1
|
Guo X, Li W, Ma W, Liu Y, Liu Z, Jiao R, Yang Z, Zhang T, Wu H, Ai X, Gu X, Wang W, Zhou H, Li X, Yang C. Daidzein alleviates skin fibrosis by suppressing TGF-β1 signaling pathway via targeting PKM2. Sci Rep 2025; 15:8649. [PMID: 40082519 PMCID: PMC11906606 DOI: 10.1038/s41598-025-93007-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
Skin fibrosis including keloids, which are characterized including excessive deposition, abnormal proliferation, aggressiveness, and migration of the extracellular matrix of dermal fibroblasts. TGF-β signaling is a classical pro-fibrotic pathway, and it plays a crucial part in the occurrence and progression of skin fibrosis. Daidzein (Dai), an isoflavone compound, has been proved to possess anti-fibrosis effect by TGF-β signaling in various inflammatory and fibrotic diseases. However, little is known about Dai on skin fibrosis. Therefore, we further explored the potential effects and mechanisms of daidzein on skin fibrosis. As expected, Dai suppressed proliferation, migration and activation mouse primary dermal fibroblasts and keloid fibroblasts. Meanwhile, Dai also ameliorated bleomycin-induced skin fibrosis and reduced fibrotic markers of keloid tissues. In addition, Dai could target PKM2 to inhibit TGF-β1/Smad signaling in skin fibrosis. Overall, our research demonstrated that Dai might become a potential therapeutic candidate drug for skin fibrosis.
Collapse
Affiliation(s)
- Xiaowei Guo
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300353, China
| | - Wenqi Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300353, China
| | - Wei Ma
- Department of Burn and Plastic Surgery, Tianjin Fourth Hospital, Tianjin, 300222, China
| | - Yuming Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300353, China
| | - Zhigang Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300353, China
| | - Ran Jiao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300353, China
| | - Zhongyi Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300353, China
| | - Tiantian Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300353, China
| | - Hongliang Wu
- Department of Anesthesiology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, China
| | - Xiaoyu Ai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300353, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China
| | - Xiaoting Gu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300353, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China
| | - Wendi Wang
- Department of Plastic and Burn Surgery, Tianjin First Central Hospital, No. 24 Kangfu Road, Nankai District, Tianjin, 300192, China
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300353, China.
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China.
| | - Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300353, China.
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China.
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, 300353, China.
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, China.
| |
Collapse
|
2
|
Liu M, Liu X, Zhang J, Liang S, Gong Y, Shi S, Yuan X. Single-cell RNA sequencing reveals the heterogeneity of myofibroblasts in wound repair. Genomics 2025; 117:110982. [PMID: 39706310 DOI: 10.1016/j.ygeno.2024.110982] [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: 03/20/2024] [Revised: 12/09/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Skin wound repair involves myofibroblasts crucial for tissue integrity. This study utilized single-cell RNA sequencing to explore myofibroblast diversity in various wound healing scenarios. Analysis of 89,148 cells from skin ulcers, keloids, and normal scars identified 13 cell clusters. Myofibroblast subcluster analysis unveiled 11 subsets, with subclusters 1 and 9 predominant in ulcers. Subcluster 1 exhibited heightened matrix metalloproteinase expression and involvement in bacterial response and angiogenesis, crucial in inflammation. Tissue validation confirmed subcluster 1 significance., while animal models supported upregulated CA12, TDO2, and IL-7R in chronic ulcers. These findings illuminate myofibroblast heterogeneity and their impact on wound healing, offering insights into potential therapeutic targets.
Collapse
Affiliation(s)
- Miaonan Liu
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxuan Liu
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jingchi Zhang
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shaocong Liang
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Gong
- Department of Burns and Wound Repairing, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shengjun Shi
- Department of Burns and Wound Repairing, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Xiaopeng Yuan
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Department of Laboratory Medicine, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University; Shenzhen 518020, Guangdong China..
| |
Collapse
|
3
|
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
|
4
|
George RE, Bay CC, Thornton SM, Knazze JT, Kane NC, Ludwig KA, Donnelly DT, Poore SO, Dingle AM. Can Electrical Stimulation Prevent Recurrence of Keloid Scars? A Scoping Review. Adv Wound Care (New Rochelle) 2024. [PMID: 38888004 DOI: 10.1089/wound.2023.0203] [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: 06/20/2024] Open
Abstract
Objective: Keloids represent a symptomatic, aberrant healing process that is difficult to treat with high recurrence rates spanning from 55% to 100% if treated via excision without adjuvant therapy. Electrical stimulation (ES) has demonstrated findings that suggest it could reduce the recurrence rate of keloids after resection. Therefore, the aim of this study is to conduct a scoping review to investigate ES as an adjuvant therapy for decreasing keloid recurrence after excision. Approach: A scoping review was performed using PubMed and Web of Science databases. The search strategy encompassed terms linking keloids and various aspects of electrical stimulation. Results: Our search yielded 2,229 articles, of which 115 articles were analyzed as full text and 1 article met inclusion criteria. Despite this, ES has demonstrated other evidence that suggests its utility. ES has been shown to counter keloidic features by reducing mast cell counts, shifting wound composition from M2 to M1 macrophages, promoting angiogenesis, and controlling fibroblast orientation and location. An alternating current will orient fibroblasts perpendicular to the current without unintended migration. Innovation: Our study indicates that, based on a compilation of clinical and preclinical in vitro data, the optimal scenario for ES in the role of keloid treatment is after excision with a biphasic pulsed application and square waveform. Conclusions: ES could serve as a multifaceted, adjuvant treatment after keloid excision, steering the healing process away from keloid-associated characteristics. Its cost-effectiveness means it could be adopted globally, providing a strategy to mitigate the burden of keloids irrespective of other available treatments or economic conditions.
Collapse
Affiliation(s)
- Robert E George
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Caroline C Bay
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Sarah M Thornton
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jessieka T Knazze
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Nicole C Kane
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kip A Ludwig
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Institute for Translational Neuroengineering, Madison, Wisconsin, USA
| | - D'Andrea T Donnelly
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Samuel O Poore
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Aaron M Dingle
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
5
|
Zhang J, Zhang X, Guo X, Li W, Zhang T, Chai D, Liu Y, Chen L, Ai X, Zhou T, Wei W, Gu X, Li X, Zhou H. Remdesivir alleviates skin fibrosis by suppressing TGF-β1 signaling pathway. PLoS One 2024; 19:e0305927. [PMID: 39024326 PMCID: PMC11257276 DOI: 10.1371/journal.pone.0305927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/05/2024] [Indexed: 07/20/2024] Open
Abstract
Fibrotic skin diseases, such as keloids, are pathological results of aberrant tissue healing and are characterized by overgrowth of dermal fibroblasts. Remdesivir (RD), an antiviral drug, has been reported to have pharmacological activities in a wide range of fibrotic diseases. However, whether RD function on skin fibrosis remains unclear. Therefore, in our study, we explored the potential effect and mechanisms of RD on skin fibrosis both in vivo and in vitro. As expected, the results demonstrated that RD alleviated BLM-induced skin fibrosis and attenuates the gross weight of keloid tissues in vivo. Further studies suggested that RD suppressed fibroblast activation and autophagy both in vivo and in vitro. In addition, mechanistic research showed that RD attenuated fibroblasts activation by the TGF-β1/Smad signaling pathway and inhibited fibroblasts autophagy by the PI3K/Akt/mTOR signaling pathway. In summary, our results demonstrate therapeutic potential of RD for skin fibrosis in the future.
Collapse
Affiliation(s)
- Jianwei Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Xiujun Zhang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Xiaowei Guo
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Wenqi Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Tiantian Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Dan Chai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Yuming Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Li Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Xiaoyu Ai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, China
| | - Tianyuan Zhou
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Wenguo Wei
- Department of Dermatology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Xiaoting Gu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| |
Collapse
|
6
|
Kwon SH, Lee J, Yoo J, Jung Y. Artificial keloid skin models: understanding the pathophysiological mechanisms and application in therapeutic studies. Biomater Sci 2024; 12:3321-3334. [PMID: 38812375 DOI: 10.1039/d4bm00005f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Keloid is a type of scar formed by the overexpression of extracellular matrix substances from fibroblasts following inflammation after trauma. The existing keloid treatment methods include drug injection, surgical intervention, light exposure, cryotherapy, etc. However, these methods have limitations such as recurrence, low treatment efficacy, and side effects. Consequently, studies are being conducted on the treatment of keloids from the perspective of inflammatory mechanisms. In this study, keloid models are created to understand inflammatory mechanisms and explore treatment methods to address them. While previous studies have used animal models with gene mutations, chemical treatments, and keloid tissue transplantation, there are limitations in fully reproducing the characteristics of keloids unique to humans, and ethical issues related to animal welfare pose additional challenges. Consequently, studies are underway to create in vitro artificial skin models to simulate keloid disease and apply them to the development of treatments for skin diseases. In particular, herein, scaffold technologies that implement three-dimensional (3D) full-thickness keloid models are introduced to enhance mechanical properties as well as biological properties of tissues, such as cell proliferation, differentiation, and cellular interactions. It is anticipated that applying these technologies to the production of artificial skin for keloid simulation could contribute to the development of inflammatory keloid treatment techniques in the future.
Collapse
Affiliation(s)
- Soo Hyun Kwon
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.
| | - Jongmin Lee
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Republic of Korea
| | - Jin Yoo
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.
| | - Youngmee Jung
- Center for Biomaterials, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.
- School of Electrical and Electronic Engineering, YU-KIST Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
7
|
Chalwa T, Lebeko M, Matobole R, P Khumalo N, Bayat A. Enhanced bioenergetic cellular activity with metabolic switch to aerobic glycolysis in Keloid and Folliculitis Keloidalis Nuchae. Arch Dermatol Res 2024; 316:412. [PMID: 38878082 PMCID: PMC11180017 DOI: 10.1007/s00403-024-03038-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/02/2024] [Accepted: 04/26/2024] [Indexed: 06/19/2024]
Abstract
Keloid scars and folliculitis keloidalis nuchae (FKN) are benign fibroproliferative dermal lesions of unknown aetiology and ill-defined treatment, which typically present in genetically susceptible individuals. Their pathognomonic hallmarks include local aggressive invasive behaviour plus high recurrence post-therapy. In view of this, we investigated proliferative and key parameters of bioenergetic cellular characteristics of site-specific keloid-derived fibroblasts (intra(centre)- and peri(margin)-lesional) and FKN compared to normal skin and normal flat non-hypertrophic scar fibroblasts as negative controls.The results showed statistically significant (P < 0.01) and variable growth dynamics with increased proliferation and migration in keloid fibroblasts, while FKN fibroblasts showed a significant (P < 0.001) increase in proliferation but similar migration profile to controls. A statistically significant metabolic switch towards aerobic glycolysis in the fibroblasts from the disease conditions was noted. Furthermore, an increase in basal glycolysis with a concomitant increase in the cellular maximum glycolytic capacity was also demonstrated in perilesional keloid and FKN fibroblasts (P < 0.05). Mitochondrial function parameters showed increased oxidative phosphorylation in the disease conditions (P < 0.05) indicating functional mitochondria. These findings further suggest that Keloids and FKN demonstrate a switch to a metabolic phenotype of aerobic glycolysis. Increased glycolytic flux inhibition is a potential mechanistic basis for future therapy.
Collapse
Affiliation(s)
- Temwani Chalwa
- MRC-SA Wound Healing and Keloid Research Unit, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Maribanyana Lebeko
- MRC-SA Wound Healing and Keloid Research Unit, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Relebohile Matobole
- MRC-SA Wound Healing and Keloid Research Unit, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Nonhlanhla P Khumalo
- MRC-SA Wound Healing and Keloid Research Unit, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Ardeshir Bayat
- MRC-SA Wound Healing and Keloid Research Unit, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
8
|
Oh S, Yeo E, Shim J, Noh H, Park J, Lee KT, Kim SH, Lee D, Lee JH. Revealing the pathogenesis of keloids based on the status: Active vs inactive. Exp Dermatol 2024; 33:e15088. [PMID: 38685820 DOI: 10.1111/exd.15088] [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: 10/10/2023] [Revised: 04/05/2024] [Accepted: 04/14/2024] [Indexed: 05/02/2024]
Abstract
Recently, the pathomechanisms of keloids have been extensively researched using transcriptomic analysis, but most studies did not consider the activity of keloids. We aimed to profile the transcriptomics of keloids according to their clinical activity and location within the keloid lesion, compared with normal and mature scars. Tissue samples were collected (keloid based on its activity (active and inactive), mature scar from keloid patients and normal scar (NS) from non-keloid patients). To reduce possible bias, all keloids assessed in this study had no treatment history and their location was limited to the upper chest or back. Multiomics assessment was performed by using single-cell RNA sequencing and multiplex immunofluorescence. Increased mesenchymal fibroblasts (FBs) was the main feature in keloid patients. Noticeably, the proportion of pro-inflammatory FBs was significantly increased in active keloids compared to inactive ones. To explore the nature of proinflammatory FBs, trajectory analysis was conducted and CCN family associated with mechanical stretch exhibited higher expression in active keloids. For vascular endothelial cells (VECs), the proportion of tip and immature cells increased in keloids compared to NS, especially at the periphery of active keloids. Also, keloid VECs highly expressed genes with characteristics of mesenchymal activation compared to NS, especially those from the active keloid center. Multiomics analysis demonstrated the distinct expression profile of active keloids. Clinically, these findings may provide the future appropriate directions for development of treatment modalities of keloids. Prevention of keloids could be possible by the suppression of mesenchymal activation between FBs and VECs and modulation of proinflammatory FBs may be the key to the control of active keloids.
Collapse
Affiliation(s)
- Sejin Oh
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eunhye Yeo
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Medical Device Management and Research, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Joonho Shim
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyungrye Noh
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jihye Park
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyeong-Tae Lee
- Department of Plastic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seok-Hyung Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dongyoun Lee
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong Hee Lee
- Department of Dermatology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Medical Device Management and Research, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
| |
Collapse
|
9
|
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
|
10
|
Kojima S, Koyama S, Ka M, Saito Y, Parrish EH, Endo M, Takata S, Mizukoshi M, Hikino K, Takeda A, Gelinas AF, Heaton SM, Koide R, Kamada AJ, Noguchi M, Hamada M, Kamatani Y, Murakawa Y, Ishigaki K, Nakamura Y, Ito K, Terao C, Momozawa Y, Parrish NF. Mobile element variation contributes to population-specific genome diversification, gene regulation and disease risk. Nat Genet 2023:10.1038/s41588-023-01390-2. [PMID: 37169872 DOI: 10.1038/s41588-023-01390-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 04/04/2023] [Indexed: 05/13/2023]
Abstract
Mobile genetic elements (MEs) are heritable mutagens that recursively generate structural variants (SVs). ME variants (MEVs) are difficult to genotype and integrate in statistical genetics, obscuring their impact on genome diversification and traits. We developed a tool that accurately genotypes MEVs using short-read whole-genome sequencing (WGS) and applied it to global human populations. We find unexpected population-specific MEV differences, including an Alu insertion distribution distinguishing Japanese from other populations. Integrating MEVs with expression quantitative trait loci (eQTL) maps shows that MEV classes regulate tissue-specific gene expression by shared mechanisms, including creating or attenuating enhancers and recruiting post-transcriptional regulators, supporting class-wide interpretability. MEVs more often associate with gene expression changes than SNVs, thus plausibly impacting traits. Performing genome-wide association study (GWAS) with MEVs pinpoints potential causes of disease risk, including a LINE-1 insertion associated with keloid and fasciitis. This work implicates MEVs as drivers of human divergence and disease risk.
Collapse
Affiliation(s)
- Shohei Kojima
- Genome Immunobiology RIKEN Hakubi Research Team, RIKEN Center for Integrative Medical Sciences and RIKEN Cluster for Pioneering Research, Yokohama, Japan.
| | - Satoshi Koyama
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Mirei Ka
- Genome Immunobiology RIKEN Hakubi Research Team, RIKEN Center for Integrative Medical Sciences and RIKEN Cluster for Pioneering Research, Yokohama, Japan
- Next-Generation Precision Medicine Development, Integrative Genomics Laboratory, Graduate School of Medicine, Department of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuka Saito
- Genome Immunobiology RIKEN Hakubi Research Team, RIKEN Center for Integrative Medical Sciences and RIKEN Cluster for Pioneering Research, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Erica H Parrish
- Genome Immunobiology RIKEN Hakubi Research Team, RIKEN Center for Integrative Medical Sciences and RIKEN Cluster for Pioneering Research, Yokohama, Japan
| | - Mikiko Endo
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Sadaaki Takata
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Misaki Mizukoshi
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Keiko Hikino
- Laboratory for Pharmacogenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Atsushi Takeda
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
- Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Asami F Gelinas
- Genome Immunobiology RIKEN Hakubi Research Team, RIKEN Center for Integrative Medical Sciences and RIKEN Cluster for Pioneering Research, Yokohama, Japan
| | - Steven M Heaton
- Genome Immunobiology RIKEN Hakubi Research Team, RIKEN Center for Integrative Medical Sciences and RIKEN Cluster for Pioneering Research, Yokohama, Japan
| | - Rie Koide
- Genome Immunobiology RIKEN Hakubi Research Team, RIKEN Center for Integrative Medical Sciences and RIKEN Cluster for Pioneering Research, Yokohama, Japan
| | - Anselmo J Kamada
- Genome Immunobiology RIKEN Hakubi Research Team, RIKEN Center for Integrative Medical Sciences and RIKEN Cluster for Pioneering Research, Yokohama, Japan
- Paleovirology Lab, Department of Biology, University of Oxford, Oxford, UK
| | - Michiya Noguchi
- Cell Engineering Division, BioResource Research Center, RIKEN, Tsukuba, Japan
| | - Michiaki Hamada
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
- Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Yoichiro Kamatani
- Laboratory of Complex Trait Genomics, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yasuhiro Murakawa
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- IFOM ETS - the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Kazuyoshi Ishigaki
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yukio Nakamura
- Cell Engineering Division, BioResource Research Center, RIKEN, Tsukuba, Japan
| | - Kaoru Ito
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- The Department of Applied Genetics, The School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Nicholas F Parrish
- Genome Immunobiology RIKEN Hakubi Research Team, RIKEN Center for Integrative Medical Sciences and RIKEN Cluster for Pioneering Research, Yokohama, Japan.
| |
Collapse
|
11
|
Meng J, Wang G, Zhou L, Jiang S, Qian S, Chen L, Wang C, Jiang R, Yang C, Niu B, Liu Y, Ding Z, Zhuo S, Liu Z. Mapping variation of extracellular matrix in human keloid scar by label-free multiphoton imaging and machine learning. JOURNAL OF BIOMEDICAL OPTICS 2023; 28:045001. [PMID: 37038546 PMCID: PMC10082605 DOI: 10.1117/1.jbo.28.4.045001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/26/2023] [Indexed: 05/18/2023]
Abstract
Significance Rapid diagnosis and analysis of human keloid scar tissues in an automated manner are essential for understanding pathogenesis and formulating treatment solutions. Aim Our aim is to resolve the features of the extracellular matrix in human keloid scar tissues automatically for accurate diagnosis with the aid of machine learning. Approach Multiphoton microscopy was utilized to acquire images of collagen and elastin fibers. Morphological features, histogram, and gray-level co-occurrence matrix-based texture features were obtained to produce a total of 28 features. The minimum redundancy maximum relevancy feature selection approach was implemented to rank these features and establish feature subsets, each of which was employed to build a machine learning model through the tree-based pipeline optimization tool (TPOT). Results The feature importance ranking was obtained, and 28 feature subsets were acquired by incremental feature selection. The subset with the top 23 features was identified as the most accurate. Then stochastic gradient descent classifier optimized by the TPOT was generated with an accuracy of 96.15% in classifying normal, scar, and adjacent tissues. The area under curve of the classification results (scar versus normal and adjacent, normal versus scar and adjacent, and adjacent versus normal and scar) was 1.0, 1.0, and 0.99, respectively. Conclusions The proposed approach has great potential for future dermatological clinical diagnosis and analysis and holds promise for the development of computer-aided systems to assist dermatologists in diagnosis and treatment.
Collapse
Affiliation(s)
- Jia Meng
- Zhejiang University, College of Optical Science and Engineering, International Research Center for Advanced Photonics, State Key Laboratory of Modern Optical Instrumentation, Hangzhou, China
| | - Guangxing Wang
- Xiamen University, School of Public Health, Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen, China
| | - Lingxi Zhou
- Zhejiang University, College of Optical Science and Engineering, International Research Center for Advanced Photonics, State Key Laboratory of Modern Optical Instrumentation, Hangzhou, China
| | - Shenyi Jiang
- Zhejiang University, College of Optical Science and Engineering, International Research Center for Advanced Photonics, State Key Laboratory of Modern Optical Instrumentation, Hangzhou, China
| | - Shuhao Qian
- Zhejiang University, College of Optical Science and Engineering, International Research Center for Advanced Photonics, State Key Laboratory of Modern Optical Instrumentation, Hangzhou, China
| | - Lingmei Chen
- Zhejiang University, College of Optical Science and Engineering, International Research Center for Advanced Photonics, State Key Laboratory of Modern Optical Instrumentation, Hangzhou, China
| | - Chuncheng Wang
- Zhejiang University, College of Optical Science and Engineering, International Research Center for Advanced Photonics, State Key Laboratory of Modern Optical Instrumentation, Hangzhou, China
| | - Rushan Jiang
- Zhejiang University, College of Optical Science and Engineering, International Research Center for Advanced Photonics, State Key Laboratory of Modern Optical Instrumentation, Hangzhou, China
| | - Chen Yang
- Zhejiang University, College of Optical Science and Engineering, International Research Center for Advanced Photonics, State Key Laboratory of Modern Optical Instrumentation, Hangzhou, China
| | - Bo Niu
- Zhejiang University, College of Optical Science and Engineering, International Research Center for Advanced Photonics, State Key Laboratory of Modern Optical Instrumentation, Hangzhou, China
| | - Yijie Liu
- Zhejiang University, College of Optical Science and Engineering, International Research Center for Advanced Photonics, State Key Laboratory of Modern Optical Instrumentation, Hangzhou, China
| | - Zhihua Ding
- Zhejiang University, College of Optical Science and Engineering, International Research Center for Advanced Photonics, State Key Laboratory of Modern Optical Instrumentation, Hangzhou, China
| | - Shuangmu Zhuo
- Jimei University, School of Science, Xiamen, China
- Address all correspondence to Zhiyi Liu, ; Shuangmu Zhuo,
| | - Zhiyi Liu
- Zhejiang University, College of Optical Science and Engineering, International Research Center for Advanced Photonics, State Key Laboratory of Modern Optical Instrumentation, Hangzhou, China
- Zhejiang University, Jiaxing Research Institute, Intelligent Optics and Photonics Research Center, Jiaxing, China
- Address all correspondence to Zhiyi Liu, ; Shuangmu Zhuo,
| |
Collapse
|
12
|
Neves LMG, Wilgus TA, Bayat A. In Vitro, Ex Vivo, and In Vivo Approaches for Investigation of Skin Scarring: Human and Animal Models. Adv Wound Care (New Rochelle) 2023; 12:97-116. [PMID: 34915768 DOI: 10.1089/wound.2021.0139] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Significance: The cutaneous repair process naturally results in different types of scarring that are classified as normal or pathological. Affected individuals are often affected from an esthetic, physical (functional), and psychosocial perspective. The distinct nature of scarring in humans, particularly the formation of pathological scars, makes the study of skin scarring a challenge for researchers in this area. Several established experimental models exist for studying scar formation. However, the increasing development and validation of newly emerging models have made it possible to carry out studies focused on different variables that influence this unique process. Recent Advances: Experimental models such as in vitro, ex vivo, and in vivo models have obtained different degrees of success in the reproduction of the scar formation in its native milieu and true environment. These models also differ in their ability to elucidate the molecular, cellular, and structural mechanisms involved in scarring, as well as for testing new agents and approaches for therapies. The models reviewed here, including cells derived from human skin and in vivo animal models, have contributed to the advancement of skin scarring research. Critical Issues and Future Directions: The absence of experimental models that faithfully reproduce the typical characteristics of the different types of human skin scars makes the improvement of validated models and the establishment of new ones a critical unmet need. The fields of wound healing research combined with tissue engineering have offered newer alternatives for experimental studies with the potential to provide clinically useful knowledge about scar formation.
Collapse
Affiliation(s)
- Lia M G Neves
- Plastic & Reconstructive Surgery Research, Centre for Dermatology Research, Wound Healing Theme, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, England, United Kingdom
| | - Traci A Wilgus
- Department of Pathology, Ohio State University, Columbus, Ohio, USA
| | - Ardeshir Bayat
- Plastic & Reconstructive Surgery Research, Centre for Dermatology Research, Wound Healing Theme, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, England, United Kingdom.,Medical Research Council (MRC) Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
13
|
Ozdemir K, Citaker S, Diker S, Keser I, Kurukahvecioglu O, Uyar Gocun P, Gulbahar O. Does Venipuncture Increase Lymphedema by Triggering Inflammation or Infection? An Experimental Rabbit Ear Lymphedema Model Study. Lymphat Res Biol 2023; 21:34-41. [PMID: 35687408 DOI: 10.1089/lrb.2021.0095] [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: 11/13/2022] Open
Abstract
Background: Recent guidelines recommend avoiding venipuncture to prevent lymphedema for breast cancer patients. This study investigated whether single or multiple sterile venipuncture procedures develop a systemic inflammation or infection and increase lymphedema in the rabbit ear lymphedema model. Methods and Results: Eighteen New Zealand white female rabbits were included. The right ear lymphedema model was created by surgical procedure; then, rabbits were divided into three randomized groups. Single and multiple venipuncture procedures were applied at least the 60th day after surgery for Group I and II, respectively. Group III was a control group. C-reactive protein (CRP) and procalcitonin (PCT) levels were analyzed to determine inflammation and infection. Ear thickness measurements were applied using a vernier caliper to assess the differences in lymphedema between the ears. All rabbits were euthanized on the 90th day after surgery. Histopathological analysis was performed to evaluate lymphedema by measuring tissue thicknesses. Ear thickness measurements showed that ear lymphedema was developed and maintained with surgical operation in all groups (p < 0.05). There was no difference in the ear thickness measurements between and within-groups results (p > 0.05). CRP and PCT levels were below the lower detection levels in all groups. According to the differences of histopathological ear distances, there were significant differences within-groups for all groups (p < 0.05), and no differences were identified between groups (p > 0.05). Conclusion: This experimental study demonstrated that single or multiple sterile venipuncture procedures did not trigger infection or inflammation and did not exacerbate ear lymphedema in the rabbit ear lymphedema model.
Collapse
Affiliation(s)
- Kadirhan Ozdemir
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Izmir Bakircay University, Izmir, Turkey
| | - Seyit Citaker
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Gazi University, Ankara, Turkey
| | - Seyda Diker
- Laboratory Animal Breeding and Experimental Research Center, Gazi University, Ankara, Turkey
| | - Ilke Keser
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Gazi University, Ankara, Turkey
| | | | - Pinar Uyar Gocun
- Department of Medical Pathology, and Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Ozlem Gulbahar
- Department of Medical Biochemistry, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
14
|
LncRNA GNAS-AS1 knockdown inhibits keloid cells growth by mediating the miR-188-5p/RUNX2 axis. Mol Cell Biochem 2022; 478:707-719. [PMID: 36036334 DOI: 10.1007/s11010-022-04538-6] [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: 06/15/2021] [Accepted: 08/08/2022] [Indexed: 10/15/2022]
Abstract
Keloid is a common dermis tumor, occurring repeatedly, affecting the quality of patients' life. Long non-coding RNAs (lncRNAs) have crucial regulatory capacities in skin scarring formation and subsequent scar carcinogenesis. The intention of this study was to investigate the mechanism and function of GNAS antisense-1 (GNAS-AS1) in keloids. Clinical samples were collected to evaluate the expression of GNAS-AS1, RUNX2, and miR-188-5p by qRT-PCR. The proliferation, migration, and invasion of HKF cells were detected by CCK-8, wound healing, and Transwell assays. The expression levels of mRNA and protein were examined through qRT-PCR and Western blot assay. Luciferase reporter assay was used to identify the binding relationship among GNAS-AS1, miR-188-5p, and Runt-related transcription factor 2 (RUNX2). GNAS-AS1 and RUNX2 expressions were remarkably enhanced, and miR-188-5p expression was decreased in keloid clinical tissues and HKF cells. GNAS-AS1 overexpression promoted cells proliferation, migration, and invasion, while GNAS-AS1 knockdown had the opposite trend. Furthermore, overexpression of GNAS-AS1 reversed the inhibitory effect of 5-FU on cell proliferation, migration, and invasion. MiR-188-5p inhibition or RUNX2 overexpression could enhance the proliferation, migration, and invasion of HKF cells. GNAS-AS1 targeted miR-188-5p to regulate RUNX2 expression. In addition, the inhibition effects of GNAS-AS1 knockdown on HKF cells could be reversed by inhibition of miR-188-5p or overexpression of RUNX2, while RUNX2 overexpression eliminated the suppressive efficaciousness of miR-188-5p mimics on HKF cells growth. GNAS-AS1 knockdown could regulate the miR-188-5p/RUNX2 signaling axis to inhibit the growth and migration in keloid cells. It is suggested that GNAS-AS1 may become a new target for the prevention and treatment of keloid.
Collapse
|
15
|
Gao Y, Hou X, Dai Y, Yang T, Chen K. Radiation-induced FAP + fibroblasts are involved in keloid recurrence after radiotherapy. Front Cell Dev Biol 2022; 10:957363. [PMID: 36092734 PMCID: PMC9449371 DOI: 10.3389/fcell.2022.957363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/04/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Keloid scars (KSs), which are composed of abnormal hyperplastic scar tissue, form during skin wound healing due to excessive fibroblast activation and collagen secretion. Although surgical resection and radiation therapy are used to prevent recurrence, KS recurrence rates range from 15 to 23%, and the underlying mechanism is unclear. Methods: To elucidate the mechanism of keloid recurrence, we established a PDX model and the grafts remained for over 20 weeks after transplantation on the bilateral backs of the NCG mice. Results: RNA-seq revealed that KS tissue gene expression was highly consistent before and after transplantation. Then, one side of the KS graft was irradiated with electron beam therapy (10 Gy), significant increases in vimentin and fibroblast activation protein alpha (FAP) expression were observed after irradiation and were accompanied by severe microvascular destruction. Surprisingly, 4 weeks after irradiation, significantly increased recurrence was observed with increased FAP + tissue and cell cycle regulator expression, resulting in a remarkable altered graft volume. Moreover, irradiation-induced FAP upregulation markedly facilitated radiation resistance and increased cell cycle progression, decreased senescence, and increased energy production. Conclusion: Our findings revealed that irradiation causes increased abundance of FAP + cells, which was associated with cell proliferation and delayed cellular senescence, accompanied by ATP production.
Collapse
Affiliation(s)
- Yan Gao
- Department of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
| | - Xue Hou
- Department of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
| | - Yuyin Dai
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, China
| | - Ting Yang
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
| | - Kexin Chen
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Kexin Chen,
| |
Collapse
|
16
|
Hsu JF, Yu RP, Stanton EW, Wang J, Wong AK. Current Advancements in Animal Models of Postsurgical Lymphedema: A Systematic Review. Adv Wound Care (New Rochelle) 2022; 11:399-418. [PMID: 34128396 PMCID: PMC9142133 DOI: 10.1089/wound.2021.0033] [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] [Indexed: 01/29/2023] Open
Abstract
Significance: Secondary lymphedema is a debilitating disease caused by lymphatic dysfunction characterized by chronic swelling, dysregulated inflammation, disfigurement, and compromised wound healing. Since there is no effective cure, animal model systems that support basic science research into the mechanisms of secondary lymphedema are critical to advancing the field. Recent Advances: Over the last decade, lymphatic research has led to the improvement of existing animal lymphedema models and the establishment of new models. Although an ideal model does not exist, it is important to consider the strengths and limitations of currently available options. In a systematic review adhering to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we present recent developments in the field of animal lymphedema models and provide a concise comparison of ease, cost, reliability, and clinical translatability. Critical Issues: The incidence of secondary lymphedema is increasing, and there is no gold standard of treatment or cure for secondary lymphedema. Future Directions: As we iterate and create animal models that more closely characterize human lymphedema, we can achieve a deeper understanding of the pathophysiology and potentially develop effective therapeutics for patients.
Collapse
Affiliation(s)
- Jerry F. Hsu
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, California, USA.,Division of Plastic Surgery, City of Hope National Medical Center, Duarte, California, USA.,Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Roy P. Yu
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Eloise W. Stanton
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Jin Wang
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, California, USA.,Division of Plastic Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Alex K. Wong
- Division of Plastic and Reconstructive Surgery, Keck School of Medicine of USC, Los Angeles, California, USA.,Division of Plastic Surgery, City of Hope National Medical Center, Duarte, California, USA.,Correspondence: Division of Plastic Surgery, City of Hope National Medical Center, 1500 E. Duarte Road, Pavillion 2216, Duarte, CA 91010, USA.
| |
Collapse
|
17
|
Xu H, Zhu Z, Hu J, Sun J, Wo Y, Wang X, Zou H, Li B, Zhang Y. Downregulated cytotoxic CD8 + T-cell identifies with the NKG2A-soluble HLA-E axis as a predictive biomarker and potential therapeutic target in keloids. Cell Mol Immunol 2022; 19:527-539. [PMID: 35039632 PMCID: PMC8975835 DOI: 10.1038/s41423-021-00834-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/23/2021] [Indexed: 11/09/2022] Open
Abstract
Keloids are an abnormal fibroproliferative wound-healing disease with a poorly understood pathogenesis, making it difficult to predict and prevent this disease in clinical settings. Identifying disease-specific signatures at the molecular and cellular levels in both the blood circulation and primary lesions is urgently needed to develop novel biomarkers for risk assessment and therapeutic targets for recurrence-free treatment. There is mounting evidence of immune cell dysregulation in keloid scarring. In this study, we aimed to profile keloid scar tissues and blood cells and found that downregulation of cytotoxic CD8+ T cells is a keloid signature in the peripheral blood and keloid lesions. Single-cell RNA sequencing revealed that the NKG2A/CD94 complex was specifically upregulated, which might contribute to the significant reduction in CTLs within the scar tissue boundary. In addition, the NKG2A/CD94 complex was associated with high serum levels of soluble human leukocyte antigen-E (sHLA-E). We subsequently measured sHLA-E in our hospital-based study cohort, consisting of 104 keloid patients, 512 healthy donors, and 100 patients with an interfering disease. The sensitivity and specificity of sHLA-E were 83.69% (87/104) and 92.16% (564/612), respectively, and hypertrophic scars and other unrelated diseases exhibited minimal interference with the test results. Furthermore, intralesional therapy with triamcinolone combined with 5-fluorouracil drastically decreased the sHLA-E levels in keloid patients with better prognostic outcomes, while an incomplete reduction in the sHLA-E levels in patient serum was associated with higher recurrence. sHLA-E may effectively serve as a diagnostic marker for assessing the risk of keloid formation and a prognostic marker for the clinical outcomes of intralesional treatment.
Collapse
Affiliation(s)
- Heng Xu
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhu Zhu
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Hu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jiawei Sun
- Genekinder Medicaltech (Shanghai) Co., Ltd, Shanghai, China
| | - Yan Wo
- Department of Anatomy and Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xianshu Wang
- Creative Biosciences (Guangzhou) Co., Ltd., Guangzhou, Guangdong, China
| | - Hongzhi Zou
- Creative Biosciences (Guangzhou) Co., Ltd., Guangzhou, Guangdong, China
- Department of Colorectal Surgery, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangdong, China
| | - Bin Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yixin Zhang
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
18
|
Katarzyna A, Agnieszka G, Paweł A, Magdalena KD, Agnieszka SW, Małgorzata D, Agnieszka L, Dominika WD. Analysis of the expression of FAP-α protein in 2D-keloid fibroblast cultures and in 3D models of keloid. J Cosmet Dermatol 2021; 21:3561-3566. [PMID: 34897935 DOI: 10.1111/jocd.14667] [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: 11/02/2021] [Accepted: 11/22/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Keloids arise most often as a result of abnormal wound healing. The lesion characterizes inflammation and fibroproliferation. Fibroblast activation protein alpha (FAP-α) is one of enzymes engaged in keloid formation. It is serine protease that facilitates cells invasion and growth. FAP-α possess also non-enzymatic activity and plays role in activation of cell signaling. The aim of the study was to assess the impact of culture conditions on the proliferation of keloid fibroblasts and the expression of FAP-α. Analysis of utility of 3D models of keloid in in vitro study of pathogenesis of keloids was also made. METHODS NHDF and KEL FIB cells were cultured in vitro in 2D cultures and 3D Matrigel models. The viability of cells was assayed spectrophotometrically with WST-1 test. FAP-α protein amount in cell cultures and 3D models was evaluated with the use of ELISA test. RESULTS KEL FIB fibroblasts exhibited higher viability than NHDF fibroblasts in all three models of keloid. The expression of FAP-α is different in normal and keloid fibroblasts. In vitro conditions influence the expression of FAP-α in NHDF cells but not in KEL FIB cells. CONCLUSIONS This preliminary study has shown that the expression of FAP-α, similarly to other enzymes engaged in keloid formation, is different in keloids in vivo and in in vitro models. FAP-α expression is modulated by in vitro conditions in normal fibroblasts but not in keloid fibroblasts.
Collapse
Affiliation(s)
- Adamczyk Katarzyna
- Department of Cosmetology, Chair of Cosmetology, School of Pharmaceutical Science, Medical University of Silesia, Katowice, Poland
| | - Garncarczyk Agnieszka
- Department of Cosmetology, Chair of Cosmetology, School of Pharmaceutical Science, Medical University of Silesia, Katowice, Poland
| | - Antończak Paweł
- Department of Cosmetology, Chair of Cosmetology, School of Pharmaceutical Science, Medical University of Silesia, Katowice, Poland
| | - Kimsa-Dudek Magdalena
- Department of Nutrigenomics and Bromatology, Chair of Molecular Biology, School of Pharmaceutical Science, Medical University of Silesia, Katowice, Poland
| | - Synowiec-Wojtarowicz Agnieszka
- Department of Nutrigenomics and Bromatology, Chair of Molecular Biology, School of Pharmaceutical Science, Medical University of Silesia, Katowice, Poland
| | - Derewniuk Małgorzata
- Department of Nutrigenomics and Bromatology, Chair of Molecular Biology, School of Pharmaceutical Science, Medical University of Silesia, Katowice, Poland
| | - Lubczyńska Agnieszka
- Department of Cosmetology, Chair of Cosmetology, School of Pharmaceutical Science, Medical University of Silesia, Katowice, Poland
| | - Wcisło-Dziadecka Dominika
- Department of Cosmetology, Chair of Cosmetology, School of Pharmaceutical Science, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
19
|
Pinocembrin Ameliorates Skin Fibrosis via Inhibiting TGF-β1 Signaling Pathway. Biomolecules 2021; 11:biom11081240. [PMID: 34439906 PMCID: PMC8393190 DOI: 10.3390/biom11081240] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 01/06/2023] Open
Abstract
Skin fibrotic diseases, such as keloids, are mainly caused by pathologic scarring of wounds during healing and characterized by benign cutaneous overgrowths of dermal fibroblasts. Current surgical and therapeutic modalities of skin fibrosis are unsatisfactory. Pinocembrin, a natural flavonoid, has been shown to possess a vast range of pharmacological activities including antimicrobial, antioxidant, anti-inflammatory, and anti-tumor activities. In this study we explored the potential effect and mechanisms of pinocembrin on skin fibrosis in vitro and in vivo. In vitro studies indicated that pinocembrin dose-dependently suppressed proliferation, migration, and invasion of keloid fibroblasts and mouse primary dermal fibroblasts. The in vivo studies showed that pinocembrin could effectively alleviate bleomycin (BLM)-induced skin fibrosis and reduce the gross weight and fibrosis-related protein expression of keloid tissues in xenograft mice. Further mechanism studies indicated that pinocembrin could suppress TGF-β1/Smad signaling and attenuate TGF-β1-induced activation of skin fibroblasts. In conclusion, our results demonstrate the therapeutic potential of pinocembrin for skin fibrosis.
Collapse
|
20
|
Memariani H, Memariani M, Moravvej H, Shahidi-Dadras M. Emerging and Novel Therapies for Keloids: A compendious review. Sultan Qaboos Univ Med J 2021; 21:e22-e33. [PMID: 33777420 PMCID: PMC7968901 DOI: 10.18295/squmj.2021.21.01.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/27/2020] [Accepted: 08/13/2020] [Indexed: 12/31/2022] Open
Abstract
Keloids are abnormal fibroproliferative scars with aggressive dermal growth expanding beyond the borders of the original injury. Different therapeutic modalities, such as corticosteroids, surgical excision, topical silicone gel sheeting, laser therapy, cryotherapy, photodynamic therapy and radiotherapy, have been used to treat keloids; however, none of these modalities has proven completely effective. Recently, researchers have devised several promising anti-keloid therapies including anti-hypertensive pharmaceuticals, calcineurin inhibitors, electrical stimulation, mesenchymal stem cell therapy, microneedle physical contact and ribonucleic acid-based therapies. The present review summarises emerging and novel treatments for keloids. PubMed® (National Library of Medicine, Bethesda, Maryland, USA), EMBASE (Elsevier, Amsterdam, Netherlands) and Web of Science (Clarivate Analytics, Philadelphia, Pennsylvania, USA) were searched for relevant literature published between January 1987 to June 2020. A total of 118 articles were included in this review. A deeper understanding of the molecular mechanisms underlying keloid scarring pathogenesis would open further avenues for developing innovative treatments.
Collapse
Affiliation(s)
- Hamed Memariani
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojtaba Memariani
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamideh Moravvej
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
21
|
Nagar H, Kim S, Lee I, Kim S, Choi SJ, Piao S, Jeon BH, Oh SH, Kim CS. Downregulation of CR6-interacting factor 1 suppresses keloid fibroblast growth via the TGF-β/Smad signaling pathway. Sci Rep 2021; 11:500. [PMID: 33436666 PMCID: PMC7804403 DOI: 10.1038/s41598-020-79785-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 12/01/2020] [Indexed: 12/29/2022] Open
Abstract
Keloids are a type of aberrant skin scarring characterized by excessive accumulation of collagen and extracellular matrix (ECM), arising from uncontrolled wound healing responses. While typically non-pathogenic, keloids are occasionally regarded as a form of benign tumor. CR6-interacting factor 1 (CRIF1) is a well-known CR6/GADD45-interacting protein, that has both nuclear and mitochondrial functions, and also exerts regulatory effects on cell growth and apoptosis. In this study, cell proliferation, cell migration, collagen production and TGF-β signaling was compared between normal fibroblasts (NFs) and keloid fibroblasts (KFs). Subsequently, the effects of CRIF1 deficiency were investigated in both NFs and KFs. Cell proliferation, cell migration, collagen production and protein expressions of TGF-β, phosphorylation of Smad2 and Smad3 were all found to be higher in KFs compared to NFs. CRIF1 deficiency in NFs and KFs inhibited cell proliferation, migration, and collagen production. In addition, phosphorylation of Smad2 and Smad3, which are transcription factors of collagen, was decreased. In contrast, mRNA expression levels of Smad7 and SMURF2, two important inhibitory proteins of Smad2/3, were increased, suggesting that CRIF1 may regulate collagen production. CRIF1 deficiency decreases the proliferation and migration of KFs, thereby inhibiting their overgrowth via the transforming growth factor-β (TGF-β)/Smad pathway. CRIF1 may therefore represent a potential therapeutic target in keloid pathogenesis.
Collapse
Affiliation(s)
- Harsha Nagar
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Physiology, School of Medicine, Chungnam National University, 55 Munhwa-ro, Jung-Gu, Daejeon, 301-131, Republic of Korea
| | - Sungmin Kim
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Physiology, School of Medicine, Chungnam National University, 55 Munhwa-ro, Jung-Gu, Daejeon, 301-131, Republic of Korea.,Department of BK21 Plus CNU Integrative Biomedical Education Initiative, Chungnam National University, Daejeon, Republic of Korea
| | - Ikjun Lee
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Physiology, School of Medicine, Chungnam National University, 55 Munhwa-ro, Jung-Gu, Daejeon, 301-131, Republic of Korea
| | - Seonhee Kim
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Physiology, School of Medicine, Chungnam National University, 55 Munhwa-ro, Jung-Gu, Daejeon, 301-131, Republic of Korea.,Department of BK21 Plus CNU Integrative Biomedical Education Initiative, Chungnam National University, Daejeon, Republic of Korea
| | - Su-Jeong Choi
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Physiology, School of Medicine, Chungnam National University, 55 Munhwa-ro, Jung-Gu, Daejeon, 301-131, Republic of Korea
| | - Shuyu Piao
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Physiology, School of Medicine, Chungnam National University, 55 Munhwa-ro, Jung-Gu, Daejeon, 301-131, Republic of Korea
| | - Byeong Hwa Jeon
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea.,Department of Physiology, School of Medicine, Chungnam National University, 55 Munhwa-ro, Jung-Gu, Daejeon, 301-131, Republic of Korea
| | - Sang-Ha Oh
- Department of Plastic and Reconstructive Surgery, School of Medicine, Chungnam National University, 282 Munhwa-ro, Jung-Gu, Daejeon, 35015, Republic of Korea. .,Brain Research Institute, School of Medicine, Chungnam National University, Daejeon, Republic of Korea.
| | - Cuk-Seong Kim
- Department of Medical Science, Chungnam National University, Daejeon, Republic of Korea. .,Department of Physiology, School of Medicine, Chungnam National University, 55 Munhwa-ro, Jung-Gu, Daejeon, 301-131, Republic of Korea. .,Department of BK21 Plus CNU Integrative Biomedical Education Initiative, Chungnam National University, Daejeon, Republic of Korea.
| |
Collapse
|
22
|
Tang Z, Ding J, Zhai X, Jing M, Guan Z, Li Y. MicroRNA-21 may be involved in the therapeutic effects of Galla chinensis ointment on keloid. J Int Med Res 2020; 48:300060520909602. [PMID: 32216491 PMCID: PMC7133421 DOI: 10.1177/0300060520909602] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Objective Galla chinensis ointment can inhibit the proliferation of keloid fibroblasts and decrease keloid formation. We investigated whether Galla chinensis ointment inhibits keloid fibroblast proliferation through expression of microRNA-21, phosphorylated (p)-phosphatidylinositol 3-kinase (p-PI3K), chromosome 10 neutropenic protein phosphatase (PTEN), protein kinase B (p-Akt), and mammalian target of rapamycin (p-mTOR). Methods A keloid mouse model and human keloid-derived fibroblasts were developed and treated with Galla chinensis. Immunohistochemistry, western blot, and reverse transcription-PCR were used to detect miR-21, PI3K, PTEN, Akt, and mTOR in keloid tissues. Results p-Akt and p-mTOR were highly expressed in the control group, PTEN was highly expressed in the treatment group, and p-PI3K was highly expressed in keloid tissue in both groups. Galla chinensis reduced miR-21 expression and increased PTEN mRNA expression in keloid fibroblasts compared with the control group, resulting in increased PTEN protein and decreased p-Akt and p-mTOR protein. Galla chinensis had no effect on p-PI3K. Conclusion Galla chinensis might inhibit proliferation of keloid fibroblasts by upregulating PTEN, thus inhibiting expression of miR-21 and downregulating p-Akt and p-mTOR expression. These results confirm the effect of Galla chinensis ointment on fibroblasts and suggest that it could be used to manage keloids clinically.
Collapse
Affiliation(s)
- Zhiming Tang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Dermatology, Xuzhou Hospital affiliated with Nanjing University of Traditional Chinese Medicine, Xuzhou, China
| | - Jicun Ding
- Department of Burns and Plastic Surgery, Xuzhou Central Hospital, Xuzhou, China
| | - Xiaoxiang Zhai
- Department of Dermatology, Shanghai Seventh People’s Hospital, Shanghai, China
- Xiaoxiang Zhai, Department of Dermatology, Shanghai Seventh People’s Hospital, No. 358 Datong Road, Gaoqiao, Pudong New Area, Shanghai 200137, China.
| | - Mengqing Jing
- Department of Dermatology, Xuzhou Hospital affiliated with Nanjing University of Traditional Chinese Medicine, Xuzhou, China
| | - Zhiqiang Guan
- Department of Dermatology, Xuzhou First People’s Hospital, Xuzhou, China
| | - Yongcong Li
- Department of Dermatology, Xuzhou Hospital affiliated with Nanjing University of Traditional Chinese Medicine, Xuzhou, China
| |
Collapse
|
23
|
Macarak EJ, Wermuth PJ, Rosenbloom J, Uitto J. Keloid disorder: Fibroblast differentiation and gene expression profile in fibrotic skin diseases. Exp Dermatol 2020; 30:132-145. [PMID: 33211348 DOI: 10.1111/exd.14243] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/10/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023]
Abstract
Keloid disorder, a group of fibroproliferative skin diseases, is characterized by unremitting accumulation of the extracellular matrix (ECM) of connective tissue, primarily collagen, to develop cutaneous tumors on the predilection sites of skin. There is a strong genetic predisposition for keloid formation, and individuals of African and Asian ancestry are particularly prone. The principal cell type responsible for ECM accumulation is the myofibroblast derived from quiescent resident skin fibroblasts either through trans-differentiation or from keloid progenitor stem cells with capacity for multi-lineage differentiation and self-renewal. The biosynthetic pathways leading to ECM accumulation are activated by several cytokines, but particularly by TGF-β signalling. The mechanical properties of the cellular microenvironment also play a critical role in the cell's response to TGF-β, as demonstrated by culturing of fibroblasts derived from keloids and control skin on substrata with different degrees of stiffness. These studies also demonstrated that culturing of fibroblasts on tissue culture plastic in vitro does not reflect their biosynthetic capacity in vivo. Collectively, our current understanding of the pathogenesis of keloids suggests a complex network of interacting cellular, molecular and mechanical factors, with distinct pathways leading to myofibroblast differentiation and activation. Keloids can serve as a model system of fibrotic diseases, a group of currently intractable disorders, and deciphering of the critical pathogenetic steps leading to ECM accumulation is expected to identify targets for pharmacologic intervention, not only for keloids but also for a number of other, both genetic and acquired, fibrotic diseases.
Collapse
Affiliation(s)
- Edward J Macarak
- The Joan and Joel Rosenbloom Center for Fibrotic Diseases, and the Jefferson Institute of Molecular Medicine, Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Peter J Wermuth
- The Joan and Joel Rosenbloom Center for Fibrotic Diseases, and the Jefferson Institute of Molecular Medicine, Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Joel Rosenbloom
- The Joan and Joel Rosenbloom Center for Fibrotic Diseases, and the Jefferson Institute of Molecular Medicine, Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Jouni Uitto
- The Joan and Joel Rosenbloom Center for Fibrotic Diseases, and the Jefferson Institute of Molecular Medicine, Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
24
|
Kumar AS, Kamalasanan K. Drug delivery to optimize angiogenesis imbalance in keloid: A review. J Control Release 2020; 329:1066-1076. [PMID: 33091533 DOI: 10.1016/j.jconrel.2020.10.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/16/2020] [Accepted: 10/17/2020] [Indexed: 12/12/2022]
Abstract
The wound healing process involves three continuous stages. Where, any imbalance can lead to the formation of unwanted keloids, hypertrophic scar, or tumors. Keloids are any unpleasant, non-compliant comorbidity affecting a major section of people around the globe who acquire it either genetically or by pathological means as a result of a skin injury. Angiogenesis is unavoidable in the healing process after an injury or disruption of skin to promote tissue regeneration. Uncontrolled angiogenesis during the healing process can initiate the unwanted response in the wound that facilitate keloid. Angiogenic therapy is adapted to accelerate healing after an injury. Else ways, there exists a risk of keloid formation due to excessive angiogenesis during the wound healing process. There are numerous strategies to treat keloid. Anti-angiogenic factors are provided to patients post-surgery to prevent the keloid formation; however, they come into the picture after the formation of keloid. The available strategies to treat keloids are steroidal injections, surgical excision of the keloid, radiotherapy, pressure therapy, the use of cryosurgery, and many more. The available treatments are not promising in reducing the recurrent rate of keloids as there are chances of high re-occurrences with similar/larger lesions on the removed keloid site. In this review, we are discussing the importance of controlled angiogenesis with the help of controlled drug delivery strategies enabling the wound healing process without the induction of keloid.
Collapse
Affiliation(s)
- Aishwari S Kumar
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, AIMS Ponekkara PO, Kochi, Kerala, 682041, India
| | - Kaladhar Kamalasanan
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, AIMS Ponekkara PO, Kochi, Kerala, 682041, India.
| |
Collapse
|
25
|
Li Q, Qin Z, Chen B, An Y, Nie F, Yang X, Pan B, Bi H. Mitochondrial Dysfunction and Morphological Abnormality in Keloid Fibroblasts. Adv Wound Care (New Rochelle) 2020. [DOI: 10.1089/wound.2019.0988] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Qi Li
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, P.R. China
| | - Zelian Qin
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, P.R. China
| | - Bin Chen
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, P.R. China
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, P.R. China
| | - Fangfei Nie
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, P.R. China
| | - Xin Yang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, P.R. China
| | - Bailin Pan
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, P.R. China
| | - Hongsen Bi
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, P.R. China
| |
Collapse
|
26
|
Stegemann A, Böhm M. Targeting the α7 nicotinic acetylcholine receptor-A novel road towards the future treatment of skin diseases. Exp Dermatol 2020; 29:924-931. [PMID: 32780438 DOI: 10.1111/exd.14173] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/11/2022]
Abstract
Nicotinic acetylcholine receptors (nAChRs) are members of the superfamily of neurotransmitter-gated ion channels. The natural ligand for nAChRs is the endogenous neurotransmitter acetylcholine. Among the nAChRs is the α7nAChR. It is not only expressed by neural tissues but also in the skin. A number of different resident cutaneous cell types including epidermal keratinocytes, sebocytes and dermal fibroblasts express functional α7nAChR. Moreover, cells of the immune system such as lymphocytes, macrophages and monocytes, playing an important role in skin homeostasis, also express α7nAChR. Translational research focusing on the exploitation of the α7nAChR in dermatology has revealed that this neuroendocrine receptor could be promising target for the treatment of inflammatory skin diseases. For example, α7nAChR agonists can counteract transforming growth factor-β1-mediated responses in dermal fibroblasts, key effector cells in scleroderma. In accordance with this α7nAChR, agonists are effective in both inflammation and non-inflammation-driven models of experimentally induced skin fibrosis. Moreover, α7nAChR agonists can modulate expression of proinflammatory cytokines in epidermal keratinocytes that are crucially involved in the pathogenesis of psoriasis and other inflammatory skin diseases. Finally, the capability of α7nAChR agonists to suppress ultraviolet light A/B-induced responses, for example production of proinflammatory cytokines and oxidative stress, the latter crucially involved in dermal photoageing, points to a potential of such agents in the prevention of extrinsic skin ageing. Therefore, emphasis on translational research targeting the α7nAChR in skin may lead to the development of new treatment and prevention modalities against fibrosclerotic skin diseases, psoriasis vulgaris, atopic dermatitis, acne, photodermatoses and extrinsic skin ageing.
Collapse
Affiliation(s)
| | - Markus Böhm
- Dept. of Dermatology, University of Münster, Germany
| |
Collapse
|
27
|
Ekstein SF, Wyles SP, Moran SL, Meves A. Keloids: a review of therapeutic management. Int J Dermatol 2020; 60:661-671. [PMID: 32905614 DOI: 10.1111/ijd.15159] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/20/2022]
Abstract
Keloid scar formation arises from a disorganized fibroproliferative collagen response that extends beyond the original wound margins because of excessive production of extracellular matrix (ECM). Despite treatment options for keloid scars including medical and surgical therapies, such as intralesional steroid injection and surgical excision, the recurrence rate remains high. Herein we consolidate recently published narrative reviews, systematic reviews, and meta-analyses to provide an overview of updated treatment recommendations for keloidal scar formation. PubMed search engine was used to access the MEDLINE database to investigate updates regarding keloid incidence and treatment. More than 100 articles were reviewed. Keloid management remains a multimodal approach. There continues to be no gold standard of treatment that provides a consistently low recurrence rate; however, the increasing number of available treatments and synergistic combinations of these treatments (i.e., laser-based devices in combination with intralesional steroids, or 5-fluorouracil (5-FU) in combination with steroid therapy) is showing favorable results. Future studies could target the efficacy of novel treatment modalities (i.e., autologous fat grafting or stem cell-based therapies) for keloid management. This review article provides updated treatment guidelines for keloids and discusses insight into management to assist patient-focused, evidence-based clinical decision making.
Collapse
Affiliation(s)
- Samuel F Ekstein
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Steven L Moran
- Division of Plastic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
28
|
Li Z, Li S, Li K, Jiang X, Zhang J, Liu H. A highly simulated scar model developed by grafting human thin split-thickness skin on back of nude mouse: The remodeling process, histological characteristics of scars. Biochem Biophys Res Commun 2020; 526:744-750. [PMID: 32265030 DOI: 10.1016/j.bbrc.2020.03.140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/17/2020] [Indexed: 12/17/2022]
Abstract
A predictive scar animal model is needed in order to study the mechanism and assess the therapies before its use in humans. However, due to the differences in wound healing patterns and regeneration ability, none of the existing models can fully simulate the characteristics of human scar. The aim of this study was to build a model that recapitulated the developing process and outcomes of human hypertrophic scar (HS). Nude mice were grafted with thin split-thickness human skins. The dynamic changes and final outcomes of the grafts were investigated. The results showed that human skin grafts survived and underwent progressive scarring remodeling in morphology and histology. Scar related markers (α-SMA, CD34, Collage I, TGF-β1) were positive in immunohistology. Protein expressions in TGF-β1/Smad2/3 pathway were increased in accordance with HS during the development process by western blotting. It was finally proved that scar reconstructed by this model matches a real-world human HS. This is a stable, easy to reproduce model for studying the scar formation process and its properties.
Collapse
Affiliation(s)
- Zehua Li
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China; Innovative Technology Research Institute of Tissue Repair and Regeneration, Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong, China
| | - Shenghong Li
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Kecheng Li
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiao Jiang
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jinrong Zhang
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China; Innovative Technology Research Institute of Tissue Repair and Regeneration, Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong, China
| | - Hongwei Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
29
|
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: 193] [Impact Index Per Article: 38.6] [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
|
30
|
Wang Z, Huang X, Zan T, Li Q, Li H. A modified scar model with controlled tension on secondary wound healing in mice. BURNS & TRAUMA 2020; 8:tkaa013. [PMID: 32395565 PMCID: PMC7201370 DOI: 10.1093/burnst/tkaa013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/19/2019] [Indexed: 11/19/2022]
Abstract
Pathological scars might cause a distorted appearance and restricted mobility, and the study of scar pathophysiology has been hindered by the absence of a reliable model. In this study, we introduce a model with a modified device to induce controlled tension on a wound healing by secondary intention to overcome the shortcomings of the model generated by Aarabi et al. We investigated and recommend an induction of 0.1 N/mm2 tension on day 7 for 14 days to mimic the characteristics of human scars. A 3.5-fold increase in scar tissue and a 2-fold increase in collagen production were induced by the modified model. Histologically, the modified method increased scar thickness. However, no significant difference was found in cell density between the two groups. This modified procedure significantly increased scar tissue, which could be used for further cellular and biomolecular research. The mechanical force applied to the wound became measurable and controllable. This method is more convenient for researchers to observe in real-time and for providing timely adjustments of the tension used in this modified model.
Collapse
Affiliation(s)
- Zi Wang
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Medical School of Shanghai Jiao Tong University, 639 Zhizaoju Road, Shanghai 200011, China
| | - Xin Huang
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Medical School of Shanghai Jiao Tong University, 639 Zhizaoju Road, Shanghai 200011, China
| | - Tao Zan
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Medical School of Shanghai Jiao Tong University, 639 Zhizaoju Road, Shanghai 200011, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Medical School of Shanghai Jiao Tong University, 639 Zhizaoju Road, Shanghai 200011, China
| | - Haizhou Li
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Medical School of Shanghai Jiao Tong University, 639 Zhizaoju Road, Shanghai 200011, China
| |
Collapse
|
31
|
Darmawan CC, Montenegro SE, Jo G, Kusumaningrum N, Lee SH, Chung JH, Mun JH. Adiponectin-Based Peptide (ADP355) Inhibits Transforming Growth Factor-β1-Induced Fibrosis in Keloids. Int J Mol Sci 2020; 21:E2833. [PMID: 32325772 PMCID: PMC7215791 DOI: 10.3390/ijms21082833] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/08/2020] [Accepted: 04/16/2020] [Indexed: 12/30/2022] Open
Abstract
Keloids, benign cutaneous overgrowths of dermal fibroblasts, are caused by pathologic scarring of wounds during healing. Current surgical and therapeutic modalities are unsatisfactory. Although adiponectin has shown an antifibrotic effect, its large size and insolubility limit its potential use in keloid treatment. We investigated the effect of a smaller and more stable adiponectin-based peptide (ADP355) on transforming growth factor β1 (TGF-β1)-induced fibrosis in a primary culture of keloid fibroblasts prepared from clinically obtained keloid samples. Xenograft of keloid tissues on athymic nude mice was used to investigate the effect of intralesional injection of ADP355. ADP355 significantly attenuated the TGF-β1-induced expression of procollagen type 1 in keloid fibroblasts (p < 0.05). Moreover, it inhibited the TGF-β1-induced phosphorylation of SMAD3 and ERK, while amplifying the phosphorylation of AMP-activated protein kinase (p < 0.05). Knockdown of adiponectin receptor 1 reversed the attenuation of procollagen expression in ADP355-treated TGF-β1-induced fibrosis (p < 0.05). ADP355 also significantly reduced the gross weight and procollagen expression of keloid tissues in xenograft mice compared to control animals. These results demonstrate the therapeutic potential of the adiponectin peptide ADP355 for keloids.
Collapse
Affiliation(s)
- Claudia C Darmawan
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Korea
| | - Sara E Montenegro
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Korea
| | - Gwanghyun Jo
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Korea
| | - Novi Kusumaningrum
- Department of Dermatology and Venereology, Diponegoro University, Semarang 50275, Indonesia
| | - Si-Hyung Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Korea
| | - Jin-Ho Chung
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Korea
| | - Je-Ho Mun
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Korea
| |
Collapse
|
32
|
Uitto J, Tirgan MH. Clinical Challenge and Call for Research on Keloid Disorder: Meeting Report from The 3rd International Keloid Research Foundation Symposium, Beijing 2019. J Invest Dermatol 2020; 140:515-518. [DOI: 10.1016/j.jid.2019.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/04/2019] [Accepted: 10/07/2019] [Indexed: 12/22/2022]
|
33
|
Sharma JR, Lebeko M, Kidzeru EB, Khumalo NP, Bayat A. In Vitro and Ex Vivo Models for Functional Testing of Therapeutic Anti-scarring Drug Targets in Keloids. Adv Wound Care (New Rochelle) 2019; 8:655-670. [PMID: 31827980 PMCID: PMC6904937 DOI: 10.1089/wound.2019.1040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 09/09/2019] [Indexed: 01/08/2023] Open
Abstract
Significance: Keloids are benign fibro-proliferative raised dermal lesions that spread beyond the original borders of the wound, continue to grow, rarely regress, and are the most common in pigmented individuals after an abnormal wound healing response. The current treatment failure and respective challenges involved highlighting the underlying issue that the etiopathogenesis of keloids is still not well understood. Disease models are required to better understand the disease pathogenesis. It is not possible to establish keloids in animals because of the uniqueness of this disease to human skin. To address this challenge, along these lines, non-animal reproducible models are vital in investigating molecular mechanisms of keloid pathogenesis and therapeutics development. Recent Advances: Various non-animal models have been developed to better understand the molecular mechanisms involved in keloid scarring and aid in identifying and evaluating the therapeutic potential of novel drug candidates. In this scenario, the current review aims at describing in vitro monocultures, co-cultures, organotypic cultures, and ex vivo whole skin keloid tissue organ culture models. Critical Issues and Future Directions: Current treatment options for keloids are far from securing a cure or preventing disease recurrence. Identifying universally accepted effective therapy for keloids has been hampered by the absence of appropriate disease model systems. Animal models do not accurately mimic the disease, thus non-animal model systems are pivotal in keloid research. The use of these models is essential not only for a better understanding of disease biology but also for identifying and evaluating novel drug targets.
Collapse
Affiliation(s)
- Jyoti R. Sharma
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Maribanyana Lebeko
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Elvis B. Kidzeru
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
- Centre for Medical Research, Institute of Medical Research and Medical Plant Studies, Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
| | - Nonhlanhla P. Khumalo
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| | - Ardeshir Bayat
- Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Faculty of Health Sciences and Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
- Plastic and Reconstructive Surgery Research, Division of Musculoskeletal & Dermatological Sciences, NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
34
|
Padmanabhan J, Maan ZN, Kwon SH, Kosaraju R, Bonham CA, Gurtner GC. In Vivo Models for the Study of Fibrosis. Adv Wound Care (New Rochelle) 2019; 8:645-654. [PMID: 31827979 PMCID: PMC6904938 DOI: 10.1089/wound.2018.0909] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/28/2019] [Indexed: 02/04/2023] Open
Abstract
Significance: Fibrosis and scar formation pose a substantial physiological and psychological burden on patients and a significant public health burden on the economy, estimated to be up to $12 billion a year. Fibrosis research is heavily reliant on in vivo models, but variations in animal models and differences between animal and human fibrosis necessitates careful selection of animal models to study fibrosis. There is also an increased need for improved animal models that recapitulate human pathophysiology. Recent Advances: Several murine and porcine models, including xenograft, drug-induced fibrosis, and mechanical load-induced fibrosis, for different types of fibrotic disease have been described in the literature. Recent findings have underscored the importance of mechanical forces in the pathophysiology of scarring. Critical Issues: Differences in skin, properties of subcutaneous tissue, and modes of fibrotic healing in animal models and humans provide challenges toward investigating fibrosis with in vivo models. While porcine models are typically better suited to study cutaneous fibrosis, murine models are preferred because of the ease of handling and availability of transgenic strains. Future Directions: There is a critical need to develop novel murine models that recapitulate the mechanical cues influencing fibrosis in humans, significantly increasing the translational value of fibrosis research. We advocate a translational pipeline that begins in mouse models with modified biomechanical environments for foundational molecular and cellular research before validation in porcine models that closely mimic the human condition.
Collapse
Affiliation(s)
- Jagannath Padmanabhan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Zeshaan N. Maan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Sun Hyung Kwon
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Revanth Kosaraju
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Clark A. Bonham
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Geoffrey C. Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
35
|
Barallobre-Barreiro J, Woods E, Bell RE, Easton JA, Hobbs C, Eager M, Baig F, Ross AM, Mallipeddi R, Powell B, Soldin M, Mayr M, Shaw TJ. Cartilage-like composition of keloid scar extracellular matrix suggests fibroblast mis-differentiation in disease. Matrix Biol Plus 2019; 4:100016. [PMID: 33543013 PMCID: PMC7852214 DOI: 10.1016/j.mbplus.2019.100016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/16/2019] [Accepted: 09/16/2019] [Indexed: 02/08/2023] Open
Abstract
Following wound damage to the skin, the scarring spectrum is wide-ranging, from a manageable normal scar through to pathological keloids. The question remains whether these fibrotic lesions represent simply a quantitative extreme, or alternatively, whether they are qualitatively distinct. A three-way comparison of the extracellular matrix (ECM) composition of normal skin, normal scar and keloids was performed using quantitative discovery-based proteomics. This approach identified 40 proteins that were significantly altered in keloids compared to normal scars, and strikingly, 23 keloid-unique proteins. The major alterations in keloids, when functionally grouped, showed many changes in proteins involved in ECM assembly and fibrillogenesis, but also a keloid-associated loss of proteases, and a unique cartilage-like composition, which was also evident histologically. The presence of Aggrecan and Collagen II in keloids suggest greater plasticity and mis-differentiation of the constituent cells. This study characterises the ECM of both scar types to a depth previously underappreciated. This thorough molecular description of keloid lesions relative to normal scars is an essential step towards our understanding of this debilitating clinical problem, and how best to treat it.
Collapse
Affiliation(s)
- Javier Barallobre-Barreiro
- King's College London, James Black Centre British Heart Foundation Centre, Denmark Hill Campus, London SE5 9NU, UK
| | - Elizabeth Woods
- Division of Biomedical Sciences, St George's University of London, London SW17 0RE, UK
| | - Rachel E. Bell
- King's College London, School of Immunology & Microbial Sciences, Department of Inflammation Biology, Centre for Inflammation Biology & Cancer Immunology, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Jennifer A. Easton
- King's College London, School of Immunology & Microbial Sciences, Department of Inflammation Biology, Centre for Inflammation Biology & Cancer Immunology, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Carl Hobbs
- King's College London, Wolfson Centre for Age Related Diseases, Guy's Campus, London SE1 1UL, UK
| | - Michael Eager
- Division of Biomedical Sciences, St George's University of London, London SW17 0RE, UK
| | - Ferheen Baig
- King's College London, James Black Centre British Heart Foundation Centre, Denmark Hill Campus, London SE5 9NU, UK
| | - Alastair Mackenzie Ross
- Guy's and St Thomas' NHS Foundation Trust, Department of Plastic Surgery, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Raj Mallipeddi
- Guy's and St Thomas' NHS Foundation Trust, St John's Institute of Dermatology, Cancer Centre, Great Maze Pond, London SE1 9RT, UK
| | - Barry Powell
- St George's University Hospitals NHS Trust, Department of Plastic and Reconstructive Surgery, Blackshaw Road, London SW17 0QT, UK
| | - Mark Soldin
- St George's University Hospitals NHS Trust, Department of Plastic and Reconstructive Surgery, Blackshaw Road, London SW17 0QT, UK
| | - Manuel Mayr
- King's College London, James Black Centre British Heart Foundation Centre, Denmark Hill Campus, London SE5 9NU, UK
| | - Tanya J. Shaw
- King's College London, School of Immunology & Microbial Sciences, Department of Inflammation Biology, Centre for Inflammation Biology & Cancer Immunology, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
36
|
Zheng B, Fang QQ, Wang XF, Shi BH, Zhao WY, Chen CY, Zhang MX, Zhang LY, Hu YY, Shi P, Ma L, Tan WQ. The effect of topical ramipril and losartan cream in inhibiting scar formation. Biomed Pharmacother 2019; 118:109394. [DOI: 10.1016/j.biopha.2019.109394] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/10/2019] [Accepted: 08/23/2019] [Indexed: 01/15/2023] Open
|
37
|
Fernández Peñuela R, Casaní Arazo L, Masiá Ayala J. Outcomes in Vascularized Lymph Node Transplantation in Rabbits: A Reliable Model for Improving the Surgical Approach to Lymphedema. Lymphat Res Biol 2019; 17:413-417. [DOI: 10.1089/lrb.2018.0038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Randolfo Fernández Peñuela
- Department of Plastic Surgery, Santa Creu i Sant Pau Hospital. (Universitat Autónoma de Barcelona), Barcelona, Spain
| | - Laura Casaní Arazo
- Catalan Institute of Cardiovascular Sciences, Santa Creu i Sant Pau Hospital, Barcelona, Spain
| | - Jaume Masiá Ayala
- Department of Plastic Surgery, Santa Creu i Sant Pau Hospital. (Universitat Autónoma de Barcelona), Barcelona, Spain
| |
Collapse
|
38
|
Boahene K, Brissett AE, Jones LR. Facial Plastic Surgery Controversies: Keloids. Facial Plast Surg Clin North Am 2018; 26:105-112. [PMID: 29636144 DOI: 10.1016/j.fsc.2017.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
There are more than 11 million people in the world affected with keloids. Nevertheless, there is a lack of agreement in keloid management. Moreover, keloid research has left gaps in the understanding of its pathogenesis. Six questions are answered by 3 clinical scientists in an attempt to address common keloid controversies.
Collapse
Affiliation(s)
- Kofi Boahene
- Facial Plastic and Reconstructive Surgery, Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institute, 601 North Caroline Street, Baltimore, MD 21287, USA
| | - Anthony E Brissett
- Division of Facial Plastic and Reconstructive Surgery, Institute of Academic Medicine, Houston Methodist ENT and Facial Plastic Surgery Associates, Houston Methodist Hospital, 6550 Fannin Street, Suite 1703, Huston, TX 77030, USA
| | - Lamont R Jones
- Department of Otolaryngology-Head and Neck Surgery, Henry Ford Health Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA.
| |
Collapse
|
39
|
Mak KK, Tan JJ, Marappan P, Balijepalli MK, Choudhury H, Ramamurthy S, Pichika MR. Galangin’s potential as a functional food ingredient. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.04.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
40
|
Fernández Peñuela R, Pons Playa G, Casaní Arazo L, Masiá Ayala J. An Experimental Lymphedema Animal Model for Assessing the Results of Lymphovenous Anastomosis. Lymphat Res Biol 2018; 16:234-239. [DOI: 10.1089/lrb.2016.0068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Randolfo Fernández Peñuela
- Department of Plastic Surgery, Hospital Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gemma Pons Playa
- Department of Plastic Surgery, Hospital Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Laura Casaní Arazo
- Catalan Institute of Cardiovascular Sciences, Hospital Santa Creu i Sant Pau, Pavilion of the Convent, Barcelona, Spain
| | - Jaume Masiá Ayala
- Department of Plastic Surgery, Hospital Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
41
|
Schaefer L. Decoding fibrosis: Mechanisms and translational aspects. Matrix Biol 2018; 68-69:1-7. [PMID: 29679639 DOI: 10.1016/j.matbio.2018.04.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 12/17/2022]
Abstract
Fibrosis, a complex process of abnormal tissue healing which inevitably leads to loss of physiological organ structure and function, is a worldwide leading cause of death. Despite a large body of research over the last two decades, antifibrotic approaches are mainly limited to organ replacement therapy generating high costs of medical care. In this translational issue, a unique group of basic and clinical researchers provide meaningful answers to a desperate call of society for effective antifibrotic treatments. Fortunately, a plethora of novel fibrogenic factors and biomarkers has been identified. Noninvasive diagnostic methods and drug delivery systems have been recently developed for the management of fibrosis. Consequently, a large number of exciting clinical trials addressing comprehensive, organ and stage-specific mechanisms of fibrogenesis are ongoing. By critically addressing previously unsuccessful and novel promising therapeutic strategies, we aim to spread hope for future treatments of the various forms of organ fibrosis.
Collapse
Affiliation(s)
- Liliana Schaefer
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main 60590, Germany.
| |
Collapse
|
42
|
Abstract
Recent advances on preclinical model based on patient-derived tumor xenografts have new insight into many clinical fields. According to our literature review, many authors believe that immunodeficient animals such as athymic rats and mice should be used to prevent tissue loss caused by acute rejection to establish patient-derived tumor xenografts models.However, recent advances showed that the microenvironment has gained attention as an important factor responsible for disease progression. Additionally, researchers attempt to come up with novel findings in chemotherapy drugs and immune modulator to control development of keloid. For these reasons, establishment of reliable animal model of keloids is very important.In this new model using an immunocompetent animal as a humanized-xenografts model, human keloid scar has been maintained for as long as 4 months. Results of migration assay have demonstrated that typical morphology of keloid fibroblast was preserved based on multiple time point observations despite its aging change. Quantitative real time polymerase chain reaction findings suggested that after implantation, there has been significant increase of vascular endothelial growth factor, CD34, and transforming growth factor beta 1 expression despite insignificant changes of hypoxia inducible factor 1 an matrix metallopeptidase 1, and matrix metallopeptidase 9 gene expression. These findings suggested that implantation of keloids within the immunocompetent animals yields is very useful experimental model in terms of fibrosis.In summary, the authors have successfully established and propagated patient-derived keloid model using the immunocompetent animals. This model could be used to test novel materials as well as combination therapies and is superior to the conventional cell line experiment models. In addition, the biology of the keloids can easily be assessed to identify predictive markers for responses to treatment regimens that are currently actively under research in various centers.
Collapse
|
43
|
Jumper N, Hodgkinson T, Paus R, Bayat A. Site-specific gene expression profiling as a novel strategy for unravelling keloid disease pathobiology. PLoS One 2017; 12:e0172955. [PMID: 28257480 PMCID: PMC5336271 DOI: 10.1371/journal.pone.0172955] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 02/13/2017] [Indexed: 12/13/2022] Open
Abstract
Keloid disease (KD) is a fibroproliferative cutaneous tumour characterised by heterogeneity, excess collagen deposition and aggressive local invasion. Lack of a validated animal model and resistance to a multitude of current therapies has resulted in unsatisfactory clinical outcomes of KD management. In order to address KD from a new perspective, we applied for the first time a site-specific in situ microdissection and gene expression profiling approach, through combined laser capture microdissection and transcriptomic array. The aim here was to analyse the utility of this approach compared with established methods of investigation, including whole tissue biopsy and monolayer cell culture techniques. This study was designed to approach KD from a hypothesis-free and compartment-specific angle, using state-of-the-art microdissection and gene expression profiling technology. We sought to characterise expression differences between specific keloid lesional sites and elucidate potential contributions of significantly dysregulated genes to mechanisms underlying keloid pathobiology, thus informing future explorative research into KD. Here, we highlight the advantages of our in situ microdissection strategy in generating expression data with improved sensitivity and accuracy over traditional methods. This methodological approach supports an active role for the epidermis in the pathogenesis of KD through identification of genes and upstream regulators implicated in epithelial-mesenchymal transition, inflammation and immune modulation. We describe dermal expression patterns crucial to collagen deposition that are associated with TGFβ-mediated signalling, which have not previously been examined in KD. Additionally, this study supports the previously proposed presence of a cancer-like stem cell population in KD and explores the possible contribution of gene dysregulation to the resistance of KD to conventional therapy. Through this innovative in situ microdissection gene profiling approach, we provide better-defined gene signatures of distinct KD regions, thereby addressing KD heterogeneity, facilitating differential diagnosis with other cutaneous fibroses via transcriptional fingerprinting, and highlighting key areas for future KD research.
Collapse
Affiliation(s)
- N. Jumper
- Plastic and Reconstructive Surgery Research, University of Manchester, Oxford Rd, Manchester, United Kingdom
| | - T. Hodgkinson
- Plastic and Reconstructive Surgery Research, University of Manchester, Oxford Rd, Manchester, United Kingdom
- Centre for Tissue Injury and Repair, University of Manchester, and MAHSC, Manchester, United Kingdom
| | - R. Paus
- Centre for Dermatology Research, University of Manchester, and MAHSC, Manchester, United Kingdom
| | - A. Bayat
- Plastic and Reconstructive Surgery Research, University of Manchester, Oxford Rd, Manchester, United Kingdom
- Centre for Dermatology Research, University of Manchester, and MAHSC, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
44
|
Jumper N, Hodgkinson T, Arscott G, Har-Shai Y, Paus R, Bayat A. The Aldo-Keto Reductase AKR1B10 Is Up-Regulated in Keloid Epidermis, Implicating Retinoic Acid Pathway Dysregulation in the Pathogenesis of Keloid Disease. J Invest Dermatol 2016; 136:1500-1512. [PMID: 27025872 DOI: 10.1016/j.jid.2016.03.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/09/2016] [Accepted: 03/07/2016] [Indexed: 12/19/2022]
Abstract
Keloid disease is a recurrent fibroproliferative cutaneous tumor of unknown pathogenesis for which clinical management remains unsatisfactory. To obtain new insights into hitherto underappreciated aspects of keloid pathobiology, we took a laser capture microdissection-based, whole-genome microarray analysis approach to identify distinct keloid disease-associated gene expression patterns within defined keloid regions. Identification of the aldo-keto reductase enzyme AKR1B10 as highly up-regulated in keloid epidermis suggested that an imbalance of retinoic acid metabolism is likely associated with keloid disease. Here, we show that AKR1B10 transfection into normal human keratinocytes reproduced the abnormal retinoic acid pathway expression pattern we had identified in keloid epidermis. Cotransfection of AKR1B10 with a luciferase reporter plasmid showed reduced retinoic acid response element activity, supporting the hypothesis of retinoic acid synthesis deficiency in keloid epidermis. Paracrine signals released by AKR1B10-overexpressing keratinocytes into conditioned medium resulted in up-regulation of transforming growth factor-β1, transforming growth factor-β2, and collagens I and III in both keloid and normal skin fibroblasts, mimicking the typical profibrotic keloid profile. Our study results suggest that insufficient retinoic acid synthesis by keloid epidermal keratinocytes may contribute to the pathogenesis of keloid disease. We refocus attention on the role of injured epithelium in keloid disease and identify AKR1B10 as a potential new target in future management of keloid disease.
Collapse
Affiliation(s)
- Natalie Jumper
- Plastic and Reconstructive Surgery Research, Manchester Institute of Biotechnology, University of Manchester, Manchester, UK
| | - Tom Hodgkinson
- Plastic and Reconstructive Surgery Research, Manchester Institute of Biotechnology, University of Manchester, Manchester, UK
| | - Guyan Arscott
- Department of Plastic and Reconstructive Surgery, University of West Indies, Kingston, Jamaica
| | - Yaron Har-Shai
- Plastic Surgery Unit, Carmel Medical Center, Haifa, Israel
| | - Ralf Paus
- Centre for Dermatology Research, Institute of Inflammation and Repair, University of Manchester, Manchester, UK; Department of Dermatology, University of Münster, D-48149, Münster, Germany
| | - Ardeshir Bayat
- Plastic and Reconstructive Surgery Research, Manchester Institute of Biotechnology, University of Manchester, Manchester, UK; Centre for Dermatology Research, Institute of Inflammation and Repair, University of Manchester, Manchester, UK.
| |
Collapse
|
45
|
Andrews JP, Marttala J, Macarak E, Rosenbloom J, Uitto J. Keloids: The paradigm of skin fibrosis - Pathomechanisms and treatment. Matrix Biol 2016; 51:37-46. [PMID: 26844756 DOI: 10.1016/j.matbio.2016.01.013] [Citation(s) in RCA: 271] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Keloids, fibroproliferative dermal tumors with effusive accumulation of extracellular matrix (ECM) components, particularly collagen, result from excessive expression of growth factors and cytokines. The etiology of keloids is unknown but they occur after dermal injury in genetically susceptible individuals, and they cause both physical and psychological distress for the affected individuals. Several treatment methods for keloids exist, including the combination therapy of surgical excision followed by intralesional steroid therapy, however, they have high recurrence rate regardless of the current treatment method. Improved understanding of the pathomechanisms leading to keloid formation will hopefully identify pathways that serve as specific targets to improve therapy for this devastating, currently intractable, disorder.
Collapse
Affiliation(s)
- Jonathan P Andrews
- Department of Dermatology and Cutaneous Biology, The Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jaana Marttala
- Department of Dermatology and Cutaneous Biology, The Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Edward Macarak
- Department of Dermatology and Cutaneous Biology, The Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; The Joan and Joel Rosenbloom Research Center for Fibrotic Diseases, Thomas Jefferson University, Philadelphia, PA, USA
| | - Joel Rosenbloom
- Department of Dermatology and Cutaneous Biology, The Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; The Joan and Joel Rosenbloom Research Center for Fibrotic Diseases, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jouni Uitto
- Department of Dermatology and Cutaneous Biology, The Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|