medium-high secretion of immune factors, among others (Dong et al., 2013; Jiao et al., 2015; Ogawa, 2017).

Scar can be classified as inflammatory, nodular, and mixed types (Yu et al., 2021). The number and type of immune cells in a patient’s scar tissue or peripheral blood may vary by type and stage. Moreover, the formation of a scar may be related to abnormal wound healing and related immune factors, especially in repeatedly recurring scars. Hence, the classification of scar types and stages provides direction for future research. In the process of scar formation, the initial activation of mast cells (MCs) is the key factor inducing chronic inflammation. Further, MCs release mediators that initiate host defense cascades leading to fibrotic processes. (Hsu et al., 2018; Tsai and Ogawa, 2019).

Current Mainstream Strategies for Inhibiting Scar Formation

Therapeutic strategies for treating hypertrophic scars include steroid injection, surgical hypertrophic scar removal, radiotherapy, compression, and cryotherapy, pulsed dye and CO2 laser, silicone gel, nanoparticles and radiotherapy, among others (Salameh et al., 2021; Ogawa, 2021; Svolacchia et al., 2016; Nicoletti et al., 2013; Riccio et al., 2019a; Jimi et al., 2020; Barrera, 2003; Aoki et al., 2020). However, existing treatments are not guaranteed to reduce recurrence of keloids, due to the high recurrence rate thereof at the hypertrophic scar excision site (Coentro et al., 2019; Kumar and Kamalasanan, 2021).

Scar-free healing is the ultimate goal of scar prevention strategies (Falanga, 2005; Ho Jeong, 2010; Heng, 2011; Leavitt et al., 2016). To develop an optimal combination strategy to this effect, it is necessary to fully reveal the causal relationship between key cells and molecules in scar pathogenesis, thus providing new therapeutic targets for scar-free healing. In addition, advances in stem cell and tissue engineering have brought more alternative therapies closer to reality. Functionalization of biomaterials through various drugs and growth factors, provides a well-controlled approach to scar therapy (Fuller et al., 2016; Rahimnejad et al., 2017; Wang et al., 2022).

Mesenchymal Stem Cells and Adipogenic Stem Cells

Mesenchymal stem cells (MSCs) are primarily responsible for the regeneration of damaged cells and can be obtained from a variety of sources, including bone marrow, umbilical cord, and adipose tissue. MSCs are abundant and have recently been attracting increasing research attention as potential treatment options in many diseases. The basic characteristics of MSCs are defined by the International Society for Cell & Gene Therapy (ISCT), as follows: 1) plasticity when cultured under standard conditions; 2) expression of a unique set of surface antigens specified by the ISCT; 3) ability to differentiate into osteoblasts, chondrocytes, and adipose cells (Dominici et al., 2006; De Francesco et al., 2017; Bougioukli et al., 2018).

Adipogenic stem cells (ASCs) are bone marrow MSCs extracted from fat cells (53). Not only do they have the general characteristics of bone marrow MSCs, but they are also easy to obtain. ASCs are found in large numbers in the human body, have high proliferation and self-renewal potential, and other advantages, based on which numerous studies have shown their potential in addressing many diseases, including hypertrophic scarring (Gir et al., 2012; Lee et al., 2012; Strong et al., 2015; O’Halloran et al., 2017; Gardin et al., 2018; Gentile and Garcovich, 2019; Ren et al., 2019; Xiong et al., 2020).

Discussion of Adipogenic Stem Cells and Hypertrophic Scarring

Existing studies have shown that ASCs’ mechanism of action entails secretion of bioactive factors (including growth factors and cytokines, among others), on the one hand, to promote cell proliferation, differentiation, and migration. On the other hand, exosomes derived from ASCs play a role (Hu et al., 2016; Li et al., 2018; Han et al., 2019; Kucharzewski et al., 2019). Moreover, ASCs can play a direct part in disease management, through their multidirectional differentiation ability, by forming complex hybrid systems with novel treatment materials.

Directed Differentiation

ASCs are pluripotent stem cells with the ability of self-renewal and multidirectional differentiation. They are not only the precursors of fat cells, but also capable of differentiating into osteoblasts, chondrocytes, muscle cells, and neuronal cells. ASCs have also been shown to differentiate into keratinocytes. These results suggest that ASCs may also differentiate directly into epidermal and dermal cells to promote tissue regeneration and prevent scarring in an injured area, during wound healing (Joshi et al., 2020; Xiong et al., 2020; Putri and Prasetyono, 2021).

Secretion of Biologically Active Factors

ASCs regulate inflammation facilitated by immune cells, through paracrine bioactive factors, inhibiting scar hyperplasia. Long-term inflammation is a main cause of hypertrophic scar formation. ASCs are known to modulate the activity of inflammatory cells, thereby attenuating periods of excessive and prolonged inflammation. This immunomodulatory activity of ASCs is conducted by paracrine secretion of several anti-fibrotic cytokines, including prostaglandin E2 (PGE2), interleukin (IL)-10, hepatocyte growth factor (HGF), and nitric oxide (NO) (Seo and Jung, 2016). In contrast, mast cells are involved in the regulation of vascular homeostasis and angiogenesis and can upregulate fibroblast proliferation, resulting in excessive collagen synthesis and differentiation of fibroblasts into myofibroblasts. ASCs can further reduce hypertrophic scarring by inhibiting the number and activity of mast cells (Putri and Prasetyono, 2021).

ASCs can also activate various anti-fibrotic molecular pathways through paracrine signaling, regulate the activity of primary fibroblast transforming growth factor, and stabilize the function of fibroblast and keratinocyte receptor sites, to achieve relevant anti-fibrosis goals (Borovikova et al., 2018). In addition, injection of ASCs can reverse the abnormal vascularization pattern of scar tissue and reshape the microvascular structure (Garza et al., 2014; Luan et al., 2016).

Release of Adipogenic Stem Cell-Derived Exosomes (Adipogenic Stem Cell-Exos)

Exosomes are a subset of small, membranous extracellular vesicles (Han et al., 2018; Jing et al., 2018) with a diameter of approximately 30–150 nm, which originate from entosis and are distributed in many bodily fluids—including blood, urine, cerebrospinal fluid, and bile—under physiological and pathological conditions. Exosomes include a variety of active biological substances such as proteins, DNA, mRNAs, and microRNAs, which can carry complex biological information and release it into target cells (Hessvik et al., 2016; Buratta et al., 2020). In addition, exosomes may exist between MSCs and target cells as paracrine mediators (Valadi et al., 2007; Chiba et al., 2012). Many studies have shown that ASC-derived exosomes (ASC-Exos) play an important role in cell migration, proliferation, and collagen synthesis (Xiong et al., 2020; Putri and Prasetyono, 2021).

For example, characteristics of scarring include collagen receptor rearrangement, activation of myofibroblasts expressing α-smooth muscle actin (α-SMA), and the production and secretion of high levels of TGF-β1 in affected tissues (Beanes et al., 2003). Xu et al. indirectly inhibited PLOD1 levels in ASCs through post-transcriptional regulation, which may significantly improve the anti-corrosion potential of ASCs during wound healing, by changing macrophage polarization and regulating scar formation (Xu et al., 2021). In addition, Wang et al. found that ASC-Exos reduced scar formation mainly by regulating the ratios of type III:type I collagen, TGF-β3:TGF-β1, matrix metalloproteinases-3 (MMP-3):tissue inhibitor of metalloproteinases-1 (TIMP-1), and promoting human dermal fibroblast HDF differentiation. This resulted in improved ECM reconstruction and other implementations (Wang et al., 2017).

Formation of Complex Hybrid Systems

ASCs can play a role in disease management by forming complex hybrid systems with novel materials. For example, Wang et al. proved that ASCs are capable of multi-potential—including lipogenic, osteogenic, and chondrogenic—differentiation, and developed ASC-Exos-delivering collagen/poly (L-lactide-co-caprolactone) (P (LLA-Cl)) nanoyarns, which conform to a material system that mimics the morphological structure of the natural tissue matrix. With sufficient biocompatibility and mechanical properties, it can effectively promote neovascularization, cell proliferation and tissue regeneration, and simultaneously limit scar formation, collagen deposition, and formation of multi-layer epithelium (Wang et al., 2022). Liu et al. showed that hyaluronic acid (HA) can be used as immobilizing agent in a constructed system of ASC-Exos with HA, to retain exosomes in the wound area and effectively play a role in wound repair. The system activated wound-based HDF activity and increased re-epithelialization, which was expected to reduce scar formation (Liu et al., 2019). In addition, Wang et al. developed an FHE hydrogel-carrier system with stimulus-responsive ASC-Exos, which significantly increased the regeneration of skin appendages and reduced scar tissue formation (Wang et al., 2019). Hector et al. also identified three types of ECM-based biomaterials—Integra™ Matrix Wound Dressing, XenoMEM™, and MatriStem™—that serve as human ASC delivery vehicles, which could inhibit scar formation (Capella-Monsonís et al., 2020).

Conclusion and Future Direction

ASCs have the advantages of being derived from a large source, offering abundant availability and high proliferative ability, having low immunogenicity in clinical applications, and being safer and more effective. ASCs histologically promote the regeneration of healthy tissue, reduce fibroblasts, and reconstruct collagen, similar to that of normal skin. At the molecular level, ASCs reduce hypertrophic scarring through direct differentiation and paracrine mechanisms. Clinically, they can improve the color, elasticity, texture, thickness, and size of hypertrophic scars. ASCs have a positive effect on alleviating hypertrophic scarring, indicating their potential as a possible treatment approach in this condition, with broad therapeutic prospect.

Author Contributions

All authors contributed to the design of the study and writing of the manuscript. KH and HC undertook the research, YW and HC wrote the main manuscript text and prepared figures. ZL revised the article critically for important intellectual content and final approval of the version to be submitted. All authors reviewed the manuscript.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Abbreviations

ASCs, adipose stem cells; ECM, extracellular matrix; HGF, hepatocyte growth factor; ISCT, International Society for Cell & Gene Therapy; IL-10, interleukin-10; mscMSCs, Mesenchymal stem cells; NO, nitric oxide; PDGF, Platelet-derived growth factor; PGE2, prostaglandin E2; TGF-β, transforming growth factor-β.

References

Aoki, M., Matsumoto, N. M., Dohi, T., Kuwahawa, H., Akaishi, S., Okubo, Y., et al. (2020). Direct Delivery of Apatite Nanoparticle-Encapsulated siRNA Targeting TIMP-1 for Intractable Abnormal Scars. Mol. Ther. - Nucleic Acids 22, 50–61. doi:10.1016/j.omtn.2020.08.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Balci, D. D., Inandi, T., Dogramaci, C. A., and Celik, E. (2009). DLQI Scores in Patients with Keloids and Hypertrophic Scars: a Prospective Case Control Study. J. Dtsch Dermatol. Ges 7 (8), 688–691. doi:10.1111/j.1610-0387.2009.07034.x

CrossRef Full Text | Google Scholar

Barrera, A. (2003). The Use of Micrografts and Minigrafts in the Aesthetic Reconstruction of the Face and Scalp. Plast. Reconstr. Surg. 112 (3), 883–890. doi:10.1097/01.PRS.0000072253.54359.7F

PubMed Abstract | CrossRef Full Text | Google Scholar

Bataille, V., Lens, M., and Spector, T. D. (2012). The Use of the Twin Model to Investigate the Genetics and Epigenetics of Skin Diseases with Genomic, Transcriptomic and Methylation Data. J. Eur. Acad. Dermatol. Venereol. 26 (9), 1067–1073. doi:10.1111/j.1468-3083.2011.04444.x

CrossRef Full Text | Google Scholar

Beanes, S. R., Dang, C., Soo, C., and Ting, K. (2003). Skin Repair and Scar Formation: the central Role of TGF-β. Expert Rev. Mol. Med. 5 (8), 1–22. doi:10.1017/S1462399403005817

CrossRef Full Text | Google Scholar

Bijlard, E., Kouwenberg, C., Timman, R., Hovius, S., Busschbach, J., and Mureau, M. (2017). Burden of Keloid Disease: A Cross-Sectional Health-Related Quality of Life Assessment. Acta Derm Venerol 97 (2), 225–229. doi:10.2340/00015555-2498

PubMed Abstract | CrossRef Full Text | Google Scholar

Borovikova, A. A., Ziegler, M. E., Banyard, D. A., Wirth, G. A., Paydar, K. Z., Evans, G. R. D., et al. (2018). Adipose-Derived Tissue in the Treatment of Dermal Fibrosis. Ann. Plast. Surg. 80 (3), 297–307. doi:10.1097/SAP.0000000000001278

PubMed Abstract | CrossRef Full Text | Google Scholar

Bougioukli, S., Sugiyama, O., Pannell, W., Ortega, B., Tan, M. H., Tang, A. H., et al. (2018). Gene Therapy for Bone Repair Using Human Cells: Superior Osteogenic Potential of Bone Morphogenetic Protein 2-Transduced Mesenchymal Stem Cells Derived from Adipose Tissue Compared to Bone Marrow. Hum. Gene Ther. 29 (4), 507–519. doi:10.1089/hum.2017.097

PubMed Abstract | CrossRef Full Text | Google Scholar

Buratta, S., Tancini, B., Sagini, K., Delo, F., Chiaradia, E., Urbanelli, L., et al. (2020). Lysosomal Exocytosis, Exosome Release and Secretory Autophagy: The Autophagic- and Endo-Lysosomal Systems Go Extracellular. Ijms 21 (7), 2576. doi:10.3390/ijms21072576

PubMed Abstract | CrossRef Full Text | Google Scholar

Capella-Monsonís, H., De Pieri, A., Peixoto, R., Korntner, S., and Zeugolis, D. I. (2020). Extracellular Matrix-Based Biomaterials as Adipose-Derived Stem Cell Delivery Vehicles in Wound Healing: a Comparative Study between a Collagen Scaffold and Two Xenografts. Stem Cel Res Ther 11 (1), 510. doi:10.1186/s13287-020-02021-x

CrossRef Full Text | Google Scholar

Chiba, M., Kimura, M., and Asari, S. (2012). Exosomes Secreted from Human Colorectal Cancer Cell Lines Contain mRNAs, microRNAs and Natural Antisense RNAs, that Can Transfer into the Human Hepatoma HepG2 and Lung Cancer A549 Cell Lines. Oncol. Rep. 28 (5), 1551–1558. doi:10.3892/or.2012.1967

PubMed Abstract | CrossRef Full Text | Google Scholar

Coentro, J. Q., Pugliese, E., Hanley, G., Raghunath, M., and Zeugolis, D. I. (2019). Current and Upcoming Therapies to Modulate Skin Scarring and Fibrosis. Adv. Drug Deliv. Rev. 146, 37–59. doi:10.1016/j.addr.2018.08.009

PubMed Abstract | CrossRef Full Text | Google Scholar

De Francesco, F., Busato, A., Mannucci, S., Zingaretti, N., Cottone, G., Amendola, F., et al. (2020). Artificial Dermal Substitutes for Tissue Regeneration: Comparison of the Clinical Outcomes and Histological Findings of Two Templates. J. Int. Med. Res. 48 (8), 030006052094550. doi:10.1177/0300060520945508

CrossRef Full Text | Google Scholar

De Francesco, F., Guastafierro, A., Nicoletti, G., Razzano, S., Riccio, M., and Ferraro, G. (2017). The Selective Centrifugation Ensures a Better In Vitro Isolation of ASCs and Restores a Soft Tissue Regeneration In Vivo. Ijms 18 (5), 1038. doi:10.3390/ijms18051038

CrossRef Full Text | Google Scholar

Dominici, M., Le Blanc, K., Mueller, I., Slaper-Cortenbach, I., Marini, F. C., Krause, D. S., et al. (2006). Minimal Criteria for Defining Multipotent Mesenchymal Stromal Cells. The International Society for Cellular Therapy Position Statement. Cytotherapy 8 (4), 315–317. doi:10.1080/14653240600855905

PubMed Abstract | CrossRef Full Text | Google Scholar

Dong, X., Mao, S., and Wen, H. (2013). Upregulation of Proinflammatory Genes in Skin Lesions May Be the Cause of Keloid Formation (Review). Biomed. Rep. 1 (6), 833–836. doi:10.3892/br.2013.169

PubMed Abstract | CrossRef Full Text | Google Scholar

Falanga, V. (2005). Wound Healing and its Impairment in the Diabetic Foot. The Lancet 366 (9498), 1736–1743. doi:10.1016/S0140-6736(05)67700-8

CrossRef Full Text | Google Scholar

Feng, J., Xue, S., Pang, Q., Rang, Z., and Cui, F. (2017). miR-141-3p Inhibits Fibroblast Proliferation and Migration by Targeting GAB1 in Keloids. Biochem. Biophysical Res. Commun. 490 (2), 302–308. doi:10.1016/j.bbrc.2017.06.040

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, S., Panayi, A., Fan, J., Mayer, H. F., Daya, M., Khouri, R. K., et al. (2021). Mechanotransduction in Wound Healing: From the Cellular and Molecular Level to the Clinic. Adv. Skin Wound Care 34 (2), 67–74. doi:10.1097/01.ASW.0000725220.92976.a7

PubMed Abstract | CrossRef Full Text | Google Scholar

Fuller, K. P., Gaspar, D., Delgado, L. M., Pandit, A., and Zeugolis, D. I. (2016). Influence of Porosity and Pore Shape on Structural, Mechanical and Biological Properties of Poly ϵ-caprolactone Electro-Spun Fibrous Scaffolds. Nanomedicine 11 (9), 1031–1040. doi:10.2217/nnm.16.21

PubMed Abstract | CrossRef Full Text | Google Scholar

Gardin, C., Ferroni, L., Bellin, G., Rubini, G., Barosio, S., and Zavan, B. (2018). Therapeutic Potential of Autologous Adipose-Derived Stem Cells for the Treatment of Liver Disease. Ijms 19 (12), 4064. doi:10.3390/ijms19124064

PubMed Abstract | CrossRef Full Text | Google Scholar

Garza, R. M., Paik, K. J., Chung, M. T., Duscher, D., Gurtner, G. C., Longaker, M. T., et al. (2014). Studies in Fat Grafting. Plast. Reconstr. Surg. 134 (2), 249–257. doi:10.1097/PRS.0000000000000326

PubMed Abstract | CrossRef Full Text | Google Scholar

Gentile, P., and Garcovich, S. (2019). Concise Review: Adipose-Derived Stem Cells (ASCs) and Adipocyte-Secreted Exosomal microRNA (A-SE-miR) Modulate Cancer Growth and proMote Wound Repair. Jcm 8 (6), 855. doi:10.3390/jcm8060855

PubMed Abstract | CrossRef Full Text | Google Scholar

Gir, P., Oni, G., Brown, S. A., Mojallal, A., and Rohrich, R. J. (2012). Human Adipose Stem Cells. Plast. Reconstr. Surg. 129 (6), 1277–1290. doi:10.1097/PRS.0b013e31824ecae6

PubMed Abstract | CrossRef Full Text | Google Scholar

Halim, A. S., Emami, A., Salahshourifar, I., and Kannan, T. P. (2012). Keloid Scarring: Understanding the Genetic Basis, Advances, and Prospects. Arch. Plast. Surg. 39 (3), 184–189. doi:10.5999/aps.2012.39.3.184

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, Y., Jia, L., Zheng, Y., and Li, W. (2018). Salivary Exosomes: Emerging Roles in Systemic Disease. Int. J. Biol. Sci. 14 (6), 633–643. doi:10.7150/ijbs.25018

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, Y., Sun, T., Han, Y., Lin, L., Liu, C., Liu, J., et al. (2019). Human Umbilical Cord Mesenchymal Stem Cells Implantation Accelerates Cutaneous Wound Healing in Diabetic Rats via the Wnt Signaling Pathway. Eur. J. Med. Res. 24 (1), 10. doi:10.1186/s40001-019-0366-9

CrossRef Full Text | Google Scholar

Harn, H. I. C., Hsu, C. K., Wang, Y. K., Huang, Y. W., Chiu, W. T., Lin, H. H., et al. (2016). Spatial Distribution of Filament Elasticity Determines the Migratory Behaviors of a Cell. Cell Adhes. Migration 10 (4), 368–377. doi:10.1080/19336918.2016.1156825

PubMed Abstract | CrossRef Full Text | Google Scholar

Harn, H. I. C., Ogawa, R., Hsu, C. K., Hughes, M. W., Tang, M. J., and Chuong, C. M. (2019). The Tension Biology of Wound Healing. Exp. Dermatol. 28 (4), 464–471. doi:10.1111/exd.13460

PubMed Abstract | CrossRef Full Text | Google Scholar

Heng, M. C. Y. (2011). Wound Healing in Adult Skin: Aiming for Perfect Regeneration. Int. J. Dermatol. 50 (9), 1058–1066. doi:10.1111/j.1365-4632.2011.04940.x

CrossRef Full Text | Google Scholar

Hessvik, N. P., Øverbye, A., Brech, A., Torgersen, M. L., Jakobsen, I. S., Sandvig, K., et al. (2016). PIKfyve Inhibition Increases Exosome Release and Induces Secretory Autophagy. Cell. Mol. Life Sci. 73 (24), 4717–4737. doi:10.1007/s00018-016-2309-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Ho Jeong, J. (2010). Adipose Stem Cells and Skin Repair. Cscr 5 (2), 137–140. doi:10.2174/157488810791268690

CrossRef Full Text | Google Scholar

Hsu, C. K., Lin, H. H., Harn, H. I. C., Hughes, M. W., Tang, M. J., and Yang, C. C. (2018). Mechanical Forces in Skin Disorders. J. Dermatol. Sci. 90 (3), 232–240. doi:10.1016/j.jdermsci.2018.03.004

CrossRef Full Text | Google Scholar

Hu, L., Wang, J., Zhou, X., Xiong, Z., Zhao, J., Yu, R., et al. (2016). Exosomes Derived from Human Adipose Mensenchymal Stem Cells Accelerates Cutaneous Wound Healing via Optimizing the Characteristics of Fibroblasts. Sci. Rep. 6, 32993. doi:10.1038/srep32993

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, C., and Ogawa, R. (2021). Keloidal Pathophysiology: Current Notions. Scars, Burns & Healing 7, 205951312098032. doi:10.1177/2059513120980320

CrossRef Full Text | Google Scholar

Hunasgi, S., Koneru, A., Vanishree, M., and Shamala, R. (2013). Keloid: A Case Report and Review of Pathophysiology and Differences between Keloid and Hypertrophic Scars. J. Oral Maxillofac. Pathol. 17 (1), 116–120. doi:10.4103/0973-029X.110701

CrossRef Full Text | Google Scholar

Jiao, H., Fan, J., Cai, J., Pan, B., Yan, L., Dong, P., et al. (2015). Analysis of Characteristics Similar to Autoimmune Disease in Keloid Patients. Aesth Plast. Surg. 39 (5), 818–825. doi:10.1007/s00266-015-0542-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Jimi, S., Takagi, S., De Francesco, F., Miyazaki, M., and Saparov, A. (2020). Acceleration of Skin Wound-Healing Reactions by Autologous Micrograft Tissue Suspension. Medicina 56 (7), 321. doi:10.3390/medicina56070321

PubMed Abstract | CrossRef Full Text | Google Scholar

Jing, H., He, X., and Zheng, J. (2018). Exosomes and Regenerative Medicine: State of the Art and Perspectives. Translational Res. 196, 1–16. doi:10.1016/j.trsl.2018.01.005

CrossRef Full Text | Google Scholar

Joshi, A., Xu, Z., Ikegami, Y., Yamane, S., Tsurashima, M., and Ijima, H. (2020). Co-culture of Mesenchymal Stem Cells and Human Umbilical Vein Endothelial Cells on Heparinized Polycaprolactone/gelatin Co-spun Nanofibers for Improved Endothelium Remodeling. Int. J. Biol. Macromolecules 151, 186–192. doi:10.1016/j.ijbiomac.2020.02.163

CrossRef Full Text | Google Scholar

Kucharzewski, M., Rojczyk, E., Wilemska-Kucharzewska, K., Wilk, R., Hudecki, J., and Los, M. J. (2019). Novel Trends in Application of Stem Cells in Skin Wound Healing. Eur. J. Pharmacol. 843, 307–315. doi:10.1016/j.ejphar.2018.12.012

CrossRef Full Text | Google Scholar

Kumar, A. S., and Kamalasanan, K. (2021). Drug Delivery to Optimize Angiogenesis Imbalance in Keloid: A Review. J. Controlled Release 329, 1066–1076. doi:10.1016/j.jconrel.2020.10.035

CrossRef Full Text | Google Scholar

Leavitt, T., Hu, M. S., Marshall, C. D., Barnes, L. A., Lorenz, H. P., and Longaker, M. T. (2016). Scarless Wound Healing: Finding the Right Cells and Signals. Cell Tissue Res 365 (3), 483–493. doi:10.1007/s00441-016-2424-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, S. H., Jin, S. Y., Song, J. S., Seo, K. K., and Cho, K. H. (2012). Paracrine Effects of Adipose-Derived Stem Cells on Keratinocytes and Dermal Fibroblasts. Ann. Dermatol. 24 (2), 136–143. doi:10.5021/ad.2012.24.2.136

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, X., Xie, X., Lian, W., Shi, R., Han, S., Zhang, H., et al. (2018). Exosomes from Adipose-Derived Stem Cells Overexpressing Nrf2 Accelerate Cutaneous Wound Healing by Promoting Vascularization in a Diabetic Foot Ulcer Rat Model. Exp. Mol. Med. 50 (4), 1–14. doi:10.1038/s12276-018-0058-5

CrossRef Full Text | Google Scholar

Limandjaja, G. C., Niessen, F. B., Scheper, R. J., and Gibbs, S. (2020). The Keloid Disorder: Heterogeneity, Histopathology, Mechanisms and Models. Front. Cel Dev. Biol. 8, 360. doi:10.3389/fcell.2020.00360

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, K., Chen, C., Zhang, H., Chen, Y., and Zhou, S. (2019). Adipose Stem Cell‐derived Exosomes in Combination with Hyaluronic Acid Accelerate Wound Healing through Enhancing Re‐epithelialization and Vascularization. Br. J. Dermatol. 181 (4), 854–856. doi:10.1111/bjd.17984

CrossRef Full Text | Google Scholar

Luan, A., Duscher, D., Whittam, A. J., Paik, K. J., Zielins, E. R., Brett, E. A., et al. (2016). Cell‐Assisted Lipotransfer Improves Volume Retention in Irradiated Recipient Sites and Rescues Radiation‐Induced Skin Changes. Stem Cells 34 (3), 668–673. doi:10.1002/stem.2256

PubMed Abstract | CrossRef Full Text | Google Scholar

Mari, W., Alsabri, S. G., Tabal, N., Younes, S., Sherif, A., and Simman, R. (2015). Novel Insights on Understanding of Keloid Scar: Article Review. J. Am. Coll. Clin. Wound Specialists 7 (1-3), 1–7. doi:10.1016/j.jccw.2016.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Morikawa, S., Iribar, H., Gutiérrez-Rivera, A., Ezaki, T., and Izeta, A. (2019). Pericytes in Cutaneous Wound Healing. Adv. Exp. Med. Biol. 1147, 1–63. doi:10.1007/978-3-030-16908-4_1

PubMed Abstract | CrossRef Full Text | Google Scholar

Nakashima, M., Chung, S., Takahashi, A., Kamatani, N., Kawaguchi, T., Tsunoda, T., et al. (2010). A Genome-wide Association Study Identifies Four Susceptibility Loci for Keloid in the Japanese Population. Nat. Genet. 42 (9), 768–771. doi:10.1038/ng.645

PubMed Abstract | CrossRef Full Text | Google Scholar

Nicoletti, G., De Francesco, F., Mele, C. M., Cataldo, C., Grella, R., Brongo, S., et al. (2013). Clinical and Histologic Effects from CO2 Laser Treatment of Keloids. Lasers Med. Sci. 28 (3), 957–964. doi:10.1007/s10103-012-1178-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Ogawa, R. (2017). Keloid and Hypertrophic Scars Are the Result of Chronic Inflammation in the Reticular Dermis. Ijms 18 (3), 606. doi:10.3390/ijms18030606

PubMed Abstract | CrossRef Full Text | Google Scholar

Ogawa, R. (2021). The Most Current Algorithms for the Treatment and Prevention of Hypertrophic Scars and Keloids: A 2020 U36pdate of the Algorithms Published 10 Years Ago. Plast. Reconstr. Surg. 125, 557–568. doi:10.1097/PRS.0000000000008667

CrossRef Full Text | Google Scholar

O’Halloran, N., Courtney, D., Kerin, M. J., and Lowery, A. J. (2017). Adipose-Derived Stem Cells in Novel Approaches to Breast Reconstruction: Their Suitability for Tissue Engineering and Oncological Safety. Breast Cancer(Auckl) 11, 117822341772677. doi:10.1177/1178223417726777

CrossRef Full Text | Google Scholar

Putri, K. T., and Prasetyono, T. O. H. (2021). A Critical Review on the Potential Role of Adipose‐derived Stem Cells for Future Treatment of Hypertrophic Scars. J. Cosmet. Dermatol. doi:10.1111/jocd.14385

CrossRef Full Text | Google Scholar

Rahimnejad, M., Derakhshanfar, S., and Zhong, W. (2017). Biomaterials and Tissue Engineering for Scar Management in Wound Care. Burns Trauma 5, 4. doi:10.1186/s41038-017-0069-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Ren, S., Chen, J., Duscher, D., Liu, Y., Guo, G., Kang, Y., et al. (2019). Microvesicles from Human Adipose Stem Cells Promote Wound Healing by Optimizing Cellular Functions via AKT and ERK Signaling Pathways. Stem Cel Res Ther 10 (1), 47. doi:10.1186/s13287-019-1152-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Riccio, M., Marchesini, A., Senesi, L., Skrami, E., Gesuita, R., and De Francesco, F. (2019a). Managing Pathologic Scars by Injecting Auto-Cross-Linked Hyaluronic Acid: A Preliminary Prospective Clinical Study. Aesth Plast. Surg. 43 (2), 480–489. doi:10.1007/s00266-018-01303-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Riccio, M., Marchesini, A., Zingaretti, N., Carella, S., Senesi, L., Onesti, M. G., et al. (2019b). A Multicentre Study: The Use of Micrografts in the Reconstruction of Full-Thickness Posttraumatic Skin Defects of the Limbs-A Whole Innovative Concept in Regenerative Surgery. Stem Cell Int. 2019, 1–10. doi:10.1155/2019/5043518

PubMed Abstract | CrossRef Full Text | Google Scholar

Rockwell, W. B., Cohen, I. K., and Ehrlich, H. P. (1989). Keloids and Hypertrophic Scars. Plast. Reconstr. Surg. 84 (5), 827–837. doi:10.1097/00006534-198911000-00021

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodrigues, M., Kosaric, N., Bonham, C. A., and Gurtner, G. C. (2019). Wound Healing: A Cellular Perspective. Physiol. Rev. 99 (1), 665–706. doi:10.1152/physrev.00067.2017

PubMed Abstract | CrossRef Full Text | Google Scholar

Salameh, F., Shumaker, P. R., Goodman, G. J., Spring, L. K., Seago, M., Alam, M., et al. (2021). Energy‐based Devices for the Treatment of Acne Scars: 2022 International Consensus Recommendations. Lasers Surg. Med. doi:10.1002/lsm.23484

CrossRef Full Text | Google Scholar

Seifert, O., and Mrowietz, U. (2009). Keloid Scarring: Bench and Bedside. Arch. Dermatol. Res. 301 (4), 259–272. doi:10.1007/s00403-009-0952-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Seo, B. F., and Jung, S. N. (2016). The Immunomodulatory Effects of Mesenchymal Stem Cells in Prevention or Treatment of Excessive Scars. Stem Cell Int. 2016, 1–8. doi:10.1155/2016/6937976

PubMed Abstract | CrossRef Full Text | Google Scholar

Strong, A. L., Burow, M. E., Gimble, J. M., and Bunnell, B. A. (2015). Concise Review: The Obesity Cancer Paradigm: Exploration of the Interactions and Crosstalk with Adipose Stem Cells. Stem Cells 33 (2), 318–326. doi:10.1002/stem.1857

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, L. M., Wang, K. H., and Lee, Y. C. G. (2014). Keloid Incidence in Asian People and its Comorbidity with Other Fibrosis-Related Diseases: a Nationwide Population-Based Study. Arch. Dermatol. Res. 306 (9), 803–808. doi:10.1007/s00403-014-1491-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Svolacchia, F., De Francesco, F., Trovato, L., Graziano, A., and Ferraro, G. A. (2016). An Innovative Regenerative Treatment of Scars with Dermal Micrografts. J. Cosmet. Dermatol. 15 (3), 245–253. doi:10.1111/jocd.12212

CrossRef Full Text | Google Scholar

Tan, S., Khumalo, N., and Bayat, A. (2019). Understanding Keloid Pathobiology from a Quasi-Neoplastic Perspective: Less of a Scar and More of a Chronic Inflammatory Disease with Cancer-like Tendencies. Front. Immunol. 10, 1810. doi:10.3389/fimmu.2019.01810

PubMed Abstract | CrossRef Full Text | Google Scholar

Tracy, L. E., Minasian, R. A., and Caterson, E. J. (2016). Extracellular Matrix and Dermal Fibroblast Function in the Healing Wound. Adv. Wound Care 5 (3), 119–136. doi:10.1089/wound.2014.0561

PubMed Abstract | CrossRef Full Text | Google Scholar

Tsai, C. H., and Ogawa, R. (2019). Keloid Research: Current Status and Future Directions. Scars, Burns & Healing 5, 205951311986865. doi:10.1177/2059513119868659

CrossRef Full Text | Google Scholar

Tulandi, T., Al-Sannan, B., Akbar, G., Ziegler, C., and Miner, L. (2011). Prospective Study of Intraabdominal Adhesions Among Women of Different Races with or without Keloids. Am. J. Obstet. Gynecol. 204 (2), e1–132. doi:10.1016/j.ajog.2010.09.005

CrossRef Full Text | Google Scholar

Valadi, H., Ekström, K., Bossios, A., Sjöstrand, M., Lee, J. J., and Lötvall, J. O. (2007). Exosome-mediated Transfer of mRNAs and microRNAs Is a Novel Mechanism of Genetic Exchange between Cells. Nat. Cel Biol 9 (6), 654–659. doi:10.1038/ncb1596

CrossRef Full Text | Google Scholar

Wang, C., Wang, M., Xu, T., Zhang, X., Lin, C., Gao, W., et al. (2019). Engineering Bioactive Self-Healing Antibacterial Exosomes Hydrogel for Promoting Chronic Diabetic Wound Healing and Complete Skin Regeneration. Theranostics 9 (1), 65–76. doi:10.7150/thno.29766

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, L., Cheng, W., Zhu, J., Li, W., Li, D., Yang, X., et al. (2022). Electrospun Nanoyarn and Exosomes of Adipose-Derived Stem Cells for Urethral Regeneration: Evaluations In Vitro and In Vivo. Colloids Surf. B: Biointerfaces 209 (Pt 2), 112218. doi:10.1016/j.colsurfb.2021.112218

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, L., Hu, L., Zhou, X., Xiong, Z., Zhang, C., Shehada, H. M. A., et al. (2017). Exosomes Secreted by Human Adipose Mesenchymal Stem Cells Promote Scarless Cutaneous Repair by Regulating Extracellular Matrix Remodelling. Sci. Rep. 7 (1), 13321. doi:10.1038/s41598-017-12919-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Z. C., Zhao, W. Y., Cao, Y., Liu, Y. Q., Sun, Q., Shi, P., et al. (2020). The Roles of Inflammation in Keloid and Hypertrophic Scars. Front. Immunol. 11, 603187. doi:10.3389/fimmu.2020.603187

PubMed Abstract | CrossRef Full Text | Google Scholar

Watt, F. M., and Fujiwara, H. (2011). Cell-extracellular Matrix Interactions in normal and Diseased Skin. Cold Spring Harbor Perspect. Biol. 3 (4)), a005124. doi:10.1101/cshperspect.a005124

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiong, M., Zhang, Q., Hu, W., Zhao, C., Lv, W., Yi, Y., et al. (2020). Exosomes from Adipose-Derived Stem Cells: The Emerging Roles and Applications in Tissue Regeneration of Plastic and Cosmetic Surgery. Front. Cel Dev. Biol. 8, 574223. doi:10.3389/fcell.2020.574223

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, M., Fang, S., and Xie, A. (2021). Posttranscriptional Control of PLOD1 in Adipose-Derived Stem Cells Regulates Scar Formation through Altering Macrophage Polarization. Ann. Transl Med. 9 (20), 1573. doi:10.21037/atm-21-4978

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, Y., Wu, H., Zhang, Q., Ogawa, R., and Fu, S. (2021). Emerging Insights into the Immunological Aspects of Keloids. J. Dermatol. 48 (12), 1817–1826. doi:10.1111/1346-8138.16149

CrossRef Full Text | Google Scholar

Zouboulis, C. C., and Orfanos, C. E. (1990). Cryosurgical Treatment of Hypertrophic Scars and Keloids. Hautarzt 41 (12), 683–688.

PubMed Abstract | Google Scholar

Keywords: hypertrofic scars, adipose derived stem cell, wound healing, stem cell therapy, mechanism, keloid

Citation: Chen H, Hou K, Wu Y and Liu Z (2022) Use of Adipose Stem Cells Against Hypertrophic Scarring or Keloid. Front. Cell Dev. Biol. 9:823694. doi: 10.3389/fcell.2021.823694

Received: 28 November 2021; Accepted: 17 December 2021;
Published: 06 January 2022.

Edited by:

Guohui Liu, Huazhong University of Science and Technology, China

Reviewed by:

Bo Yin, Chinese Academy of Medical Sciences and Peking Union Medical College, China
Chanyuan Jiang, Chinese Academy of Medical Sciences and Peking Union Medical College, China

Copyright © 2022 Chen, Hou, Wu and Liu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Zeming Liu, 6myt@163.com

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.