1
|
Xie R, Yun J, Li C, Zhang S, Zhong A, Wu J, Cen Y, Li Z, Chen J. Identification of potential therapeutic target SPP1 and related RNA regulatory pathway in keloid based on bioinformatics analysis. Ann Med 2024; 56:2382949. [PMID: 39041063 PMCID: PMC11268233 DOI: 10.1080/07853890.2024.2382949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 06/05/2024] [Indexed: 07/24/2024] Open
Abstract
OBJECTIVE To explore the complex mechanisms of keloid, new approaches have been developed by different strategies. However, conventional treatment did not significantly reduce the recurrence rate. This study aimed to identify new biomarkers and mechanisms for keloid progression through bioinformatics analyses. METHODS In our study, microarray datasets for keloid were downloaded from the GEO database. Differentially expressed genes (DEGs) were identified by R software. Multiple bioinformatics tools were used to identify hub genes, and reverse predict upstream miRNAs and lncRNA molecules of target hub genes. Finally, the total RNA-sequencing technique and miRNA microarray were combined to validate the identified genes. RESULTS Thirty-one DEGs were screened out and the upregulated hub gene SPP1 was finally identified, which was consistent with our RNA-sequencing analysis results and validation dataset. In addition, a ceRNA network of mRNA (SPP1)-miRNA (miR-181a-5p)-lncRNA (NEAT1, MALAT1, LINC00667, NORAD, XIST and MIR4458HG) was identified by the bioinformatics databases. The results of our miRNA microarray showed that miR-181a-5p was upregulated in keloid, also we found that the lncRNA NEAT1 could affect keloid progression by retrieving the relevant literature. CONCLUSIONS We speculate that SPP1 is a potential candidate biomarker and therapeutic target for patients with keloid, and NEAT1/miR-181a-5p/SPP1 might be the RNA regulatory pathway that regulates keloid formation.
Collapse
Affiliation(s)
- Ruxin Xie
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jiao Yun
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Chenyu Li
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Shiwei Zhang
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Ai Zhong
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Junliang Wu
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Ying Cen
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zhengyong Li
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Junjie Chen
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Milara J, Ribera P, Marín S, Montero P, Roger I, Tenor H, Cortijo J. Phosphodiesterase 4 is overexpressed in human keloids and its inhibition reduces fibroblast activation and skin fibrosis. Chem Biol Interact 2024; 402:111211. [PMID: 39197814 DOI: 10.1016/j.cbi.2024.111211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/05/2024] [Accepted: 08/26/2024] [Indexed: 09/01/2024]
Abstract
There is a pressing medical need for improved treatments in skin fibrosis including keloids and hypertrophic scars (HTS). This study aimed to characterize the role of phosphodiesterase 4 (PDE4), specifically PDE4B in fibrotic skin remodeling in vitro and in vivo. In vitro, effects of PDE4A-D (Roflumilast) or PDE4B (siRNA) inhibition on TGFβ1-induced myofibroblast differentiation and dedifferentiation were studied in normal (NHDF) and keloid (KF) human dermal fibroblasts. In vivo, the role of PDE4 on HOCl-induced skin fibrosis in mice was addressed in preventive and therapeutic protocols. PDE4B (mRNA, protein) was increased in Keloid > HTS compared to healthy skin and in TGFβ-stimulated NHDF and KF. In Keloid > HTS, collagen Iα1, αSMA, TGFβ1 and NOX4 mRNA were all elevated compared to healthy skin confirming skin fibrosis. In vitro, inhibition of PDE4A-D and PDE4B similarly prevented TGFβ1-induced Smad3 and ERK1/2 phosphorylation and myofibroblast differentiation, elevated NOX4 protein and proliferation in NHDF. PDE4A-D inhibition enabled myofibroblast dedifferentiation and curbed TGFβ1-induced reactive oxygen species and fibroblast senescence. In KF PDE4A-D inhibition restrained TGFβ1-induced Smad3 and ERK1/2 phosphorylation, myofibroblast differentiation and senescence. Mechanistically, PDE4A-D inhibition rescued from TGFβ1-induced loss in PPM1A, a Smad3 phosphatase. In vivo, PDE4 inhibition mitigated HOCl-induced skin fibrosis in mice in preventive and therapeutic protocols. The current study provides novel evidence evolving rationale for PDE4 inhibitors in skin fibrosis (including keloids and HTS) and delivered evidence for a functional role of PDE4B in this fibrotic condition.
Collapse
Affiliation(s)
- Javier Milara
- CIBER de Enfermedades Respiratorias, Health Institute Carlos III, Valencia, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, Spain; Pharmacy Unit, University General Hospital Consortium of Valencia, Spain.
| | - Pilar Ribera
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Spain
| | - Severiano Marín
- Plastic Surgery Unit, University General Hospital Consortium, 46014, Valencia, Spain
| | - Paula Montero
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Spain; Faculty of Health Sciences, Universidad Europea de Valencia, 46010, Valencia, Spain
| | - Inés Roger
- CIBER de Enfermedades Respiratorias, Health Institute Carlos III, Valencia, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, Spain; Faculty of Health Sciences, Universidad Europea de Valencia, 46010, Valencia, Spain
| | | | - Julio Cortijo
- CIBER de Enfermedades Respiratorias, Health Institute Carlos III, Valencia, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, Spain
| |
Collapse
|
3
|
Li DJ, Berry CE, Wan DC, Longaker MT. Clinical, mechanistic, and therapeutic landscape of cutaneous fibrosis. Sci Transl Med 2024; 16:eadn7871. [PMID: 39321265 DOI: 10.1126/scitranslmed.adn7871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 09/03/2024] [Indexed: 09/27/2024]
Abstract
When dysregulated, skin fibrosis can lead to a multitude of pathologies. We provide a framework for understanding the wide clinical spectrum, mechanisms, and management of cutaneous fibrosis encompassing a variety of matrix disorders, fibrohistiocytic neoplasms, injury-induced scarring, and autoimmune scleroses. Underlying such entities are common mechanistic pathways that leverage morphogenic signaling, immune activation, and mechanotransduction to modulate fibroblast function. In light of the limited array of available treatments for cutaneous fibrosis, scientific insights have opened new therapeutic and investigative avenues for conditions that still lack effective interventions.
Collapse
Affiliation(s)
- Dayan J Li
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA 94063, USA
| | - Charlotte E Berry
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Derrick C Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael T Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
4
|
Chu X, Sun J, Dai S, Liang Y, Qian X, Xu J, Zhang J. AURKA Activates FOXO3a to Form a Positive Feedback Loop in the Proliferation and Migration of Keloid Fibroblasts. Adv Wound Care (New Rochelle) 2024. [PMID: 39078320 DOI: 10.1089/wound.2024.0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024] Open
Abstract
Objective: Keloids are benign fibroproliferative disorders with invasive growth exceeding the wound boundary. Aurora kinase A (AURKA) is a serine/threonine kinase highly expressed in various tumors, facilitating tumor growth and invasion. Currently, the role of AURKA in keloid remains unclear. Approach: Fibroblasts were isolated from keloid and normal skin samples. AURKA was evaluated by qPCR, Western blot, and immunohistochemistry. Transcriptome sequencing and dual-luciferase reporter assays were applied to figure out targets of AURKA. Following expression alteration and MLN8237 (an AURKA kinase inhibitor, AKI) treatment, phenotypical experiments were conducted to clarify biological functions of AURKA along with its target, and to probe into the clinical potential of AURKA inhibition. Results: AURKA was upregulated in keloid tissues and fibroblasts. Forkhead box O 3a (FOXO3a) was verified as a downstream of AURKA. Further experiments demonstrated that AURKA transactivated FOXO3a by binding to FOXO3a, while FOXO3a directly transactivated AURKA. Functionally, AURKA and FOXO3a cooperated in enhancing the proliferation and migration of keloid fibroblasts via protein kinase B (AKT) phosphorylation. Although MLN8237 weakened the proliferation and migration in keloid fibroblasts, the transactivation of AURKA on FOXO3a was independent of kinase activity. Innovation: This study reveals that AURKA and FOXO3a compose a transactivation loop in enhancing the proliferative and migrative properties of keloid fibroblasts, and proposes AURKA as a promising target. Conclusion: AURKA/FOXO3a loop promotes the proliferation and migration of keloid fibroblasts via AKT signaling. Despite the anti-keloid effects of AKIs, AURKA acts as a transcription factor independently of kinase activity, deepening our understanding on AKI insensitivity.
Collapse
Affiliation(s)
- Xi Chu
- Department of Plastic and Cosmetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Jiaqi Sun
- Department of Plastic Surgery, The Third Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Siya Dai
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yehua Liang
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xifei Qian
- School of Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinghong Xu
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jufang Zhang
- Department of Plastic and Cosmetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| |
Collapse
|
5
|
Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C, He J. TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther 2024; 9:61. [PMID: 38514615 PMCID: PMC10958066 DOI: 10.1038/s41392-024-01764-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 08/31/2023] [Accepted: 01/31/2024] [Indexed: 03/23/2024] Open
Abstract
Transforming growth factor (TGF)-β is a multifunctional cytokine expressed by almost every tissue and cell type. The signal transduction of TGF-β can stimulate diverse cellular responses and is particularly critical to embryonic development, wound healing, tissue homeostasis, and immune homeostasis in health. The dysfunction of TGF-β can play key roles in many diseases, and numerous targeted therapies have been developed to rectify its pathogenic activity. In the past decades, a large number of studies on TGF-β signaling have been carried out, covering a broad spectrum of topics in health, disease, and therapeutics. Thus, a comprehensive overview of TGF-β signaling is required for a general picture of the studies in this field. In this review, we retrace the research history of TGF-β and introduce the molecular mechanisms regarding its biosynthesis, activation, and signal transduction. We also provide deep insights into the functions of TGF-β signaling in physiological conditions as well as in pathological processes. TGF-β-targeting therapies which have brought fresh hope to the treatment of relevant diseases are highlighted. Through the summary of previous knowledge and recent updates, this review aims to provide a systematic understanding of TGF-β signaling and to attract more attention and interest to this research area.
Collapse
Affiliation(s)
- Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
6
|
Hong YK, Lin YC, Cheng TL, Lai CH, Chang YH, Huang YL, Hung CY, Wu CH, Hung KS, Ku YC, Ho YT, Tang MJ, Lin SW, Shi GY, McGrath JA, Wu HL, Hsu CK. TEM1/endosialin/CD248 promotes pathologic scarring and TGF-β activity through its receptor stability in dermal fibroblasts. J Biomed Sci 2024; 31:12. [PMID: 38254097 PMCID: PMC10804696 DOI: 10.1186/s12929-024-01001-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Pathologic scars, including keloids and hypertrophic scars, represent a common form of exaggerated cutaneous scarring that is difficult to prevent or treat effectively. Additionally, the pathobiology of pathologic scars remains poorly understood. We aim at investigating the impact of TEM1 (also known as endosialin or CD248), which is a glycosylated type I transmembrane protein, on development of pathologic scars. METHODS To investigate the expression of TEM1, we utilized immunofluorescence staining, Western blotting, and single-cell RNA-sequencing (scRNA-seq) techniques. We conducted in vitro cell culture experiments and an in vivo stretch-induced scar mouse model to study the involvement of TEM1 in TGF-β-mediated responses in pathologic scars. RESULTS The levels of the protein TEM1 are elevated in both hypertrophic scars and keloids in comparison to normal skin. A re-analysis of scRNA-seq datasets reveals that a major profibrotic subpopulation of keloid and hypertrophic scar fibroblasts greatly expresses TEM1, with expression increasing during fibroblast activation. TEM1 promotes activation, proliferation, and ECM production in human dermal fibroblasts by enhancing TGF-β1 signaling through binding with and stabilizing TGF-β receptors. Global deletion of Tem1 markedly reduces the amount of ECM synthesis and inflammation in a scar in a mouse model of stretch-induced pathologic scarring. The intralesional administration of ontuxizumab, a humanized IgG monoclonal antibody targeting TEM1, significantly decreased both the size and collagen density of keloids. CONCLUSIONS Our data indicate that TEM1 plays a role in pathologic scarring, with its synergistic effect on the TGF-β signaling contributing to dermal fibroblast activation. Targeting TEM1 may represent a novel therapeutic approach in reducing the morbidity of pathologic scars.
Collapse
Affiliation(s)
- Yi-Kai Hong
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Chen Lin
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan
| | - Tsung-Lin Cheng
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- College of Professional Studies, National Pingtung University of Science Technology, Pingtung, Taiwan
| | - Chao-Han Lai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Han Chang
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Lun Huang
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Yi Hung
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chen-Han Wu
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan
| | - Kuo-Shu Hung
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Chu Ku
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen-Ting Ho
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Ming-Jer Tang
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Wha Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University Hospital, Taipei, Taiwan
| | - Guey-Yueh Shi
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - John A McGrath
- St John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, London, UK
| | - Hua-Lin Wu
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan.
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Chao-Kai Hsu
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- International Center for Wound Repair and Regeneration (iWRR), National Cheng Kung University, Tainan, Taiwan.
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
7
|
Palić J, Saey V, van der Lugt J, Baumgärtner W, Hülskötter K. Cytologic, histopathologic, and immunohistochemical features of keloidal fibroma in a dog. Vet Clin Pathol 2023; 52:676-680. [PMID: 37661191 DOI: 10.1111/vcp.13282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/12/2023] [Accepted: 06/04/2023] [Indexed: 09/05/2023]
Abstract
A five-year-old male English Bulldog was presented with a firm, well-circumscribed, 1 cm in diameter cutaneous mass on the left flank. Fine-needle aspiration (FNA) biopsy samples were collected for cytologic analysis. Cytology revealed a highly cellular sample consisting of spindle cells, numerous bundles of thick, glassy eosinophilic material (hyalinized collagen), and inflammatory cells. Spindle cells showed moderate anisocytosis and anisokaryosis, had oval nuclei with coarsely stippled chromatin, 1-3 prominent round nucleoli, and moderate amounts of wispy cytoplasm. Cells were occasionally associated with an eosinophilic extracellular matrix. Binucleated and trinucleated spindle cells were often noted. Low numbers of macrophages, small lymphocytes, and individual well-granulated mast cells were also present. The lesion was excised and submitted for histopathologic examination, revealing a well-delineated, nonencapsulated mass composed of hyalinized collagen fibers separated by spindle-shaped mesenchymal cells in the deep dermis and subcutis. Mild anisocytosis and anisokaryosis and less than one mitosis per 10 × high power fields were present. Excision of the mass was complete. The findings were consistent with a keloidal fibroma, a rare benign variant of fibroma. Neoplastic cells showed positive immunoreactivity for vimentin, and a small-to-moderate number of tumor cells showed positive immunoreactivity for α-smooth muscle actin. This is the first cytologic description of a keloidal fibroma correlated with histopathologic findings and immunolabeling. In cases where keloidal neoplasia is suspected, and since moderate cellular atypia can be present on cytologic examination even in cases of keloidal fibroma, histopathologic examination is necessary to differentiate between keloidal fibroma and keloidal fibrosarcoma.
Collapse
Affiliation(s)
- Jelena Palić
- Vet Med Labor GmbH Division of IDEXX Laboratories, Kornwestheim, Germany
| | - Veronique Saey
- Vet Med Labor GmbH Division of IDEXX Laboratories, Kornwestheim, Germany
| | - Jaco van der Lugt
- Vet Med Labor GmbH Division of IDEXX Laboratories, Kornwestheim, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Kirsten Hülskötter
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
8
|
Eftimie R, Rolin G, Adebayo OE, Urcun S, Chouly F, Bordas SPA. Modelling Keloids Dynamics: A Brief Review and New Mathematical Perspectives. Bull Math Biol 2023; 85:117. [PMID: 37855947 DOI: 10.1007/s11538-023-01222-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 10/02/2023] [Indexed: 10/20/2023]
Abstract
Keloids are fibroproliferative disorders described by excessive growth of fibrotic tissue, which also invades adjacent areas (beyond the original wound borders). Since these disorders are specific to humans (no other animal species naturally develop keloid-like tissue), experimental in vivo/in vitro research has not led to significant advances in this field. One possible approach could be to combine in vitro human models with calibrated in silico mathematical approaches (i.e., models and simulations) to generate new testable biological hypotheses related to biological mechanisms and improved treatments. Because these combined approaches do not really exist for keloid disorders, in this brief review we start by summarising the biology of these disorders, then present various types of mathematical and computational approaches used for related disorders (i.e., wound healing and solid tumours), followed by a discussion of the very few mathematical and computational models published so far to study various inflammatory and mechanical aspects of keloids. We conclude this review by discussing some open problems and mathematical opportunities offered in the context of keloid disorders by such combined in vitro/in silico approaches, and the need for multi-disciplinary research to enable clinical progress.
Collapse
Affiliation(s)
- R Eftimie
- Laboratoire de Mathématiques de Besançon, Université de Franche-Comté, 25000, Besançon, France.
| | - G Rolin
- INSERM CIC-1431, CHU Besançon, F-25000, Besançon, France
- EFS, INSERM, UMR 1098 RIGHT, Université de Franche-Comté, F-25000, Besançon, France
| | - O E Adebayo
- Laboratoire de Mathématiques de Besançon, Université de Franche-Comté, 25000, Besançon, France
| | - S Urcun
- Institute for Computational Engineering, Faculty of Science, Technology and Communication, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - F Chouly
- Institut de Mathématiques de Bourgogne, Université de Franche-Comté, 21078, Dijon, France
- Center for Mathematical Modelling and Department of Mathematical Engineering, University of Chile and IRL 2807 - CNRS, Santiago, Chile
- Departamento de Ingeniería Matemática, CI2MA, Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - S P A Bordas
- Institute for Computational Engineering, Faculty of Science, Technology and Communication, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
9
|
Chen JY, Feng QL, Pan HH, Zhu DH, He RL, Deng CC, Yang B. An Open-Label, Uncontrolled, Single-Arm Clinical Trial of Tofacitinib, an Oral JAK1 and JAK3 Kinase Inhibitor, in Chinese Patients with Keloid. Dermatology 2023; 239:818-827. [PMID: 37643592 DOI: 10.1159/000532064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 07/13/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND The keloid treatment is still a thorny and complicated clinical problem, especially in multiple keloids induced by wound, severe burn, ethnic background or cultural behaviors, or unexplained skin healing. Mainstream treatments have limited efficacy in treating multiple keloids. As no oral treatment with painlessness and convenience is available, oral treatment strategies should be formulated. OBJECTIVES This study aimed to investigate the efficacy and therapeutic mechanism of oral tofacitinib in keloid patients. METHODS We recruited the 7 patients with keloid scars and prescribed 5 mg of tofacitinib twice a day orally with a maximum follow-up of 12 weeks. The Patient and Observer Scar Assessment Scale (POSAS), the Vancouver scar scale (VSS), ANTERA 3D camera, and the DUB Skin Scanner 75 were used to assess the characteristics of the lesion. Immunohistochemistry was performed to evaluate collagen synthesis, proliferation, and relative molecular pathways. Moreover, the effects of tofacitinib were assessed on keloid fibroblast in vitro. RESULTS After 12 weeks of oral tofacitinib, significant improvement in POSAS, VSS, and Dermatology Life Quality Index (DLQI) scores was observed (p < 0.05). The volume, lesion height, and dermis thickness of the keloid decreased (p < 0.05). Moreover, significant decreases in the expression of collagen I, Ki67, p-STAT 3, and p-SMAD2 were observed after 12 weeks of administration. In vitro experiments suggested that tofacitinib treatment inhibits fibroblast proliferation and collagen I synthesis via suppression of STAT3 and SMAD2 pathway. CONCLUSION Tofacitinib, a new candidate oral drug for keloid, could reduce keloid lesion volume by inhibiting collagen synthesis and inhibiting fibroblast proliferation, and alleviate itch and pain to obtain a better life quality.
Collapse
Affiliation(s)
- Jun-Yi Chen
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China,
- Dermatology Hospital, Southern Medical University, Guangzhou, China,
| | - Qing-Lan Feng
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Hui-Hui Pan
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Ding-Heng Zhu
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Ren-Liang He
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Cheng-Cheng Deng
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Dirand Z, Tissot M, Chatelain B, Viennet C, Rolin G. Is Spheroid a Relevant Model to Address Fibrogenesis in Keloid Research? Biomedicines 2023; 11:2350. [PMID: 37760792 PMCID: PMC10526056 DOI: 10.3390/biomedicines11092350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023] Open
Abstract
Keloid refers to a fibro-proliferative disorder characterized by an accumulation of extracellular matrix at the dermis level, overgrowing beyond the initial wound and forming tumor-like nodule areas. The absence of treatment for keloid is clearly related to limited knowledge about keloid etiology. In vitro, keloids were classically studied through fibroblasts monolayer culture, far from keloid in vivo complexity. Today, cell aggregates cultured as 3D spheroid have gained in popularity as new tools to mimic tissue in vitro. However, no previously published works on spheroids have specifically focused on keloids yet. Thus, we hypothesized that spheroids made of keloid fibroblasts (KFs) could be used to model fibrogenesis in vitro. Our objective was to qualify spheroids made from KFs and cultured in a basal or pro-fibrotic environment (+TGF-β1). As major parameters for fibrogenesis assessment, we evaluated apoptosis, myofibroblast differentiation and response to TGF-β1, extracellular matrix (ECM) synthesis, and ECM-related genes regulation in KFs spheroids. We surprisingly observed that fibrogenic features of KFs are strongly downregulated when cells are cultured in 3D. In conclusion, we believe that spheroid is not the most appropriate model to address fibrogenesis in keloid, but it constitutes an efficient model to study the deactivation of fibrotic cells.
Collapse
Affiliation(s)
- Zélie Dirand
- Université de Franche-Comté, Sciences Médicales et Pharmaceutiques, EFS, INSERM, UMR RIGHT, 25000 Besançon, France; (Z.D.)
| | - Marion Tissot
- Université de Franche-Comté, Sciences Médicales et Pharmaceutiques, EFS, INSERM, UMR RIGHT, 25000 Besançon, France; (Z.D.)
| | - Brice Chatelain
- Service de Chirurgie Maxillo-Faciale, Stomatologie et Odontologie Hospitalière, CHU Besançon, 25000 Besançon, France
| | - Céline Viennet
- Université de Franche-Comté, Sciences Médicales et Pharmaceutiques, EFS, INSERM, UMR RIGHT, 25000 Besançon, France; (Z.D.)
| | - Gwenaël Rolin
- Université de Franche-Comté, Sciences Médicales et Pharmaceutiques, CHU Besançon EFS, INSERM, UMR RIGHT, 25000 Besançon, France
| |
Collapse
|
11
|
Qiu ZK, Zhang MZ, Zhang WC, Li ZJ, Si LB, Long X, Yu NZ, Wang XJ. Role of HIF-1α in pathogenic mechanisms of keloids. J Cosmet Dermatol 2023; 22:1436-1448. [PMID: 36718786 DOI: 10.1111/jocd.15601] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/08/2022] [Accepted: 12/12/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUDS AND OBJECTIVE Keloids are defined as overrepairing products that develop after skin lesions. Keloids are characterized by the proliferation of fibroblasts and the overaccumulation of extracellular matrix components (mainly collagen), leading to a locally hypoxic microenvironment. Hence, this article was aimed to review hypoxia in pathogenesis of keloids. METHODS We reviewed and summarized the relevant published studies. RESULTS Hypoxia results in the accumulation of hypoxia-inducible factor 1α (HIF-1α) in keloids, contributing to overactivation of the fibrotic signaling pathway, epithelial-mesenchymal transition, and changes in metabolism, eventually leading to aggravated fibrosis, infiltrative growth, and radiotherapy resistance. CONCLUSION It is, therefore, essential to understand the role of HIF-1α in the pathogenic mechanisms of keloids in order to develop new therapeutic approaches.
Collapse
Affiliation(s)
- Zi-Kai Qiu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Zi Zhang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen-Chao Zhang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Jin Li
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lou-Bin Si
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Long
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan-Ze Yu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Jun Wang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
12
|
He L, Zhu C, Dou H, Yu X, Jia J, Shu M. Keloid Core Factor CTRP3 Overexpression Significantly Controlled TGF- β1-Induced Propagation and Migration in Keloid Fibroblasts. DISEASE MARKERS 2023; 2023:9638322. [PMID: 37091895 PMCID: PMC10115533 DOI: 10.1155/2023/9638322] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 04/25/2023]
Abstract
Purpose Keloid is a type of benign fibrous proliferative tumor characterized by excessive scarring. C1q/TNF-related protein 3 (CTRP3) has been proven to possess antifibrotic effect. Here, we explored the role of CTRP3 in keloid. In the current research, we examined the influence of CTRP3 on keloid fibroblasts (KFs) and investigated the potential molecular mechanism. Methods KF tissue specimens and adjacent normal fibroblast (NF) tissues were collected cultured from 10 keloid participants. For the TGF-β1 stimulation group, KFs were processed with human recombinant TGF-β1. Cell transfection of pcDNA3.1-CTRP3 or pcDNA3.1 was performed. The siRNA of CTRP3 (si-CTRP3) or negative control siRNA (si-scramble) was transfected into KFs. Results CTRP3 was downregulated in keloid tissues and KFs. CTRP3 overexpression significantly controlled TGF-β1-induced propagation and migration in KFs. Col I, α-SMA, and fibronectin mRNA and protein levels were enhanced by TGF-β1 stimulation, whereas they were inhibited by CTRP3 overexpression. In contrast, CTRP3 knockdown exhibited the opposite effect. In addition, CTRP3 attenuated TGF-β receptors TRI and TRII in TGF-β1-induced KFs. Furthermore, CTRP3 prevented TGF-β1-stimulated nuclear translocation of smad2 and smad3 and suppressed the expression levels of p-smad2 and p-smad3 in KFs. Conclusion CTRP3 exerted an antifibrotic role through inhibiting proliferation, migration, and ECM accumulation of KFs via regulating TGF-β1/Smad signal path.
Collapse
Affiliation(s)
- Lin He
- Department of Plastic, Aesthetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Chan Zhu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Huicong Dou
- Department of Plastic, Aesthetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xueyuan Yu
- Department of Plastic, Aesthetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jing Jia
- Department of Plastic, Aesthetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Maoguo Shu
- Department of Plastic, Aesthetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
13
|
Suwanchinda A, Nararatwanchai T. Efficacy and safety of the innovative cold atmospheric-pressure plasma technology in the treatment of keloid: A randomized controlled trial. J Cosmet Dermatol 2022; 21:6788-6797. [PMID: 36120805 DOI: 10.1111/jocd.15397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/10/2022] [Accepted: 09/16/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Keloid (KD) treatment is challenging for both physicians and patients. It can be functional debilitating and psychologically distressing. Available current therapeutics modalities give inconsistently effective results. OBJECTIVES To evaluate the efficacy and safety of innovative cold atmospheric plasma (CAP) technology in the treatment of keloid. METHODS This prospective, randomized control trial, the assessor-blinded trial, includes 18 patients with keloids. The keloid lesion was divided into two halves. One side was randomly treated with CAP technology biweekly on the same treated side for five sessions with a follow-up 30 days after finishing the final treatment. Another half was left untreated as a control. Efficacy assessment using POSAS, VSS, Patients' satisfaction scale, Antera 3D® skin imaging system. The safety assessment using VAS and adverse effects monitoring was completed. RESULTS Objective assessment using Antera 3D® skin imaging system (Miravex, Dublin, Ireland) showed statistically significant improvement (p-value <0.05) on the treated side compared with the untreated side in all parameters, color, melanin, hemoglobin, texture, except for volume. POSAS, patient, and observer overall opinion score, and patient and observer total score in the summary of all rated characteristics, comparing the treated and untreated areas, showed a statistically significant reduction in all parameters after two treatments (*p-value <0.05). VSS showed statistically significant improvement after the second treatment and continued to the last follow-up. Most patients rated satisfaction scales up to 72.2% as moderate improvement, 11.1% as great improvement, 11.1% as slight improvement, and 5.6% as no change. The adverse effect was only a small scab in one patient. CONCLUSION CAP technology could be considered an alternative treatment for keloid offering mild-to-moderate improvement with minimal side effects.
Collapse
Affiliation(s)
- Atchima Suwanchinda
- Department of Dermatology, School of Anti-aging and Regenerative Medicine, Mae Fah Luang University, Bangkok, Thailand
| | - Thamthiwat Nararatwanchai
- Department of Dermatology, School of Anti-aging and Regenerative Medicine, Mae Fah Luang University, Bangkok, Thailand
| |
Collapse
|
14
|
Macrophages Are Polarized toward an Inflammatory Phenotype by their Aged Microenvironment in the Human Skin. J Invest Dermatol 2022; 142:3136-3145.e11. [PMID: 35850208 DOI: 10.1016/j.jid.2022.06.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 01/05/2023]
Abstract
Aging of the skin is accompanied by cellular as well as tissue environmental changes, ultimately reducing the ability of the tissue to regenerate and adequately respond to external stressors. Macrophages are important gatekeepers of tissue homeostasis, and it has been reported that their number and phenotype change during aging in a site-specific manner. How aging affects human skin macrophages and what implications this has for the aging process in the tissue are still not fully understood. Using single-cell RNA-sequencing analysis, we show that there is at least a 50% increase of macrophages in human aged skin, which appear to have developed from monocytes and exhibit more proinflammatory M1-like characteristics. In contrast, the cell-intrinsic ability of aged monocytes to differentiate into M1 macrophages was reduced. Using coculture experiments with aged dermal fibroblasts, we show that it is the aged microenvironment that drives a more proinflammatory phenotype of macrophages in the skin. This proinflammatory M1-like phenotype in turn negatively influenced the expression of extracellular matrix proteins by fibroblasts, emphasizing the impact of the aged macrophages on the skin phenotype.
Collapse
|
15
|
Liu D, Zhang Y, Zhen L, Xu R, Ji Z, Ye Z. Activation of the NFκB signaling pathway in IL6+CSF3+ vascular endothelial cells promotes the formation of keloids. Front Bioeng Biotechnol 2022; 10:917726. [PMID: 36082167 PMCID: PMC9445273 DOI: 10.3389/fbioe.2022.917726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/29/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Keloid is a disease caused by abnormal proliferation of skin fibres, the causative mechanism of which remains unclear. Method: In this study, endothelial cells of keloids were studied using scRNAseq combined with bulk-RNAseq data from keloids. The master regulators driving keloid development were identified by transcription factor enrichment analysis. The pattern of changes in vascular endothelial cells during keloid development was explored by inferring endothelial cell differentiation trajectories. Deconvolution of bulkRNAseq by CIBERSORTX verified the pattern of keloidogenesis. Immunohistochemistry for verification of the lesion process in keloid endothelial cells. Results: The endothelial cells of keloids consist of four main cell populations (MMP1+ Endo0, FOS + JUN + Endo1, IL6+CSF3+Endo2, CXCL12 + Endo3). Endo3 is an endothelial progenitor cell, Endo1 is an endothelial cell in the resting state, Endo2 is an endothelial cell in the activated state and Endo0 is an endothelial cell in the terminally differentiated state. Activation of the NFΚB signaling pathway is a typical feature of Endo2 and represents the early skin state of keloids. Conclusion: We have identified patterns of vascular endothelial cell lesions during keloidogenesis and development, and have found that activation of the NFΚB signaling pathway is an essential feature of keloid formation. These findings are expected to contribute to the understanding of the pathogenesis of keloids and to the development of new targeted therapeutic agents for the lesional characteristics of vascular endothelial cells.
Collapse
Affiliation(s)
- Delin Liu
- Department of General Surgery, Institute for Minimally Invasive Surgery, Affiliated Zhongda Hospital, Medical School, Southeast University, Nanjing, China
- Department of Endcrinology, Affiliated Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Yidi Zhang
- Department of Endcrinology, Affiliated Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Lisha Zhen
- School of Statistics, Renmin University of China, Beijing, China
- Beijing Sankuai Online Technology Co.,Ltd, Dhaka, Bangladesh
| | - Rong Xu
- Department of Endcrinology, Affiliated Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Zhenling Ji
- Department of General Surgery, Institute for Minimally Invasive Surgery, Affiliated Zhongda Hospital, Medical School, Southeast University, Nanjing, China
- *Correspondence: Zhenling Ji, ; Zheng Ye,
| | - Zheng Ye
- Department of Endcrinology, Affiliated Zhongda Hospital, Medical School, Southeast University, Nanjing, China
- *Correspondence: Zhenling Ji, ; Zheng Ye,
| |
Collapse
|
16
|
Feng QL, Gu JJ, Chen JY, Zheng WY, Pan HH, Xu XY, Deng CC, Yang B. TSP1 promotes fibroblast proliferation and extracellular matrix deposition via the IL6/JAK2/STAT3 signalling pathway in keloids. Exp Dermatol 2022; 31:1533-1542. [PMID: 35661430 DOI: 10.1111/exd.14623] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/06/2022] [Accepted: 06/02/2022] [Indexed: 11/29/2022]
Abstract
Keloids are benign fibroproliferative diseases with abnormally proliferated bulges beyond the edge of the skin lesions, and they are characterized by uncontrolled fibroblast proliferation and excessive extracellular matrix deposition in the dermis. However, the definite mechanisms that increase fibroblast proliferation and collagen deposition in keloids remain unclear. Thrombospondin 1 (TSP1) has been suggested to play an important role in wound healing and fibrotic disorders, but its role in keloids is unknown. In this study, we aimed to clarify the specific role of TSP1 in keloids and explore the potential mechanism. Our results demonstrated that TSP1 was highly expressed in keloid lesions compared to normal skin. Knockdown of TSP1 in keloid fibroblasts decreased cell proliferation and collagen I deposition. Exogenous TSP1 treatment increased cell proliferation and collagen I deposition in normal fibroblasts. We further investigated the underlying mechanism and found that TSP1 promoted fibroblast proliferation and extracellular matrix deposition by upregulating the IL6/JAK2/STAT3 pathway. Moreover, we verified that TSP1 expression was positively correlated with IL6/STAT3 signalling activity in keloids. Taken together, our findings indicate that TSP1 promotes keloid development via the IL6/JAK2/STAT3 signalling pathway and blocking TSP1 may represent a potential strategy for keloid therapy.
Collapse
Affiliation(s)
- Qing-Lan Feng
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Jing Gu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Jun-Yi Chen
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Wen-Yue Zheng
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Hui-Hui Pan
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Xue-Yan Xu
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Cheng-Cheng Deng
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
17
|
Carpenter DJ, Hamdi OA, Finberg AM, Daniero JJ. Laryngotracheal stenosis: Mechanistic review. Head Neck 2022; 44:1948-1960. [PMID: 35488503 PMCID: PMC9543412 DOI: 10.1002/hed.27079] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 11/24/2022] Open
Abstract
Background The purpose of this review article is to summarize the existing literature surrounding wound healing mechanisms in laryngotracheal stenosis. Methods A review of general wound healing pathophysiology, followed by a focused review of iatrogenic laryngotracheal stenosis (iLTS) and idiopathic subglottic stenosis (iSGS) as conditions of aberrant wound healing. Results iLTS is the scarring of the laryngotracheal complex, coming secondary to injury from prolonged intubation. iSGS is a chronic fibroinflammatory scarring and narrowing of the subglottic airway in the absence of any obvious preceding injury or trauma. They are both thought to result from a prolonged and dysregulated wound healing response that promotes the deposition of pathologic scar in the airway. Conclusions Understanding the mechanisms that underlie wound healing will help identify and intervene on the process early in its development and discover future therapies that target individual wound healing mechanisms limiting the incidence of this recalcitrant disease process.
Collapse
Affiliation(s)
- Delaney J Carpenter
- Department of Otolaryngology - Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Osama A Hamdi
- Department of Otolaryngology - Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Ariel M Finberg
- Department of Otolaryngology - Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, USA
| | - James J Daniero
- Department of Otolaryngology - Head and Neck Surgery, University of Virginia Health System, Charlottesville, Virginia, USA
| |
Collapse
|
18
|
Abstract
Significance: Keloid scarring is cosmetically disfiguring, psychosocially distressing, and can be physically disabling. The pathophysiology of keloid formation is poorly understood and subsequently, treatment options are ill defined, limited, and largely unsatisfactory. Therefore, in view of its unsatisfactory and recalcitrant management, keloid therapy is often seen as a financial burden affecting both patients and the health care systems. Recent Advances: Increased research on the genetic and epigenetic mechanisms in keloids has broadened our understanding of keloid pathobiology. Epigenetic mechanisms, mainly DNA methylation, histone modification, and noncoding RNAs, are currently being widely investigated. Advances in genetic sequencing technology and reduced cost have aided this endeavor. Studies on blood and patient-derived keloid tissue are being done with therapeutic agents targeting epigenetic and genetic markers with the shared goal of identifying the pathways underlying the initiation and maintenance of keloids. These advances have informed us of multiple complex molecular pathways implicated in keloids, which are yet to be fully elucidated. Critical Issues: Improved understanding of the genetic and epigenetic causes implicated in keloids will enhance our knowledge of this enigmatic disorder and likely lead to the development of therapeutic targets based on the available clinical and experimental studies. Due to the incomplete knowledge of molecular targets involved in keloid scarring pathways, therapeutics is still lagging for this clinically and scientifically important condition. Future Directions: Focused research on the identification of molecular targets and mechanistic pathways implicated in keloids is required to generate novel antifibrotic therapeutic options to decrease or eradicate recurrence of the disease as well as associated morbidity and improve the quality of life of those affected with keloids.
Collapse
Affiliation(s)
- Dennias Tonderai Nyika
- MRC Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Nonhlanhla P. Khumalo
- MRC Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Ardeshir Bayat
- MRC Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
- Plastic and Reconstructive Surgery Research, Wound Healing Unit, NIHR Manchester Biomedical Research, Centre for Dermatology Research, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
19
|
OMICS Approaches Evaluating Keloid and Hypertrophic Scars. Int J Inflam 2022; 2022:1490492. [PMID: 36483731 PMCID: PMC9722497 DOI: 10.1155/2022/1490492] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/01/2022] [Indexed: 11/18/2022] Open
Abstract
Abnormal scar formation during wound healing can result in keloid and hypertrophic scars, which is a major global health challenge. Such abnormal scars can cause significant physiological pain and psychological distress and become a financial burden. Due to the biological complexity of scar formation, the pathogenesis of such scars and how to prevent them from forming remains elusive. In this review paper, we delve into the world of "omics" approaches to study abnormal scars and provide examples of genomics, transcriptomics, proteomics, epigenomics, and metabolomics. The benefits of "omics" approaches are that they allow for high-throughput studies and the analysis of 100s to 1000s of genes and proteins with the accumulation of large quantities of data. Currently in the field, there is a lack of "omics" review articles describing pathological scars. In this review, we summarize genome-wide linkage analysis, genome-wide association studies, and microarray data to name a few omics technologies. Such data can provide novel insights into different molecular pathways and identify novel factors which may not be captured through small-scale laboratory techniques.
Collapse
|
20
|
Kim KI, Kwon CI, Lee JH, Kim CD, Yoon TJ. Inhibitory Effect of Mitoxantrone on Collagen Synthesis in Dermal Fibroblasts. Ann Dermatol 2022; 34:206-211. [PMID: 35721328 PMCID: PMC9171176 DOI: 10.5021/ad.2022.34.3.206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 12/21/2021] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
Background Fibroblasts produce collagen molecules that support the structure of the skin. The decrease and hypersynthesis of collagen causes skin problems such as skin atrophy, wrinkles and scars. Objective The purpose of this study is to investigate the mechanism of mitoxantrone on collagen synthesis in fibroblasts. Methods Cultured fibroblasts were treated with mitoxantrone, and then collagen synthesis was confirmed by reverse transcription-polymerase chain reaction and Western blot. Results Mitoxantrone inhibited the expression of type I collagen in fibroblasts at both the mRNA and protein levels. In the collagen gel contraction assay, mitoxantrone significantly inhibited gel contraction compared to the control group. Mitoxantrone inhibited transforming growth factor (TGF)-β-induced phosphorylation of SMAD3. Finally, mitoxantrone inhibited the expression of LARP6, an RNA-binding protein that regulates collagen mRNA stability. Conclusion These results suggest that mitoxantrone reduces collagen synthesis by inhibiting TGF-β/SMAD signaling and LARP6 expression in fibroblasts, which can be developed as a therapeutic agent for diseases caused by collagen hypersynthesis.
Collapse
Affiliation(s)
- Kyung-Il Kim
- Department of Dermatology, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Dermatology, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Chang-Il Kwon
- Department of Dermatology, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
- Gyeongsang Institute of Health Sciences, Jinju, Korea
| | | | - Chang-Deok Kim
- Department of Dermatology, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Tae-Jin Yoon
- Department of Dermatology, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
- Gyeongsang Institute of Health Sciences, Jinju, Korea
| |
Collapse
|
21
|
Kawagishi-Hotta M, Hasegawa S, Inoue Y, Hasebe Y, Arima M, Iwata Y, Sugiura K, Akamatsu H. Gremlin 2 suppresses differentiation of stem/progenitor cells in the human skin. Regen Ther 2021; 18:191-201. [PMID: 34307797 PMCID: PMC8280529 DOI: 10.1016/j.reth.2021.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/30/2021] [Accepted: 06/24/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION The skin is comprised of various kinds of cells and has three layers, the epidermis, dermis and subcutaneous adipose tissue. Stem cells in each tissue duplicate themselves and differentiate to supply new cells that function in the tissue, and thereby maintain the tissue homeostasis. In contrast, senescent cells accumulate with age and secrete senescence-associated secretory phenotype (SASP) factors that impair surrounding cells and tissues, which lowers the capacity to maintain homeostasis in each tissue. Previously, we found Gremlin 2 (GREM2) as a novel SASP factor in the skin and reported that GREM2 suppressed the differentiation of adipose-derived stromal/stem cells. In the present study, we investigated the effects of GREM2 on stem cells in the epidermis and dermis. METHODS To examine whether GREM2 expression and the differentiation levels in the epidermis and dermis are correlated, the expressions of GREM2, stem cell markers, an epidermal differentiation marker Keratin 10 (KRT10) and a dermal differentiation marker type 3 procollagen were examined in the skin samples (n = 14) randomly chosen from the elderly where GREM2 expression level is high and the individual differences of its expression are prominent. Next, to test whether GREM2 affects the differentiation of skin stem cells, cells from two established lines (an epidermal and a dermal stem/progenitor cell model) were cultured and induced to differentiate, and recombinant GREM2 protein was added. RESULTS In the human skin, the expression levels of GREM2 varied among individuals both in the epidermis and dermis. The expression level of GREM2 was not correlated with the number of stem cells, but negatively correlated with those of both an epidermal and a dermal differentiation markers. The expression levels of epidermal differentiation markers were significantly suppressed by the addition of GREM2 in the three-dimensional (3D) epidermis generated with an epidermal stem/progenitor cell model. In addition, by differentiation induction, the expressions of dermal differentiation markers were induced in cells from a dermal stem/progenitor cell model, and the addition of GREM2 significantly suppressed the expressions of the dermal differentiation markers. CONCLUSIONS GREM2 expression level did not affect the numbers of stem cells in the epidermis and dermis but affects the differentiation and maturation levels of the tissues, and GREM2 suppressed the differentiation of stem/progenitor cells in vitro. These findings suggest that GREM2 may contribute to the age-related reduction in the capacity to maintain skin homeostasis by suppressing the differentiation of epidermal and dermal stem/progenitor cells.
Collapse
Affiliation(s)
- Mika Kawagishi-Hotta
- Research Laboratories, Nippon Menard Cosmetic Co., LTD., Japan
- Nagoya University-MENARD Collaborative Research Chair, Nagoya University Graduate School of Medicine, Japan
- Department of Applied Cell and Regenerative Medicine, Fujita Health University School of Medicine, Japan
| | - Seiji Hasegawa
- Research Laboratories, Nippon Menard Cosmetic Co., LTD., Japan
- Nagoya University-MENARD Collaborative Research Chair, Nagoya University Graduate School of Medicine, Japan
- Department of Dermatology, Fujita Health University School of Medicine, Japan
| | - Yu Inoue
- Research Laboratories, Nippon Menard Cosmetic Co., LTD., Japan
- Nagoya University-MENARD Collaborative Research Chair, Nagoya University Graduate School of Medicine, Japan
| | - Yuichi Hasebe
- Research Laboratories, Nippon Menard Cosmetic Co., LTD., Japan
- Nagoya University-MENARD Collaborative Research Chair, Nagoya University Graduate School of Medicine, Japan
| | - Masaru Arima
- Department of Dermatology, Fujita Health University School of Medicine, Japan
| | - Yohei Iwata
- Department of Dermatology, Fujita Health University School of Medicine, Japan
| | - Kazumitsu Sugiura
- Department of Dermatology, Fujita Health University School of Medicine, Japan
| | - Hirohiko Akamatsu
- Department of Applied Cell and Regenerative Medicine, Fujita Health University School of Medicine, Japan
| |
Collapse
|
22
|
Zheng W, Lin G, Wang Z. Bioinformatics study on different gene expression profiles of fibroblasts and vascular endothelial cells in keloids. Medicine (Baltimore) 2021; 100:e27777. [PMID: 34964740 PMCID: PMC8615345 DOI: 10.1097/md.0000000000027777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 10/28/2021] [Indexed: 01/05/2023] Open
Abstract
Keloid is a benign fibroproliferative skin tumor. The respective functions of fibroblasts and vascular endothelial cells in keloid have not been fully studied. The purpose of this study is to identify the respective roles and key genes of fibroblasts and vascular endothelial cells in keloids, which can be used as new targets for diagnosis or treatment.The microarray datasets of keloid fibroblasts and vascular endothelial cells were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were screened out. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used for functional enrichment analysis. The search tool for retrieval of interacting genes and Cytoscape were used to construct protein-protein interaction (PPI) networks and analyze gene modules. The hub genes were screened out, and the relevant interaction networks and biological process analysis were carried out.In fibroblasts, the DEGs were significantly enriched in collagen fibril organization, extracellular matrix organization and ECM-receptor interaction. The PPI network was constructed, and the most significant module was selected, which is mainly enriched in ECM-receptor interaction. In vascular endothelial cells, the DEGs were significantly enriched in cytokine activity, growth factor activity and transforming growth factor-β (TGF-β) signaling pathway. Module analysis was mainly enriched in TGF-β signaling pathway. Hub genes were screened out separately.In summary, the DEGs and hub genes discovered in this study may help us understand the molecular mechanisms of keloid, and provide potential targets for diagnosis and treatment.
Collapse
Affiliation(s)
- Weihan Zheng
- School of Basic Medicine, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Guojian Lin
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, PR China
| | - Zhizhou Wang
- School of Basic Medicine, Fujian Medical University, Fuzhou, Fujian, PR China
| |
Collapse
|
23
|
Metformin Inhibits Transforming Growth Factor β-Induced Fibrogenic Response of Human Dermal Fibroblasts and Suppresses Fibrosis in Keloid Spheroids. Ann Plast Surg 2021; 89:340-341. [PMID: 34670977 DOI: 10.1097/sap.0000000000003002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
24
|
Sawant M, Hinz B, Schönborn K, Zeinert I, Eckes B, Krieg T, Schuster R. A story of fibers and stress: Matrix-embedded signals for fibroblast activation in the skin. Wound Repair Regen 2021; 29:515-530. [PMID: 34081361 DOI: 10.1111/wrr.12950] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/13/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022]
Abstract
Our skin is continuously exposed to mechanical challenge, including shear, stretch, and compression. The extracellular matrix of the dermis is perfectly suited to resist these challenges and maintain integrity of normal skin even upon large strains. Fibroblasts are the key cells that interpret mechanical and chemical cues in their environment to turnover matrix and maintain homeostasis in the skin of healthy adults. Upon tissue injury, fibroblasts and an exclusive selection of other cells become activated into myofibroblasts with the task to restore skin integrity by forming structurally imperfect but mechanically stable scar tissue. Failure of myofibroblasts to terminate their actions after successful repair or upon chronic inflammation results in dysregulated myofibroblast activities which can lead to hypertrophic scarring and/or skin fibrosis. After providing an overview on the major fibrillar matrix components in normal skin, we will interrogate the various origins of fibroblasts and myofibroblasts in the skin. We then examine the role of the matrix as signaling hub and how fibroblasts respond to mechanical matrix cues to restore order in the confusing environment of a healing wound.
Collapse
Affiliation(s)
- Mugdha Sawant
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Katrin Schönborn
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Isabel Zeinert
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Beate Eckes
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Thomas Krieg
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Ronen Schuster
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Canada.,PhenomicAI, MaRS Centre, 661 University Avenue, Toronto, Canada
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW The loss of contractile function after heart injury remains one of the major healthcare issues of our time. One strategy to deal with this problem would be to increase the number of cardiomyocytes to enhance cardiac function. In the last couple of years, reactivation of cardiomyocyte proliferation has repeatedly demonstrated to aid in functional recovery after cardiac injury. RECENT FINDINGS The Tgf-β superfamily plays key roles during development of the heart and populating the embryonic heart with cardiomyocytes. In this review, we discuss the role of Tgf-β signaling in regulating cardiomyocyte proliferation during development and in the setting of cardiac regeneration. Although various pathways to induce cardiomyocyte proliferation have been established, the extent to which cardiomyocyte proliferation requires or involves activation of the Tgf-β superfamily is not entirely clear. More research is needed to better understand cross-talk between pathways that regulate cardiomyocyte proliferation.
Collapse
Affiliation(s)
- Daniel W Sorensen
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.,Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Jop H van Berlo
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA. .,Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA. .,Integrative Biology and Physiology graduate program, University of Minnesota, Minneapolis, MN, USA. .,Cancer and Cardiovascular Research Building, University of Minnesota, 2231 6th St SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
26
|
Jeon HB, Roh H, Ahn HM, Lee JH, Yun CO, Roh TS, Lee WJ. Metformin Inhibits Transforming Growth Factor β-Induced Fibrogenic Response of Human Dermal Fibroblasts and Suppresses Fibrosis in Keloid Spheroids. Ann Plast Surg 2021; 86:406-411. [PMID: 33141769 DOI: 10.1097/sap.0000000000002574] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Accumulation of excessive extracellular matrix (ECM) and aberrant transforming growth factor β (TGF-β) signaling pathway function can be potential therapeutic targets for keloid treatment. In this study, we examined the antifibrotic effect of metformin as a suppressor of TGF-β signaling pathways in human dermal fibroblasts (HDFs) and keloid spheroids. Human dermal fibroblasts were stimulated with TGF-β (10 ng/mL) and treated with metformin (10 mM). The mRNA and protein expression of ECM components were evaluated by quantitative polymerase chain reaction, western blot, and immunofluorescence assay. In addition, we immunohistochemically examined the expression levels of ECM proteins in keloid spheroids. After addition of metformin (10 mM), collagen types I and III and elastin mRNA levels were significantly decreased in HDFs, and collagen type I protein level was significantly decreased. In addition, the expression levels of collagen types I and III, fibronectin, and elastin were significantly reduced in keloid spheroids after treatment with metformin (100 mM). Collagen types I and III and p-Smad2/3 complex proteins were decreased in metformin-treated keloid spheroids. These findings indicated that metformin inhibits the expression of ECM components in TGF-β-stimulated HDFs and keloid spheroids. Therefore, we suggest the potential of metformin as an effective agent for the treatment of keloids.
Collapse
Affiliation(s)
- Hong Bae Jeon
- From the Department of Medicine, Yonsei University Graduate School
| | - Hyun Roh
- Institute for Human Tissue Restoration and Department of Plastic & Reconstructive Surgery, Severance Hospital, Yonsei University College of Medicine
| | - Hyo Min Ahn
- Department of Bioengineering, College of Engineering, Hanyang University
| | - Ju Hee Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Chae Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University
| | - Tai Suk Roh
- Institute for Human Tissue Restoration and Department of Plastic & Reconstructive Surgery, Severance Hospital, Yonsei University College of Medicine
| | - Won Jai Lee
- Institute for Human Tissue Restoration and Department of Plastic & Reconstructive Surgery, Severance Hospital, Yonsei University College of Medicine
| |
Collapse
|
27
|
Li Q, Cheng F, Zhou K, Fang L, Wu J, Xia Q, Cen Y, Chen J, Qing Y. Increased sensitivity to TNF-α promotes keloid fibroblast hyperproliferation by activating the NF-κB, JNK and p38 MAPK pathways. Exp Ther Med 2021; 21:502. [PMID: 33791011 PMCID: PMC8005672 DOI: 10.3892/etm.2021.9933] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 01/18/2021] [Indexed: 02/05/2023] Open
Abstract
Hyperproliferation of fibroblasts is the main cause of keloid formation. However, the pathogenesis of keloids has yet to be fully elucidated. Tumor necrosis factor (TNF)-α may play an important role in the formation and proliferation of keloids, as it is implicated in the pathogenesis of various fibrous disorders. In the present study, the expression level of TNF-α and its receptors, soluble TNF receptor (sTNFR)1 and sTNFR2, in the peripheral blood and skin tissues was detected by ELISA, reverse transcription-quantitative PCR or immunohistochemistry. There was no statistically significant difference in the expression of TNF-α and sTNFR2 in the peripheral blood and skin tissues between patients with keloids and healthy participants (P>0.05), while the sTNFR1 mRNA level in fibroblasts cultured in vitro and its protein level in keloid skin samples were significantly higher compared with those in normal skin (P<0.05). Subsequently, TNF-α recombinant protein was used to treat keloid-derived and normal skin fibroblasts, and it was observed that TNF-α promoted the proliferation of keloid fibroblasts (KFs), but had little effect on normal skin fibroblasts. Furthermore, it was observed that TNF-α stimulation led to the activation of the nuclear factor (NF)-κB, c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) pathways in KFs. In conclusion, KFs exhibited increased expression of sTNFR1, which may contribute to the increased sensitivity to TNF-α, resulting in low concentrations of TNF-α activating the NF-κB, JNK and p38 MAPK pathways, thereby promoting the sustained and excessive proliferation of KFs.
Collapse
Affiliation(s)
- Qijie Li
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Fengrui Cheng
- Department of Plastic and Burn Surgery, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Kai Zhou
- Department of Plastic and Burn Surgery, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lu Fang
- Department of Plastic and Burn Surgery, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Junliang Wu
- Department of Plastic and Burn Surgery, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qingjie Xia
- Department of Anesthesiology, Institute of Neurological Diseases, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ying Cen
- Department of Plastic and Burn Surgery, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Junjie Chen
- Department of Plastic and Burn Surgery, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yong Qing
- Department of Plastic and Burn Surgery, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
28
|
Marty P, Chatelain B, Lihoreau T, Tissot M, Dirand Z, Humbert P, Senez C, Secomandi E, Isidoro C, Rolin G. Halofuginone regulates keloid fibroblast fibrotic response to TGF-β induction. Biomed Pharmacother 2021; 135:111182. [PMID: 33433355 DOI: 10.1016/j.biopha.2020.111182] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/14/2020] [Accepted: 12/26/2020] [Indexed: 01/06/2023] Open
Abstract
Keloids are characterized by increased deposition of fibrous tissue in the skin and subcutaneous tissue following an abnormal wound healing process. Although keloid etiology is yet to be fully understood, fibroblasts are known to be key players in its development. Here we analyze the antifibrotic mechanisms of Halofuginone (HF), a drug reportedly able to inhibit the TGF-β1-Smad3 pathway and to attenuate collagen synthesis, in an in-vitro keloid model using patient-derived Keloid Fibroblasts (KFs) isolated from fibrotic tissue collected during the "Scar Wars" clinical study (NCT NCT03312166). TGF-β1 was used as a pro-fibrotic agent to stimulate fibroblasts response under HF treatment. The fibrotic related properties of KFs, including survival, migration, proliferation, myofibroblasts conversion, ECM synthesis and remodeling, were investigated in 2D and 3D cultures. HF at 50 nM concentration impaired KFs proliferation, and decreased TGF-β1-induced expression of α-SMA and type I procollagen production. HF treatment also reduced KFs migration, prevented matrix contraction and increased the metallo-proteases/inhibitors (MMP/TIMP) ratio. Overall, HF elicits an anti-fibrotic contrasting the TGF-β1 stimulation of KFs, thus supporting its therapeutic use for keloid prevention and management.
Collapse
Affiliation(s)
- Pierre Marty
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25000, Besançon, France; Service de Chirurgie Maxillo-faciale, Stomatologie et Odontologie Hospitalière, CHU Besançon, F-25000, Besançon, France
| | - Brice Chatelain
- Service de Chirurgie Maxillo-faciale, Stomatologie et Odontologie Hospitalière, CHU Besançon, F-25000, Besançon, France
| | | | - Marion Tissot
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25000, Besançon, France
| | - Zélie Dirand
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25000, Besançon, France
| | - Philippe Humbert
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25000, Besançon, France
| | - Clémence Senez
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25000, Besançon, France
| | - Eleonora Secomandi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy.
| | - Gwenaël Rolin
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25000, Besançon, France; INSERM CIC-1431, CHU Besançon, F-25000, Besançon, France.
| |
Collapse
|
29
|
Tsiapalis D, Kearns S, Kelly JL, Zeugolis DI. Growth factor and macromolecular crowding supplementation in human tenocyte culture. BIOMATERIALS AND BIOSYSTEMS 2021; 1:100009. [PMID: 36825160 PMCID: PMC9934496 DOI: 10.1016/j.bbiosy.2021.100009] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 11/18/2020] [Accepted: 01/22/2021] [Indexed: 01/20/2023] Open
Abstract
Cell-assembled tissue engineering strategies hold great potential in regenerative medicine, as three-dimensional tissue-like modules can be produced, even from a patient's own cells. However, the development of such implantable devices requires prolonged in vitro culture time, which is associated with cell phenotypic drift. Considering that the cells in vivo are subjected to numerous stimuli, multifactorial approaches are continuously gaining pace towards controlling cell fate during in vitro expansion. Herein, we assessed the synergistic effect of simultaneous and serial growth factor supplementation (insulin growth factor-1, platelet-derived growth factor ββ, growth differentiation factor 5 and transforming growth factor β3) to macromolecular crowding (carrageenan) in human tenocyte function; collagen synthesis and deposition; and gene expression. TGFβ3 supplementation (without/with carrageenan) induced the highest (among all groups) DNA content. In all cases, tenocyte proliferation was significantly increased as a function of time in culture, whilst metabolic activity was not affected. Carrageenan supplementation induced significantly higher collagen deposition than groups without carrageenan (without/with any growth factor). Of all the growth factors used, TGFβ3 induced the highest collagen deposition when used together with carrageenan in both simultaneous and serial fashion. At day 13, gene expression analysis revealed that TGFβ3 in serial supplementation to carrageenan upregulated the most and downregulated the least collagen- and tendon- related genes and upregulated the least and downregulated the most osteo-, chondro-, fibrosis- and adipose- related trans-differentiation genes. Collectively, these data clearly advocate the beneficial effects of multifactorial approaches (in this case, growth factor and macromolecular crowding supplementation) in the development of functional cell-assembled tissue surrogates.
Collapse
Affiliation(s)
- Dimitrios Tsiapalis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | | | | | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Lugano, Switzerland
- Corresponding authors.
| |
Collapse
|
30
|
Mirmajidi T, Chogan F, Rezayan AH, Sharifi AM. In vitro and in vivo evaluation of a nanofiber wound dressing loaded with melatonin. Int J Pharm 2021; 596:120213. [PMID: 33493599 DOI: 10.1016/j.ijpharm.2021.120213] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/19/2020] [Accepted: 12/25/2020] [Indexed: 12/12/2022]
Abstract
Wound healing is a complicated process that takes a long time to complete. The three-layer nanofiber wound dressing containing melatonin is highly expected to show remarkable wound repair by reducing the wound healing time. In this study, chitosan (Cs)-polycaprolactone (PCL)/ polyvinylalcohol (PVA)-melatonin (MEL)/ chitosan-polycaprolactone three-layer nanofiber wound dressing was prepared by electrospinning for melatonin sustained release. The characteristics of the wound dressing were further evaluated. The wound dressing had a high water uptake after 24 h (401%), and the water contact angle results showed that it had hydrophilicity effect that supported the cell attachment. The wound healing effect of wound dressing was examined using a full-thickness excisional model of rat skin by the local administration of MEL. The gene expressions of transforming growth factor-beta (TGF-β1), alpha-smooth muscle actin (α-SMA), collagen type I (COL1A1), and collagen type III (COL3A1) were further studied. The histopathological evaluation showed the complete regeneration of the epithelial layer, remodeling of wounds, collagen synthesis, and reduction in inflammatory cells. The NF + 20% MEL significantly increased TGF-β1, COL1A1, COL3A1, and α-SMA mRNA expressions. This wound dressing may have a considerable potential as a wound dressing to accelerate the wound healing.
Collapse
Affiliation(s)
- Tahereh Mirmajidi
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, P.O. Box 14395-1561, Tehran, Iran
| | - Faraz Chogan
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, P.O. Box 14395-1561, Tehran, Iran
| | - Ali Hossein Rezayan
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, P.O. Box 14395-1561, Tehran, Iran.
| | - Ali Mohammad Sharifi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran; Razi Drug Research Center, Department of Pharmacology, Iran University of Medical Sciences, Tehran, Iran; Tissue Engineering Group (NOCERAL), Department of Orthopedics Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
31
|
Dual inhibition of HDAC and tyrosine kinase signaling pathways with CUDC-907 attenuates TGFβ1 induced lung and tumor fibrosis. Cell Death Dis 2020; 11:765. [PMID: 32943605 PMCID: PMC7499263 DOI: 10.1038/s41419-020-02916-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/17/2022]
Abstract
TGFβ1 signaling is a critical driver of collagen accumulation in pulmonary fibrotic diseases and a well-characterized regulator of cancer associated fibroblasts (CAF) activation in lung cancer. Myofibroblasts induced by TGFβ1 and other factors are key players in the pathogenesis of lung fibrosis and tumor. Tremendous attention has been gained to targeting myofibroblasts in order to inhibit the progression of fibrosis and myofibroblast-induced tumor progression and metastasis. Here we determined the therapeutic efficacy of simultaneously targeting PI3K and HDAC pathways in lung myofibroblasts and CAF with a single agent and to evaluate biomarkers of treatment response. CUDC-907 is a first-in-class compound, functioning as a dual inhibitor of HDACs and PI3K/AKT pathway. We investigated its effects in counteracting the activity of TGFβ1-induced myofibroblasts/CAF in regard to cell proliferation, migration, invasion, apoptosis in vitro antifibrosis efficiency in vivo. We found that CUDC-907 inhibited myofibroblasts/CAF cell proliferation, migration and apoptosis in a dose-dependent manner and caused cell cycle arrest at G1-S phase. CUDC-907 not only inhibited myofibroblasts markers expression, but also significantly inhibited the phosphorylation level of AKT, mTOR, Smad2/3, and promoted acetylation of histones. Furthermore, the observed inhibitory effect was also confirmed in bleomycin-induced mice lung fibrosis and nude mouse transplanted tumor model. Overall, these data suggest that dual inhibition of HDAC and the tyrosine kinase signaling pathways with CUDC-907 is a promising treatment strategy for TGFβ1-induced lung and tumor fibrosis.
Collapse
|
32
|
Zhou BY, Wang WB, Wu XL, Zhang WJ, Zhou GD, Gao Z, Liu W. Nintedanib inhibits keloid fibroblast functions by blocking the phosphorylation of multiple kinases and enhancing receptor internalization. Acta Pharmacol Sin 2020; 41:1234-1245. [PMID: 32327724 PMCID: PMC7608201 DOI: 10.1038/s41401-020-0381-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 02/13/2020] [Indexed: 01/22/2023] Open
Abstract
Keloid is a benign skin tumor characterized by its cell hyperproliferative activity, invasion into normal skin, uncontrolled growth, overproduction and deposition of extracellular matrices and high recurrence rate after various therapies. Nintedanib is a receptor tyrosine kinase inhibitor targeting VEGF, PDGF, FGF, and TGF-β receptors with proved efficacy in anti-angiogenesis and in treating various types of cancers. In this study, we investigated the effects of nintedanib on keloid fibroblasts in both in vitro and ex vivo models. Keloid fibroblasts were prepared from 54 keloid scar samples in active stages collected from 49 patients. We found that nintedanib (1−4 μM) dose-dependently suppressed cell proliferation, induced G0/G1 cell cycle arrest, and inhibited migration and invasion of keloid fibroblasts. The drug also significantly inhibited the gene and protein expression of collagen I (COL-1) and III (COL-3), fibronectin (FN), and connective growth factor (CTGF), as well as the gene expression of other pathological factors, such as alpha smooth muscle actin (α-SMA), plasminogen activator inhibitor-1 (PAI-1), FK506-binding protein 10 (FKBP10), and heat shock protein 47 (HSP47) in keloid fibroblasts. Furthermore, nintedanib treatment significantly suppressed the phosphorylation of p38, JNK, ERK, STAT3, and Smad, enhanced endocytosis of various growth factor receptors. Using an ex vivo tissue explant model, we showed that nintedanib significantly suppressed cell proliferation, migration, and collagen production. The drug also significantly disrupted microvessel structure ex vivo. In summary, our results demonstrate that nintedanib is likely to become a potential targeted drug for keloid systemic therapy.
Collapse
|
33
|
Kim HS, Chen J, Wu LP, Wu J, Xiang H, Leong KW, Han J. Prevention of excessive scar formation using nanofibrous meshes made of biodegradable elastomer poly(3-hydroxybutyrate- co-3-hydroxyvalerate). J Tissue Eng 2020; 11:2041731420949332. [PMID: 32922720 PMCID: PMC7448259 DOI: 10.1177/2041731420949332] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 07/23/2020] [Indexed: 11/24/2022] Open
Abstract
To reduce excessive scarring in wound healing, electrospun nanofibrous meshes, composed of haloarchaea-produced biodegradable elastomer poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV), are fabricated for use as a wound dressing. Three PHBV polymers with different 3HV content are used to prepare either solution-cast films or electrospun nanofibrous meshes. As 3HV content increases, the crystallinity decreases and the scaffolds become more elastic. The nanofibrous meshes exhibit greater elasticity and elongation at break than films. When used to culture human dermal fibroblasts in vitro, PHBV meshes give better cell attachment and proliferation, less differentiation to myofibroblasts, and less substrate contraction. In a full-thickness mouse wound model, treatment with films or meshes enables regeneration of pale thin tissues without scabs, dehydration, or tubercular scar formation. The epidermis of wounds treated with meshes develop small invaginations in the dermis within 2 weeks, indicating hair follicle and sweat gland regeneration. Consistent with the in vitro results, meshes reduce myofibroblast differentiation in vivo through downregulation of α-SMA and TGF-β1, and upregulation of TGF-β3. The regenerated wounds treated with meshes are softer and more elastic than those treated with films. These results demonstrate that electrospun nanofibrous PHBV meshes mitigate excessive scar formation by regulating myofibroblast formation, showing their promise for use as wound dressings.
Collapse
Affiliation(s)
- Hye Sung Kim
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.,Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Republic of Korea
| | - Junyu Chen
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Lin-Ping Wu
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jihua Wu
- PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Hua Xiang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jing Han
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
34
|
Rakshit M, Gautam A, Toh LZ, Lee YS, Lai HY, Wong TT, Ng KW. Hydroxyapatite Particles Induced Modulation of Collagen Expression and Secretion in Primary Human Dermal Fibroblasts. Int J Nanomedicine 2020; 15:4943-4956. [PMID: 32764927 PMCID: PMC7367744 DOI: 10.2147/ijn.s245500] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/09/2020] [Indexed: 01/31/2023] Open
Abstract
Background Hydroxyapatite (HA) [Ca5(PO4)3(OH)] is a naturally occurring calcium phosphate which makes up 60–70% of the dry weight of human bones. Nano-scale HA particles are increasingly being used as carriers for controlled and targeted delivery of bioactive agents like drugs, proteins, and nucleic acids due to their high porosity, negative charge, and biodegradability. Purpose Although much effort has been devoted to understanding the delivery kinetics and effects of the payloads in such carriers, a thorough understanding of the influence of the carriers themselves is lacking. Methods HA particles (300 µg/mL) were administered to primary human dermal fibroblasts (HDFs). The uptake and intracellular localization of the particles were determined by flow cytometry, confocal imaging, and transmission electron microscopy (TEM). Immunological assays and PCR were performed to determine the levels of pro-inflammatory cytokines and collagens in cell lysates and media supernatant. Results The current study explores the effects of poly-dispersed HA particles on primary HDFs as a model system. The majority of the particles were determined to range between 150 and 200 nm in diameter. Upon exposure to HA suspensions, primary HDFs internalized the particles by endocytosis within 6 hours of exposure, showing maximum uptake at 72 hours following which the particles were exocytosed by 168 hours. This correlated to reduced secretion of various pro-inflammatory and pro-collagenic cytokines. Biochemical analysis further revealed a reduction in Type I collagen expression and secretion. Conclusion HA particles have an immune-modulatory effect on dermal fibroblasts and reduce collagen production, which may impact the integrity of the extracellular matrix (ECM). This study demonstrates the need to consider the secondary effects of particulate carriers like HA, beyond basic cytotoxicity, in the specific tissue environment where the intended function is to be realized.
Collapse
Affiliation(s)
- Moumita Rakshit
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Archana Gautam
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Li Zhen Toh
- Ocular Therapeutics and Drug Delivery, Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Ying Shi Lee
- Ocular Therapeutics and Drug Delivery, Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Hui Ying Lai
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Tina T Wong
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore.,Ocular Therapeutics and Drug Delivery, Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Kee Woei Ng
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore.,Environmental Chemistry and Materials Centre, Nanyang Environment & Water Research Institute, Singapore 637141, Singapore.,Skin Research Institute of Singapore, Singapore 138648, Singapore.,Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Cambridge, MA 02115, USA
| |
Collapse
|
35
|
Limandjaja GC, Niessen FB, Scheper RJ, Gibbs S. The Keloid Disorder: Heterogeneity, Histopathology, Mechanisms and Models. Front Cell Dev Biol 2020; 8:360. [PMID: 32528951 PMCID: PMC7264387 DOI: 10.3389/fcell.2020.00360] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/22/2020] [Indexed: 12/22/2022] Open
Abstract
Keloids constitute an abnormal fibroproliferative wound healing response in which raised scar tissue grows excessively and invasively beyond the original wound borders. This review provides a comprehensive overview of several important themes in keloid research: namely keloid histopathology, heterogeneity, pathogenesis, and model systems. Although keloidal collagen versus nodules and α-SMA-immunoreactivity have been considered pathognomonic for keloids versus hypertrophic scars, conflicting results have been reported which will be discussed together with other histopathological keloid characteristics. Importantly, histopathological keloid abnormalities are also present in the keloid epidermis. Heterogeneity between and within keloids exists which is often not considered when interpreting results and may explain discrepancies between studies. At least two distinct keloid phenotypes exist, the superficial-spreading/flat keloids and the bulging/raised keloids. Within keloids, the periphery is often seen as the actively growing margin compared to the more quiescent center, although the opposite has also been reported. Interestingly, the normal skin directly surrounding keloids also shows partial keloid characteristics. Keloids are most likely to occur after an inciting stimulus such as (minor and disproportionate) dermal injury or an inflammatory process (environmental factors) at a keloid-prone anatomical site (topological factors) in a genetically predisposed individual (patient-related factors). The specific cellular abnormalities these various patient, topological and environmental factors generate to ultimately result in keloid scar formation are discussed. Existing keloid models can largely be divided into in vivo and in vitro systems including a number of subdivisions: human/animal, explant/culture, homotypic/heterotypic culture, direct/indirect co-culture, and 3D/monolayer culture. As skin physiology, immunology and wound healing is markedly different in animals and since keloids are exclusive to humans, there is a need for relevant human in vitro models. Of these, the direct co-culture systems that generate full thickness keloid equivalents appear the most promising and will be key to further advance keloid research on its pathogenesis and thereby ultimately advance keloid treatment. Finally, the recent change in keloid nomenclature will be discussed, which has moved away from identifying keloids solely as abnormal scars with a purely cosmetic association toward understanding keloids for the fibroproliferative disorder that they are.
Collapse
Affiliation(s)
- Grace C. Limandjaja
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center (location VUmc), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Frank B. Niessen
- Department of Plastic Surgery, Amsterdam University Medical Center (location VUmc), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Rik J. Scheper
- Department of Pathology, Amsterdam University Medical Center (location VUmc), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center (location VUmc), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
36
|
Park J, Kim YC. Topical delivery of 5-fluorouracil-loaded carboxymethyl chitosan nanoparticles using microneedles for keloid treatment. Drug Deliv Transl Res 2020; 11:205-213. [PMID: 32383003 DOI: 10.1007/s13346-020-00781-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Keloids are induced by skin injuries such as surgeries, skin piercings, burns, and trauma. The intra-lesional injection of 5-fluorouracil (5-FU) is a promising therapy to treat keloid. However, local 5-FU injections have caused several side effects such as pain at administration and hyperpigmentation. This study suggests a safer and more effective 5-FU delivery system. We used microneedles to treat keloid because this method has the feasibility of self-administration without pain. In this study, 5-FU-loaded carboxymethyl chitosan (CMC) nanoparticles were prepared and characterized by various analytical methods and then coated on stainless solid microneedles. The blank CMC nanoparticles caused an increase in cell viability on human normal fibroblasts to 150%. In particular, the 5-FU-loaded CMC nanoparticles showed a significant inhibitory effect on the human keloid fibroblast to 16%. The intercellular uptake of the 5-FU-loaded CMC nanoparticles was observed on both human normal and keloid fibroblasts by using a confocal microscope. In addition, it was found that the nanoparticles showed an inhibition of TGF-β1 by ELISA. For topical drug delivery, it was confirmed that the nanoparticles coated onto the microneedles were dissolved and diffused at the administration site in the porcine dorsal skin model. According to these results, the suggested microneedle-mediated drug delivery system not only inhibits the human keloid fibroblasts by delivering drugs effectively into the keloids but also has the feasibility to self-administer without pain. Therefore, this new system including 5-FU-loaded CMC nanoparticles and microneedles has the potential to treat keloid scars. Graphical abstract.
Collapse
Affiliation(s)
- Juhyun Park
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yeu-Chun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
37
|
Tang R, Wang YC, Mei X, Shi N, Sun C, Ran R, Zhang G, Li W, Staveley-O'Carroll KF, Li G, Chen SY. LncRNA GAS5 attenuates fibroblast activation through inhibiting Smad3 signaling. Am J Physiol Cell Physiol 2020; 319:C105-C115. [PMID: 32374674 DOI: 10.1152/ajpcell.00059.2020] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Transforming growth factor-β (TGF-β)-induced fibroblast activation is a key pathological event during tissue fibrosis. Long noncoding RNA (lncRNA) is a class of versatile gene regulators participating in various cellular and molecular processes. However, the function of lncRNA in fibroblast activation is still poorly understood. In this study, we identified growth arrest-specific transcript 5 (GAS5) as a novel regulator for TGF-β-induced fibroblast activation. GAS5 expression was downregulated in cultured fibroblasts by TGF-β and in resident fibroblasts from bleomycin-treated skin tissues. Overexpression of GAS5 suppressed TGF-β-induced fibroblast to myofibroblast differentiation. Mechanistically, GAS5 directly bound mothers against decapentaplegic homolog 3 (Smad3) and promoted Smad3 binding to Protein phosphatase 1A (PPM1A), a Smad3 dephosphatase, and thus accelerated Smad3 dephosphorylation in TGF-β-treated fibroblasts. In addition, GAS5 inhibited fibroblast proliferation. Importantly, local delivery of GAS5 via adenoviral vector suppressed bleomycin-induced skin fibrosis in mice. Collectively, our data revealed that GAS5 suppresses fibroblast activation and fibrogenesis through inhibiting TGF-β/Smad3 signaling, which provides a rationale for an lncRNA-based therapy to treat fibrotic diseases.
Collapse
Affiliation(s)
- Rui Tang
- Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia
| | - Yung-Chun Wang
- Department of Surgery, University of Missouri School of Medicine, Columbia, Missouri
| | - Xiaohan Mei
- Department of Surgery, University of Missouri School of Medicine, Columbia, Missouri.,Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia
| | - Ning Shi
- Department of Surgery, University of Missouri School of Medicine, Columbia, Missouri
| | - Chenming Sun
- Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia
| | - Ran Ran
- Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia
| | - Gui Zhang
- Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia
| | - Wenjing Li
- Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia
| | - Kevin F Staveley-O'Carroll
- Department of Surgery, University of Missouri School of Medicine, Columbia, Missouri.,The Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri
| | - Guangfu Li
- Department of Surgery, University of Missouri School of Medicine, Columbia, Missouri
| | - Shi-You Chen
- Department of Surgery, University of Missouri School of Medicine, Columbia, Missouri.,Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia.,Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| |
Collapse
|
38
|
Knockdown of fibronectin extra domain B suppresses TGF-β1-mediated cell proliferation and collagen deposition in keloid fibroblasts via AKT/ERK signaling pathway. Biochem Biophys Res Commun 2020; 526:1131-1137. [PMID: 32317186 DOI: 10.1016/j.bbrc.2020.04.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/08/2020] [Indexed: 01/17/2023]
Abstract
Keloids represent a dermal fibrotic disease characterized by excess collagen deposition and invasion of normal skin beyond the wound boundary, similar to malignant tumor features. Fibronectin extra domain B (EDB) is highly expressed in many tumors but has not been studied in keloids. The present study aimed to investigate the expression and the influence of EDB on keloid and elucidate the putative signaling pathway. We examined expression of EDB and the effects of EDB on fibroblast proliferation, apoptosis and the expression of the related proteins and genes. The level of phosphorylation of Smad, ERK, and AKT was estimated to elucidate the signaling pathways. The results showed that EDB in human keloid tissues and fibroblasts was overexpressed. EDB knockdown suppressed the cell proliferation of keloid fibroblasts (KFs) treated by transforming growth factor-β1 (TGF-β1). Also, the phosphorylation of Smad, ERK, and AKT in TGF-β1-induced KFs was inhibited In addition, the low expression of pro-collagen-I (Col-I) and Col-III protein and mRNA level was observed in the siEDB group. EDB knockdown inhibited cell proliferation and suppressed collagen deposition in TGF-1-induced KFs. The underlying mechanism is the activation of TGF-β1/Smad, ERK, and AKT signaling pathways. Together, the results suggested that EDB is a promising therapeutic target for keloid clinical treatment.
Collapse
|
39
|
Li XY, Weng XJ, Li XJ, Tian XY. TSG-6 Inhibits the Growth of Keloid Fibroblasts Via Mediating the TGF-β1/Smad Signaling Pathway. J INVEST SURG 2020; 34:947-956. [PMID: 31986937 DOI: 10.1080/08941939.2020.1716894] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Xin-Yi Li
- Department of Plastic Surgery, First Affiliated Hospital of Anhui Medical University, China
| | - Xiao-Juan Weng
- Department of Plastic Surgery, First Affiliated Hospital of Anhui Medical University, China
| | - Xiao-Jing Li
- Department of Plastic Surgery, First Affiliated Hospital of Anhui Medical University, China
| | - Xiao-Yu Tian
- Department of Plastic Surgery, First Affiliated Hospital of Anhui Medical University, China
| |
Collapse
|
40
|
Xu JH, Zhao WY, Fang QQ, Wang XF, Zhang DD, Hu YY, Zheng B, Tan WQ. Co-transfection of hepatocyte growth factor and truncated TGF-β type II receptor inhibit scar formation. ACTA ACUST UNITED AC 2020; 53:e9144. [PMID: 31939600 PMCID: PMC6967399 DOI: 10.1590/1414-431x20199144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/28/2019] [Indexed: 11/22/2022]
Abstract
Wound scarring remains a major challenge for plastic surgeons. Transforming growth factor (TGF)-β plays a key role in the process of scar formation. Previous studies have demonstrated that truncated TGF-β type II receptor (t-TGF-βRII) is unable to continue signal transduction but is still capable of binding to TGF-β, thereby blocking the TGF-β signaling pathway. Hepatocyte growth factor (HGF) is a multifunctional growth factor that promotes tissue regeneration and wound healing. Theoretically, the combination of HGF and t-TGF-βRII would be expected to exert a synergistic effect on promoting wound healing and reducing collagen formation. In the present study, lentivirus-mediated transfection of the two genes (t-TGF-βRII/HGF) into fibroblasts in vitro and in a rat model in vivo was used. The results demonstrated that the expression of t-TGF-βRII and HGF in NIH-3T3 cells was successfully induced. The expression of both molecules significantly reduced collagen I and III expression, and also inhibited fibroblast proliferation. Furthermore, histological examination and scar quantification revealed less scarring in the experimental wound in a rat model. Moreover, on macroscopic inspection, the experimental wound exhibited less visible scarring compared with the control. Therefore, the present study demonstrated that the combination gene therapy of t-TGF-βRII and HGF promoted wound healing, with less scarring and more epithelial tissue formation, not only by suppressing the overgrowth of collagen due to its antifibrotic effect, but also by promoting tissue regeneration.
Collapse
Affiliation(s)
- Ji-Hua Xu
- Department of Hand Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wan-Yi Zhao
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qing-Qing Fang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Xiao-Feng Wang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Ding-Ding Zhang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Yan-Yan Hu
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Bin Zheng
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wei-Qiang Tan
- Department of Hand Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
41
|
Eng TY, Abugideiri M, Chen TW, Madden N, Morgan T, Tanenbaum D, Wandrey N, Westergaard S, Xu K, Jane Sudmeier L. Radiation Therapy for Benign Disease: Keloids, Macular Degeneration, Orbital Pseudotumor, Pterygium, Peyronie Disease, Trigeminal Neuralgia. Hematol Oncol Clin North Am 2019; 34:229-251. [PMID: 31739946 DOI: 10.1016/j.hoc.2019.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although the use of ionizing radiation on malignant conditions has been well established, its application on benign conditions has not been fully accepted and has been inadequately recognized by health care providers outside of radiation therapy. Most frequently, radiation therapy in these benign conditions is used along with other treatment modalities, such as surgery, when the condition causes significant disability or could even lead to death. Radiation therapy can be helpful for inflammatory/proliferative disorders. This article discusses the present use of radiation therapy for some of the most common benign conditions.
Collapse
Affiliation(s)
- Tony Y Eng
- Radiation Oncology Department, Winship Cancer Institute of Emory University, 1365 Clifton Road Northeast, Building C, Atlanta, GA 30322, USA.
| | - Mustafa Abugideiri
- Radiation Oncology Department, Winship Cancer Institute of Emory University, 1365 Clifton Road Northeast, Building C, Atlanta, GA 30322, USA
| | - Tiffany W Chen
- Department of Radiation Oncology, University of Texas Health Science Center at San Antonio, 7979 Wurzbach Road, San Antonio, TX 78229, USA
| | - Nicholas Madden
- Radiation Oncology Department, Winship Cancer Institute of Emory University, 1365 Clifton Road Northeast, Building C, Atlanta, GA 30322, USA
| | - Tiffany Morgan
- Radiation Oncology Department, Winship Cancer Institute of Emory University, 1365 Clifton Road Northeast, Building C, Atlanta, GA 30322, USA
| | - Daniel Tanenbaum
- Radiation Oncology Department, Winship Cancer Institute of Emory University, 1365 Clifton Road Northeast, Building C, Atlanta, GA 30322, USA
| | - Narine Wandrey
- Department of Radiation Oncology, University of Texas Health Science Center at San Antonio, 7979 Wurzbach Road, San Antonio, TX 78229, USA
| | - Sarah Westergaard
- Radiation Oncology Department, Winship Cancer Institute of Emory University, 1365 Clifton Road Northeast, Building C, Atlanta, GA 30322, USA
| | - Karen Xu
- Radiation Oncology Department, Winship Cancer Institute of Emory University, 1365 Clifton Road Northeast, Building C, Atlanta, GA 30322, USA
| | - Lisa Jane Sudmeier
- Radiation Oncology Department, Winship Cancer Institute of Emory University, 1365 Clifton Road Northeast, Building C, Atlanta, GA 30322, USA
| |
Collapse
|
42
|
Tu T, Huang J, Lin M, Gao Z, Wu X, Zhang W, Zhou G, Wang W, Liu W. CUDC‑907 reverses pathological phenotype of keloid fibroblasts in vitro and in vivo via dual inhibition of PI3K/Akt/mTOR signaling and HDAC2. Int J Mol Med 2019; 44:1789-1800. [PMID: 31545402 PMCID: PMC6777681 DOI: 10.3892/ijmm.2019.4348] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/02/2019] [Indexed: 02/06/2023] Open
Abstract
Keloids are benign skin tumors with a high recurrence rate following surgical excision. Abnormal intracellular signaling is one of the key mechanisms involved in its pathogenesis. Over-activated phosphoinositide 3-kinase/RAC-alpha serine/threonine-protein kinase/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway and overproduction of histone deacetylases 2 (HDAC2) have also been observed in keloid fibroblasts (KFs). The present study aimed to explore the possibility of reversing the KF pathological phenotype using CUDC-907, a dual inhibitor of PI3K/Akt/mTOR pathway and HDACs. KFs and keloid xenografts were treated with CUDC-907 to examine its inhibitory effects on the pathological activities of KFs in vitro and in vivo. CUDC-907 inhibited cell proliferation, migration, invasion and extracellular matrix deposition of in vitro cultured KFs and also suppressed collagen accumulation and disrupted the capillaries of keloid explants ex vivo and in vivo. A mechanistic study of CUDC-907 revealed the initiation of cell cycle arrest at G2/M phase along with the enhanced expression of cyclin-dependent kinase inhibitor 1 and decreased expression of cyclin B in cells treated with CUDC-907. CUDC-907 not only inhibited AKT and mTOR phosphorylation and promoted the acetylation of histone H3, but also significantly inhibited the phosphorylation levels of Smad2/3 and Erk. These preclinical data demonstrating its anti-keloid effects suggest that CUDC-907 may represent a candidate drug for systemic keloid therapy.
Collapse
Affiliation(s)
- Tian Tu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Jia Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Miaomiao Lin
- Department of Otolaryngology, Suzhou First People's Hospital, Suzhou, Anhui 234000, P.R. China
| | - Zhen Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Xiaoli Wu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Wenbo Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
43
|
Bertelsen C, Shoffel-Havakuk H, O'Dell K, Johns MM, Reder LS. Serial In-Office Intralesional Steroid Injections in Airway Stenosis. JAMA Otolaryngol Head Neck Surg 2019; 144:203-210. [PMID: 29327045 DOI: 10.1001/jamaoto.2017.2800] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Importance Endoscopic dilation is the mainstay treatment strategy for subglottic and proximal tracheal stenosis (SGS/PTS). Its major limitation is restenosis requiring repeated surgery. Intralesional steroid injection (ISI) is a promising adjunctive treatment aimed at prolonging the effects of dilation. Objective To evaluate the association of serial in-office ISI after endoscopic dilation with surgery-free interval (SFI) in adults with SGS/PTS. Design, Setting, and Participants A retrospective study of adults with SGS/PTS who underwent at least 2 consecutive in-office ISI at the University of Southern California, Keck School of Medicine, over a 3-year period was conducted. Exposure Serial ISI with triamcinolone 40 mg/mL using topical anesthesia, spaced 3 to 6 weeks apart. Main Outcomes and Measures Surgery-free interval, number of dilations, need for open airway surgery, decannulation rate, and adverse events. Patients with previous dilations and sufficient follow-up time were included in a comparative analysis of SFI before and after ISI. The Mann-Whitney U test was applied for comparisons. Results Twenty-four patients met eligibility criteria. Mean (SD) age was 50.1 (15.1) years; 18 (75%) were female. Ten (42%) patients had idiopathic, 8 (33%) had traumatic, and 6 (25%) had rheumatologic-related SGS/PTS. Mean (SD) follow-up time was 32.3 (33.4) months. Patients underwent mean (SD) 4.08 (1.91) injections. Seventeen (71%) patients have not undergone further surgery after ISI. Mean (SD) SFI was 17.8 (12.8) months overall and was 15.7 (10.6) months for idiopathic, 13.8 (9.9) for traumatic, and 26.7 (16.9) for rheumatologic-related SGS/PTS. Twenty-one (88%) patients underwent dilation(s) prior to ISI. Among patients who fulfilled eligibility criteria for comparison of SFI before and after ISI, SFI improved from 10.1 months before, to 22.6 months after ISI (mean difference, 12.5 months; 95% CI, -2.1 to 27.2 months). Three of 6 patients (all with traumatic SGS/PTS) presenting with a tracheotomy were decannulated. No patients required open airway surgery after ISI. There were no adverse events associated with ISI. Conclusions and Relevance Serial in-office ISI are safe and well-tolerated in adults with SGS/PTS. This technique can reduce the surgical burden on these patients and may obviate the need for future airway intervention.
Collapse
Affiliation(s)
- Caitlin Bertelsen
- Division of Laryngology, USC Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of USC, Los Angeles, California
| | - Hagit Shoffel-Havakuk
- Division of Laryngology, USC Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of USC, Los Angeles, California
| | - Karla O'Dell
- Division of Laryngology, USC Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of USC, Los Angeles, California
| | - Michael M Johns
- Division of Laryngology, USC Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of USC, Los Angeles, California
| | - Lindsay S Reder
- Division of Laryngology, USC Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of USC, Los Angeles, California
| |
Collapse
|
44
|
Triamcinolone Acetonide Suppresses Keloid Formation Through Enhancing Apoptosis in a Nude Mouse Model. Ann Plast Surg 2019; 83:S50-S54. [PMID: 31513066 DOI: 10.1097/sap.0000000000002090] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Current understanding of steroid treatments for keloids is in regards to modulation of inflammation, proliferation, and apoptosis, with no in vivo study on the latter. Using a nude mouse model, we investigated whether triamcinolone acetonide (TA) injections induce keloids regression through enhancing apoptosis. MATERIALS AND METHODS Thirty-six keloid specimens (1 × 1 cm) were harvested from 6 patients and separated into sets of 2 from the same patient: no treatment and intralesional TA injection (0.4 mg/mL/kg) at 8 weeks of postimplantation. One set was implanted in each of 18 randomly selected nude mice, which were separated into 3 groups based on time of keloid harvesting after treatment: group A, 2 weeks; group B, 8 weeks; and group C, 14 weeks. Each group had 1 set of specimen from each patient. Histological staining was performed with hematoxylin and eosin stain. Immunohistochemistry staining was performed for human-prolyl 4-hydroxylase (hPH4) and caspase 3 protein, along with terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. RESULTS All keloid specimens survived, with no noted overgrowth. Hematoxylin and eosin staining revealed dense extracellular matrix and viable fibroblasts, and hPH4 immunohistochemistry revealed strong expression, demonstrating keloid viability. Caspase 3 protein and TUNEL expressions were significantly increased in the treatment versus control groups, demonstrating that TA injections induced apoptosis. CONCLUSIONS Triamcinolone acetonide intralesional injections significantly increased apoptosis in keloids, represented by increased caspase 3 protein and TUNEL expressions, supporting that steroids suppress keloids in part owing to enhancement of apoptosis.
Collapse
|
45
|
Antifibrotic Effects of High-Mobility Group Box 1 Protein Inhibitor (Glycyrrhizin) on Keloid Fibroblasts and Keloid Spheroids through Reduction of Autophagy and Induction of Apoptosis. Int J Mol Sci 2019; 20:ijms20174134. [PMID: 31450620 PMCID: PMC6747471 DOI: 10.3390/ijms20174134] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 01/07/2023] Open
Abstract
Overabundance of extracellular matrix resulting from hyperproliferation of keloid fibroblasts (KFs) and dysregulation of apoptosis represents the main pathophysiology underlying keloids. High-mobility group box 1 (HMGB1) plays important roles in the regulation of cellular death. Suppression of HMGB1 inhibits autophagy while increasing apoptosis. Suppression of HMGB1 with glycyrrhizin has therapeutic benefits in fibrotic diseases. In this study, we explored the possible involvement of autophagy and HMGB1 as a cell death regulator in keloid pathogenesis. We have highlighted the potential utility of glycyrrhizin as an antifibrotic agent via regulation of the aberrant balance between autophagy and apoptosis in keloids. Higher HMGB1 expression and enhanced autophagy were observed in keloids. The proliferation of KFs was decreased following glycyrrhizin treatment. While apoptosis was enhanced in keloids after glycyrrhizin treatment, autophagy was significantly reduced. The expressions of ERK1/2, Akt, and NF-κB, were enhanced in HMGB1-teated fibroblasts, but decreased following glycyrrhizin treatment. The expression of extracellular matrix (ECM) components was reduced in glycyrrhizin-treated keloids. TGF-β, Smad2/3, ERK1/2, and HMGB1 were decreased in glycyrrhizin-treated keloids. Treatment with the autophagy inhibitor 3-MA resulted in a decrease of autophagy markers and collagen in the TGF-β-treated fibroblasts. The results indicated that autophagy plays an important role in the pathogenesis of keloids. Because glycyrrhizin appears to reduce ECM and downregulate autophagy in keloids, its potential use for treatment of keloids is indicated.
Collapse
|
46
|
Tan S, Khumalo N, Bayat A. 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 2019; 10:1810. [PMID: 31440236 PMCID: PMC6692789 DOI: 10.3389/fimmu.2019.01810] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 07/17/2019] [Indexed: 01/01/2023] Open
Abstract
Keloids are considered as benign fibroproliferative skin tumors growing beyond the site of the original dermal injury. Although traditionally viewed as a form of skin scarring, keloids display many cancer-like characteristics such as progressive uncontrolled growth, lack of spontaneous regression and extremely high rates of recurrence. Phenotypically, keloids are consistent with non-malignant dermal tumors that are due to the excessive overproduction of collagen which never metastasize. Within the remit of keloid pathobiology, there is increasing evidence for the various interplay of neoplastic-promoting and suppressing factors, which may explain its aggressive clinical behavior. Amongst the most compelling parallels between keloids and cancer are their shared cellular bioenergetics, epigenetic methylation profiles and epithelial-to-mesenchymal transition amongst other disease biological (genotypic and phenotypic) behaviors. This review explores the quasi-neoplastic or cancer-like properties of keloids and highlights areas for future study.
Collapse
Affiliation(s)
- Silvian Tan
- Plastic and Reconstructive Surgery Research, Centre for Dermatology Research, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, United Kingdom
| | - Nonhlanhla Khumalo
- Hair and Skin Research Laboratory, Department of Dermatology, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Ardeshir Bayat
- Plastic and Reconstructive Surgery Research, Centre for Dermatology Research, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, United Kingdom
- Hair and Skin Research Laboratory, Department of Dermatology, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
47
|
Petrou IG, Jugun K, Rüegg EM, Zilli T, Modarressi A, Pittet-Cuénod B. Keloid treatment: what about adjuvant radiotherapy? Clin Cosmet Investig Dermatol 2019; 12:295-301. [PMID: 31190938 PMCID: PMC6526192 DOI: 10.2147/ccid.s202884] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/01/2019] [Indexed: 11/23/2022]
Abstract
Background: Keloids are debilitating fibrous skin proliferations with a high recurrence rate after surgical treatment. Postoperative radiotherapy (PORT) is a well-tolerated adjuvant treatment to reduce the risk of recurrence, but the optimal regimen for this combined treatment remains unknown. The aim of this study is to evaluate the efficacy of combining surgical excision and immediate PORT. Methods: We retrospectively reviewed the records of patients with keloid lesions treated with adjuvant PORT in the period 2005–2014 at Geneva University Hospitals. Main outcomes were the rates of complications and recurrence in patients with a minimal follow-up of 1 year, including the Patient and Observer Scar Assessment Scale satisfaction scores. Results: 10 patients with 16 keloids were eligible (mean follow-up, 37 months). Only one recurrence was reported (6%). In 12.5% of cases, mild erythema appeared in the early postoperative period. No major complications were observed. The overall patient and observer satisfaction rate was excellent. Conclusion: Surgical excision combined with immediate PORT is an effective and easy treatment with good esthetic results and an acceptable recurrence rate. It should be considered for patients with persistent keloid formation after failure of other treatments and those at high risk of relapse.
Collapse
Affiliation(s)
- Ilias G Petrou
- Division of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | | | - Eva Meia Rüegg
- Division of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Thomas Zilli
- Division of Radiation Oncology, Geneva University Hospitals, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Ali Modarressi
- Division of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, University of Geneva Faculty of Medicine, Geneva, Switzerland
| | - Brigitte Pittet-Cuénod
- Division of Plastic, Reconstructive and Aesthetic Surgery, Geneva University Hospitals, University of Geneva Faculty of Medicine, Geneva, Switzerland
| |
Collapse
|
48
|
Extensive CD34-to-CD90 Fibroblast Transition Defines Regions of Cutaneous Reparative, Hypertrophic, and Keloidal Scarring. Am J Dermatopathol 2019; 41:16-28. [PMID: 30320623 DOI: 10.1097/dad.0000000000001254] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND CD90 fibroblasts have been described arising from and replacing the homeostatic CD34 network in scleroderma, but have not been specifically examined in other forms of cutaneous fibrosis. OBJECTIVES To address expression, timelines, and spatial relationships of CD90, CD34, and smooth muscle actin (SMA) expressing fibroblasts in scars and to examine for the presence of a CD34-to-CD90 transition. METHODS One hundred and seventeen scars (reparative/hypertrophic/keloidal) were evaluated for CD90, CD34, and SMA expression. Double-staining immunohistochemistry for CD90/CD34 was performed to identify CD90/CD34 transitioning cells, confirmed by double-color immunofluorescence. In addition, some scars were double-stained with CD90/SMA, CD90/procollagen-1, or SMA/procollagen-1 to evaluate spatial relationships and active collagen synthesis. Expression was graded as diffuse, minority, and negative. RESULTS Most scars demonstrate a CD90/CD34 pattern, and dual CD90/CD34 fibroblasts were observed in 91% of scars. In reparative scars, CD90 expression reverses to a CD34/CD90 state with maturation. Pathologic scars exhibit prolonged CD90 expression. Both CD90 and SMA fibroblasts collagenize scars, although CD90 fibroblasts are more prevalent. CONCLUSIONS CD90 fibroblasts likely arise from the resting CD34 fibroblastic network. Actively collagenizing scar fibroblasts exhibit a CD90/CD34 phenotype, which is prolonged in pathologic scars. CD90 fibroblasts are likely important players in cutaneous scarring.
Collapse
|
49
|
TRAF4 Promotes Fibroblast Proliferation in Keloids by Destabilizing p53 via Interacting with the Deubiquitinase USP10. J Invest Dermatol 2019; 139:1925-1935.e5. [PMID: 30940456 DOI: 10.1016/j.jid.2019.03.1136] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 02/27/2019] [Accepted: 03/05/2019] [Indexed: 12/16/2022]
Abstract
Keloids represent one extreme of aberrant dermal wound healing. One of the important characteristics of keloids is uncontrolled fibroblasts proliferation. However, the mechanism of excessive proliferation of fibroblasts in keloids remains elusive. In this study, we demonstrated that TRAF4 was highly expressed in keloid fibroblasts and promoted fibroproliferation. We investigated the underlying molecular mechanism and found that TRAF4 suppressed the p53 pathway independent of its E3 ubiquitin ligase activity. Specifically, TRAF4 interacted with the deubiquitinase USP10 and blocked the access of p53 to USP10, resulting in p53 destabilization. Knockdown of p53 rescued cell proliferation in TRAF4-knockdown keloid fibroblasts, suggesting that the regulation of proliferation by TRAF4 in keloids relied on p53. Furthermore, in keloid patient samples, TRAF4 expression was inversely correlated with p53-p21 signaling activity. These findings help to elucidate the mechanisms underlying keloid development and indicate that blocking TRAF4 could represent a potential strategy for keloid therapy in the future.
Collapse
|
50
|
Huang J, Tu T, Wang W, Gao Z, Zhou G, Zhang W, Wu X, Liu W. Aligned topography mediated cell elongation reverses pathological phenotype of
in vitro
cultured keloid fibroblasts. J Biomed Mater Res A 2019; 107:1366-1378. [DOI: 10.1002/jbm.a.36650] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/17/2018] [Accepted: 02/04/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Jia Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Key Laboratory of Tissue Engineering Research, National Tissue Engineering Center of ChinaShanghai Jiao Tong University School of Medicine Shanghai People's Republic of China
| | - Tian Tu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Key Laboratory of Tissue Engineering Research, National Tissue Engineering Center of ChinaShanghai Jiao Tong University School of Medicine Shanghai People's Republic of China
| | - Wenbo Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Key Laboratory of Tissue Engineering Research, National Tissue Engineering Center of ChinaShanghai Jiao Tong University School of Medicine Shanghai People's Republic of China
| | - Zhen Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Key Laboratory of Tissue Engineering Research, National Tissue Engineering Center of ChinaShanghai Jiao Tong University School of Medicine Shanghai People's Republic of China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Key Laboratory of Tissue Engineering Research, National Tissue Engineering Center of ChinaShanghai Jiao Tong University School of Medicine Shanghai People's Republic of China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Key Laboratory of Tissue Engineering Research, National Tissue Engineering Center of ChinaShanghai Jiao Tong University School of Medicine Shanghai People's Republic of China
| | - Xiaoli Wu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Key Laboratory of Tissue Engineering Research, National Tissue Engineering Center of ChinaShanghai Jiao Tong University School of Medicine Shanghai People's Republic of China
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Key Laboratory of Tissue Engineering Research, National Tissue Engineering Center of ChinaShanghai Jiao Tong University School of Medicine Shanghai People's Republic of China
| |
Collapse
|