1
|
Wijesinghe RE, Kahatapitiya NS, Lee C, Han S, Kim S, Saleah SA, Seong D, Silva BN, Wijenayake U, Ravichandran NK, Jeon M, Kim J. Growing Trend to Adopt Speckle Variance Optical Coherence Tomography for Biological Tissue Assessments in Pre-Clinical Applications. MICROMACHINES 2024; 15:564. [PMID: 38793137 PMCID: PMC11122893 DOI: 10.3390/mi15050564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024]
Abstract
Speckle patterns are a generic feature in coherent imaging techniques like optical coherence tomography (OCT). Although speckles are granular like noise texture, which degrades the image, they carry information that can be benefited by processing and thereby furnishing crucial information of sample structures, which can serve to provide significant important structural details of samples in in vivo longitudinal pre-clinical monitoring and assessments. Since the motions of tissue molecules are indicated through speckle patterns, speckle variance OCT (SV-OCT) can be well-utilized for quantitative assessments of speckle variance (SV) in biological tissues. SV-OCT has been acknowledged as a promising method for mapping microvasculature in transverse-directional blood vessels with high resolution in micrometers in both the transverse and depth directions. The fundamental scope of this article reviews the state-of-the-art and clinical benefits of SV-OCT to assess biological tissues for pre-clinical applications. In particular, focus on precise quantifications of in vivo vascular response, therapy assessments, and real-time temporal vascular effects of SV-OCT are primarily emphasized. Finally, SV-OCT-incorporating pre-clinical techniques with high potential are presented for future biomedical applications.
Collapse
Affiliation(s)
- Ruchire Eranga Wijesinghe
- Department of Electrical and Electronic Engineering, Faculty of Engineering, Sri Lanka Institute of Information Technology, Malabe 10115, Sri Lanka;
- Center for Excellence in Intelligent Informatics, Electronics & Transmission (CIET), Sri Lanka Institute of Information Technology, Malabe 10115, Sri Lanka
| | - Nipun Shantha Kahatapitiya
- Department of Computer Engineering, Faculty of Engineering, University of Sri Jayewardenepura, Nugegoda 10250, Sri Lanka; (N.S.K.); (U.W.)
| | - Changho Lee
- Department of Artificial Intelligence Convergence, Chonnam National University, Gwangju 61186, Republic of Korea
- Department of Nuclear Medicine, Chonnam National University Medical School & Hwasun Hospital, 264, Seoyang-ro, Hwasun 58128, Republic of Korea
| | - Sangyeob Han
- ICT Convergence Research Center, Kyungpook National University, 80, Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Shinheon Kim
- ICT Convergence Research Center, Kyungpook National University, 80, Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Sm Abu Saleah
- ICT Convergence Research Center, Kyungpook National University, 80, Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Daewoon Seong
- School of Electronic and Electrical Engineering, College of IT Engineering, Kyungpook National University, 80, Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Bhagya Nathali Silva
- Center for Excellence in Intelligent Informatics, Electronics & Transmission (CIET), Sri Lanka Institute of Information Technology, Malabe 10115, Sri Lanka
- Faculty of Computing, Sri Lanka Institute of Information Technology, Malabe 10115, Sri Lanka
| | - Udaya Wijenayake
- Department of Computer Engineering, Faculty of Engineering, University of Sri Jayewardenepura, Nugegoda 10250, Sri Lanka; (N.S.K.); (U.W.)
| | - Naresh Kumar Ravichandran
- Center for Scientific Instrumentation, Korea Basic Science Institute, 169-148, Gwahak-ro, Yuseong-gu, Daejeon 34133, Republic of Korea
| | - Mansik Jeon
- School of Electronic and Electrical Engineering, College of IT Engineering, Kyungpook National University, 80, Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Jeehyun Kim
- School of Electronic and Electrical Engineering, College of IT Engineering, Kyungpook National University, 80, Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| |
Collapse
|
2
|
Murakami T, Shigeki S. Pharmacotherapy for Keloids and Hypertrophic Scars. Int J Mol Sci 2024; 25:4674. [PMID: 38731893 PMCID: PMC11083137 DOI: 10.3390/ijms25094674] [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: 03/14/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Keloids (KD) and hypertrophic scars (HTS), which are quite raised and pigmented and have increased vascularization and cellularity, are formed due to the impaired healing process of cutaneous injuries in some individuals having family history and genetic factors. These scars decrease the quality of life (QOL) of patients greatly, due to the pain, itching, contracture, cosmetic problems, and so on, depending on the location of the scars. Treatment/prevention that will satisfy patients' QOL is still under development. In this article, we review pharmacotherapy for treating KD and HTS, including the prevention of postsurgical recurrence (especially KD). Pharmacotherapy involves monotherapy using a single drug and combination pharmacotherapy using multiple drugs, where drugs are administered orally, topically and/or through intralesional injection. In addition, pharmacotherapy for KD/HTS is sometimes combined with surgical excision and/or with physical therapy such as cryotherapy, laser therapy, radiotherapy including brachytherapy, and silicone gel/sheeting. The results regarding the clinical effectiveness of each mono-pharmacotherapy for KD/HTS are not always consistent but rather scattered among researchers. Multimodal combination pharmacotherapy that targets multiple sites simultaneously is more effective than mono-pharmacotherapy. The literature was searched using PubMed, Google Scholar, and Online search engines.
Collapse
Affiliation(s)
- Teruo Murakami
- Laboratory of Biopharmaceutics and Pharmacokinetics, Faculty of Pharmaceutical Sciences, Hiroshima International University, Higashi-Hiroshima 731-2631, Japan;
| | - Sadayuki Shigeki
- Department of Rehabilitation, Faculty of Rehabilitation, Hiroshima International University, Higashi-Hiroshima 731-2631, Japan
| |
Collapse
|
3
|
Mokhtar SMA, Derrick-Roberts ALK, Evans DR, Strudwick XL. Cell Viability Assessment of PEDOT Conducting Polymer-Coated Microneedles for Skin Sampling. ACS APPLIED BIO MATERIALS 2023; 6:4662-4671. [PMID: 37902811 DOI: 10.1021/acsabm.3c00416] [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] [Indexed: 10/31/2023]
Abstract
Recently, transdermal monitoring and drug delivery have gained much interest, owing to the introduction of the minimally invasive microneedle (MN) device. The advancement of electroactive MNs electrically assisted in the capture of biomarkers or the triggering of drug release. Recent works have combined conducting polymers (CPs) onto MNs owing to the soft nature of the polymers and their tunable ionic and electronic conductivity. Though CPs are reported to work safely in the body, their biocompatibility in the skin has been insufficiently investigated. Furthermore, during electrical biasing of CPs, they undergo reduction or oxidation, which in practical terms leads to release/exchange of ions, which could pose biological risks. This work investigates the viability and proliferation of skin cells upon exposure to an electrochemically biased MN pair comprising two differently doped poly(3,4-ethylenedioxy-thiophene) (PEDOT) polymers that have been designed for skin sampling use. The impact of biasing on human keratinocytes and dermal fibroblasts was determined at different initial cell seeding densities and incubation periods. Indirect testing was employed, whereby the culture media was first exposed to PEDOTs prior to the addition of this extract to cells. In all conditions, both unbiased and biased PEDOT extracts showed no cytotoxicity, but the viability and proliferation of cells cultured at a low cell seeding density were lower than those of the control after 48 h of incubation.
Collapse
Affiliation(s)
- Siti Musliha Ajmal Mokhtar
- Future Industries Institute, UniSA STEM, University of South Australia, Mawson Lakes, SA 5095, Australia
- College of Engineering, Universiti Teknologi MARA, Johor Branch, Pasir Gudang Campus, Masai, Johor 81750, Malaysia
| | | | - Drew R Evans
- Future Industries Institute, UniSA STEM, University of South Australia, Mawson Lakes, SA 5095, Australia
| | - Xanthe L Strudwick
- Future Industries Institute, UniSA STEM, University of South Australia, Mawson Lakes, SA 5095, Australia
| |
Collapse
|
4
|
Yuan B, Upton Z, Leavesley D, Fan C, Wang XQ. Vascular and Collagen Target: A Rational Approach to Hypertrophic Scar Management. Adv Wound Care (New Rochelle) 2023; 12:38-55. [PMID: 34328823 PMCID: PMC9595647 DOI: 10.1089/wound.2020.1348] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Significance: Hypertrophic scarring is a challenging issue for patients and clinicians. The prevalence of hypertrophic scarring can be up to 70% after burns, and patients suffer from pain, itching, and loss of joint mobility. To date, the exact mechanisms underlying hypertrophic scar formation are unclear, and clinical options remain limited. Recent Advances: Several studies have demonstrated that pathological scars are a type of hyperactive vascular response to wounding. Scar regression has been found to be accompanied by microvessel occlusion, which causes severe hypoxia, malnutrition, and endothelial dysfunction, suggesting the essential roles of microvessels in scar regression. Therefore, interventions that target the vasculature, such as intense pulsed light, pulsed dye lasers, vascular endothelial growth factor antibodies, and Endostar, represent potential treatments. In addition, the mass of scar-associated collagen is usually not considered by current treatments. However, collagen-targeted therapies such as fractional CO2 laser and collagenase have shown promising outcomes in scar treatment. Critical Issues: Traditional modalities used in current clinical practice only partially target scar-associated microvessels or collagen. As a result, the effectiveness of current treatments is limited and is too often accompanied by undesirable side effects. The formation of scars in the early stage is mainly affected by microvessels, whereas the scars in later stages are mostly composed of residual collagen. Traditional therapies do not utilize specific targets for scars at different stages. Therefore, more precise treatment strategies are needed. Future Directions: Scars should be classified as either "vascular-dominant" or "collagen-dominant" before selecting a treatment. In this way, strategies that are vascular-targeted, collagen-targeted, or a combination thereof could be recommended to treat scars at different stages.
Collapse
Affiliation(s)
- Bo Yuan
- Burns and Plastic Surgery Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Zee Upton
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Skin Research Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - David Leavesley
- Skin Research Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Chen Fan
- Skin Research Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
- Correspondence: Chen Fan, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Xi-Qiao Wang
- Burns and Plastic Surgery Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
- Correspondence: Xi-Qiao Wang, Burns and Plastic Surgery Department, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, P.R. China
| |
Collapse
|
5
|
Wang X, Liang B, Li J, Pi X, Zhang P, Zhou X, Chen X, Zhou S, Yang R. Identification and characterization of four immune-related signatures in keloid. Front Immunol 2022; 13:942446. [PMID: 35967426 PMCID: PMC9365668 DOI: 10.3389/fimmu.2022.942446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/27/2022] [Indexed: 11/29/2022] Open
Abstract
A keloid is a fibroproliferative disorder of unknown etiopathogenesis that requires ill-defined treatment. Existing evidence indicates that the immune system plays an important role in the occurrence and development of keloid. However, there is still a lack of research on the immune-related signatures of keloid. Here we identified immune-related signatures in keloid and explored their pathological mechanisms. Transcriptomic datasets (GSE7890, GSE92566, and GSE44270) of keloid and normal skin tissues were obtained from the Gene Expression Omnibus database. The overlap of differentially expressed genes and immune-related genes was considered as differentially expressed immune-related genes (DEIGs). Functional analysis, expression, and distribution were applied to explore the function and characteristics of DEIGs, and the expression of these DEIGs in keloid and normal skin tissues was verified by immunohistochemistry. Finally, we conducted interactive network analysis and immune infiltration analysis to determine the therapeutic potential and immune correlation. We identified four DEIGs (LGR5, PTN, JAG1, and DKK1). In these datasets, only GSE7890 met the screening criteria. In the GSE7890 dataset, DKK1 and PTN were downregulated in keloid, whereas JAG1 and LGR5 were upregulated in keloid. In addition, we obtained the same conclusion through immunohistochemistry. Functional analysis indicated that these four DEIGs were mainly involved in stem cell, cell cycle, UV response, and therapy resistance. Through interactive network analysis, we found that these DEIGs were associated with drugs currently used to treat keloid, such as hydrocortisone, androstanolone, irinotecan, oxaliplatin, BHQ-880, and lecoleucovorin. Finally, many immune cells, including CD8+ T cells, resting memory CD4+ T cells, and M1 macrophages, were obtained by immune infiltration analysis. In conclusion, we identified four immune signaling molecules associated with keloid (LGR5, PTN, JAG1, and DKK1). These immune-related signaling molecules may be important modules in the pathogenesis of keloid. Additionally, we developed novel therapeutic targets for the treatment of this challenging disease.
Collapse
Affiliation(s)
- Xiaoxiang Wang
- Guangdong Medical University, Zhanjiang, China
- Department of Burn Surgery, The First People’s Hospital of Foshan, Foshan, China
| | - Bo Liang
- The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiehua Li
- Department of Dermatology, The First People’s Hospital of Foshan, Foshan, China
| | - Xiaobing Pi
- Department of Dermatology, The First People’s Hospital of Foshan, Foshan, China
| | - Peng Zhang
- Neijiang Health Vocational College, Neijiang, China
| | - Xinzhu Zhou
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xiaodong Chen
- Department of Burn Surgery, The First People’s Hospital of Foshan, Foshan, China
- *Correspondence: Xiaodong Chen, ; Sitong Zhou, ; Ronghua Yang,
| | - Sitong Zhou
- Department of Dermatology, The First People’s Hospital of Foshan, Foshan, China
- *Correspondence: Xiaodong Chen, ; Sitong Zhou, ; Ronghua Yang,
| | - Ronghua Yang
- Guangdong Medical University, Zhanjiang, China
- Department of Burn and Plastic Surgery, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
- *Correspondence: Xiaodong Chen, ; Sitong Zhou, ; Ronghua Yang,
| |
Collapse
|
6
|
Angiogenic gene characterization and vessel permeability of dermal microvascular endothelial cells isolated from burn hypertrophic scar. Sci Rep 2022; 12:12222. [PMID: 35851095 PMCID: PMC9293893 DOI: 10.1038/s41598-022-16376-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 07/08/2022] [Indexed: 02/06/2023] Open
Abstract
Hypertrophic scar (HTS) formation is a common challenge for patients after burn injury. Dermal microvascular endothelial cells (DMVECs) are an understudied cell type in HTS. An increase in angiogenesis and microvessel density can be observed in HTS. Endothelial dysfunction may play a role in scar development. This study aims to generate a functional and expression profile of HTS DMVECs. We hypothesize that transcript and protein-level responses in HTS DMVECs differ from those in normal skin (NS). HTSs were created in red Duroc pigs. DMVECs were isolated using magnetic-activated cell sorting with ulex europaeus agglutinin 1 (UEA-1) lectin. Separate transwell inserts were used to form monolayers of HTS DMVECs and NS DMVECs. Cell injury was induced and permeability was assessed. Gene expression in HTS DMVECS versus NS DMVECs was measured. Select differentially expressed genes were further investigated. HTS had an increased area density of dermal microvasculature compared to NS. HTS DMVECs were 17.59% less permeable than normal DMVECs (p < 0.05). After injury, NS DMVECs were 28.4% and HTS DMVECs were 18.8% more permeable than uninjured controls (28.4 ± 4.8 vs 18.8 ± 2.8; p = 0.11). PCR array identified 31 differentially expressed genes between HTS and NS DMVECs, of which 10 were upregulated and 21 were downregulated. qRT-PCR and ELISA studies were in accordance with the array. DMVECs expressed a mixed profile of factors that can contribute to and inhibit scar formation. HTS DMVECs have both a discordant response to cellular insults and baseline differences in function, supporting their proposed role in scar pathology. Further investigation of DMVECs is warranted to elucidate their contribution to HTS pathogenesis.
Collapse
|
7
|
Immunohistochemical Analysis of Postburn Scars following Treatment Using Dermal Substitutes. Anal Cell Pathol (Amst) 2022; 2022:3686863. [PMID: 35251908 PMCID: PMC8896958 DOI: 10.1155/2022/3686863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 12/08/2021] [Accepted: 01/20/2022] [Indexed: 11/28/2022] Open
Abstract
Background Post-burn hypertrophic scars commonly occur after burns. Studies that compare dermal substitutes with other treatment methods are insufficient. The purpose was to analyze the histopathological differences in hypertrophic burn scars after Matriderm®+split-thickness skin graft (STSG) and compare with AlloDerm®+STSG, STSG, full-thickness skin graft (FTSG), and normal skin. Methods Samples of unburned, normal skin and deep 2nd or 3rd degree burns were obtained from patients who experienced a burn injury in the past to at least 6 months before biopsy, which was performed between 2011 and 2012. All subjects received >6 months of treatment before the biopsy. Intervention groups were normal (63), STSG (28), FTSG (6), Matriderm® (11), and AlloDerm® (18). Immunohistochemical analyses of elastin, collagen I, collagen III, cluster of differentiation 31 (CD31), smooth muscle actin (α-SMA), and laminin from scar and control tissues were performed and compared. Results α-SMA vascular quantity and vessel width, stromal CD31, and basement membrane laminin expression were not significantly different between normal and intervention groups. Matriderm® group showed no significant difference in elastin, collagen III, stromal CD31 and α-SMA, CD31 vessel width, stromal α-SMA, vessel quantity and width, and laminin length compared to the normal group, meaning they were not significantly different from the normal skin traits. Conclusion Dermal substitutes may be an optimal alternative to address the cosmetic and functional limitations posed by other treatment methods.
Collapse
|
8
|
Zheng W, Lin G, Wang Z. Bioinformatics study on different gene expression profiles of fibroblasts and vascular endothelial cells in keloids. Medicine (Baltimore) 2021; 100:e27777. [PMID: 34964740 PMCID: PMC8615345 DOI: 10.1097/md.0000000000027777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 10/28/2021] [Indexed: 01/05/2023] Open
Abstract
Keloid is a benign fibroproliferative skin tumor. The respective functions of fibroblasts and vascular endothelial cells in keloid have not been fully studied. The purpose of this study is to identify the respective roles and key genes of fibroblasts and vascular endothelial cells in keloids, which can be used as new targets for diagnosis or treatment.The microarray datasets of keloid fibroblasts and vascular endothelial cells were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were screened out. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used for functional enrichment analysis. The search tool for retrieval of interacting genes and Cytoscape were used to construct protein-protein interaction (PPI) networks and analyze gene modules. The hub genes were screened out, and the relevant interaction networks and biological process analysis were carried out.In fibroblasts, the DEGs were significantly enriched in collagen fibril organization, extracellular matrix organization and ECM-receptor interaction. The PPI network was constructed, and the most significant module was selected, which is mainly enriched in ECM-receptor interaction. In vascular endothelial cells, the DEGs were significantly enriched in cytokine activity, growth factor activity and transforming growth factor-β (TGF-β) signaling pathway. Module analysis was mainly enriched in TGF-β signaling pathway. Hub genes were screened out separately.In summary, the DEGs and hub genes discovered in this study may help us understand the molecular mechanisms of keloid, and provide potential targets for diagnosis and treatment.
Collapse
Affiliation(s)
- Weihan Zheng
- School of Basic Medicine, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Guojian Lin
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, PR China
| | - Zhizhou Wang
- School of Basic Medicine, Fujian Medical University, Fuzhou, Fujian, PR China
| |
Collapse
|
9
|
Chen C, Zhang M, Yu N, Zhang W, Long X, Wang Y, Wang X. Heterogeneous Features of Keloids Assessed by Laser Speckle Contrast Imaging: A Cross-Sectional Study. Lasers Surg Med 2020; 53:865-871. [PMID: 33027537 DOI: 10.1002/lsm.23331] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 09/01/2020] [Accepted: 09/20/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND OBJECTIVES Keloids are described as benign dermal fibroproliferative lesions, and vascularization may play a significant role in their pathogenesis. In this study, laser speckle contrast imaging (LSCI) was used to assess perfusion within keloids and surrounding skin, and perfusion of keloids at different stages was compared. STUDY DESIGN/MATERIALS AND METHODS A total of 59 patients with 110 untreated keloids on the anterior chest were enrolled in this study. Different keloid stages (progressive, stable, and regressive) were defined according to patients' descriptions of whether keloids became larger, stable, or smaller during the previous year. Vancouver Scar Scale (VSS) was assessed by a plastic surgeon, and patient reports on pain and itching were documented. LSCI was used to evaluate blood perfusion of keloids (K), skin adjacent to keloids (A), and nonadjacent skin (N). The mean perfusion of these regions was determined, and ratios (K/N, A/N) were calculated. RESULTS A heterogeneous perfusion map was observed among the keloid groups, as well as within each keloid. A positive correlation was found between keloid perfusion and VSS. There were 62 (56.4%) keloids in the progressive stage, 33 (30.0%) keloids in the stable stage, and 15 (13.6%) keloids in the regressive stage. The mean K/N ratios in the progressive, stable, and regressive stages were 2.3 ± 0.5, 1.8 ± 0.3, and 1.5 ± 0.5, respectively. The mean A/N ratios were 1.2 ± 0.4, 1.2 ± 0.2, and 1.0 ± 0.5, respectively. Within each keloid, significantly higher perfusion was noted in the keloid and adjacent skin compared with nonadjacent skin. CONCLUSION These results indicate that LSCI is a promising technique for evaluating keloid blood perfusion and distinguishing heterogeneous keloids. Lasers Surg. Med. © 2020 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.,Department of Ultrasound, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Mingzi Zhang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Nanze Yu
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Wenchao Zhang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xiao Long
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Youbin Wang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xiaojun Wang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| |
Collapse
|
10
|
Wang Q, Miao M, Qin Z, Li B, Niu X. Lower Metal Element Levels in Hypertrophic Scars: A Potential Mechanism of Aberrant Cicatrix Hyperplasia. Med Sci Monit 2020; 26:e925202. [PMID: 32895361 PMCID: PMC7500125 DOI: 10.12659/msm.925202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background We investigated levels of the metal elements Ca, Mg, Zn, Fe, and Cu in blood, normal skin (NS), and different types of scar tissue and aimed to elucidate the pathogenesis of hypertrophic scars (HS). Material/Methods Tissue specimens were excised from 3 groups of research participants: scar-free, flat scar (FS), and HS groups. Levels of the study elements were measured in blood, NS, and scar tissues with a spectrophotometer. The levels in plasma or in different types of specimens were compared among subgroups. In the FS and HS groups, levels were compared between the scar tissue and NS of each individual. In addition, element differences in exposed and unexposed areas of NS were investigated in the scar-free group. HS fibroblasts (HFB) were cultured in medium with various reduced levels of metal elements to determine the influence of metal elements on fibroblast growth. Results Levels of trace elements, including Zn, Fe, and Cu, were significantly lower in HS than in FS. The levels of Ca, Zn, Fe, and Cu were markedly lower in HS than in the patients’ own NS, while the Cu/Zn ratio was higher. However, no such difference was observed in the FS group. No significant difference in element levels was found in either plasma or NS among the 3 groups. Reduced levels of the elements promoted HFB proliferation within 24 h while an inhibition effect was observed at 72 h. Conclusions Our findings indicate reduced levels of metal elements in part of the healing microenvironment, suggesting that decreased metal levels may be involved in the pathogenesis of HS.
Collapse
Affiliation(s)
- Qifei Wang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China (mainland)
| | - Mi Miao
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China (mainland)
| | - Zelian Qin
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China (mainland)
| | - Bolun Li
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China (mainland)
| | - Xingtao Niu
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China (mainland)
| |
Collapse
|
11
|
Matsumoto NM, Aoki M, Okubo Y, Kuwahara K, Eura S, Dohi T, Akaishi S, Ogawa R. Gene Expression Profile of Isolated Dermal Vascular Endothelial Cells in Keloids. Front Cell Dev Biol 2020; 8:658. [PMID: 32850798 PMCID: PMC7403211 DOI: 10.3389/fcell.2020.00658] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/01/2020] [Indexed: 01/01/2023] Open
Abstract
Wound healing is a complex biological process, and imbalances of various substances in the wound environment may prolong healing and lead to excessive scarring. Keloid is abnormal proliferation of scar tissue beyond the original wound margins with excessive deposition of extracellular matrix (ECM) and chronic inflammation. Despite numerous previous research efforts, the pathogenesis of keloid remains unknown. Vascular endothelial cells (VECs) are a major type of inductive cell in inflammation and fibrosis. Despite several studies on vascular morphology in keloid formation, there has been no functional analysis of the role of VECs. In the present study, we isolated living VECs from keloid tissues and investigated gene expression patterns using microarray analysis. We obtained 5 keloid tissue samples and 6 normal skin samples from patients without keloid. Immediately after excision, tissue samples were gently minced and living cells were isolated. Magnetic-activated cell sorting of VECs was performed by negative selection of fibroblasts and CD45+ cells and by positive selection of CD31+cells. After RNA extraction, gene expression analysis was performed to compare VECs isolated from keloid tissue (KVECs) with VECs from normal skin (NVECs). After cell isolation, the percentage of CD31+ cells as measured by flow cytometry ranged from 81.8%–98.6%. Principal component analysis was used to identify distinct molecular phenotypes in KVECs versus NVECs and these were divided into two subgroups. In total, 15 genes were upregulated, and 3 genes were downregulated in KVECs compared with NVECs using the t-test (< 0.05). Quantitative RT-PCR and immunohistochemistry showed 16-fold and 11-fold overexpression of SERPINA3 and LAMC2, respectively. SERPINA3 encodes the serine protease inhibitor, α1-antichymotripsin. Laminin γ2-Chain (LAMC2) is a subunit of laminin-5 that induces retraction of vascular endothelial cells and enhances vascular permeability. This is the first report of VEC isolation and gene expression analysis in keloid tissue. Our data suggest that SERPINA3 and LAMC2 upregulation in KVECs may contribute to the development of fibrosis and prolonged inflammation in keloid. Further functional investigation of these genes will help clarify the mechanisms of abnormal scar tissue proliferation.
Collapse
Affiliation(s)
- Noriko M Matsumoto
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Masayo Aoki
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan.,Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Yuri Okubo
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Kosuke Kuwahara
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Shigeyoshi Eura
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Teruyuki Dohi
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Satoshi Akaishi
- Department of Plastic Surgery, Nippon Medical School Musashi Kosugi Hospital, Kawasaki, Japan
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
12
|
Zeng J, Jiang B, Xiao X, Zhang R. Inhibition of sphingosine kinase 2 attenuates hypertrophic scar formation via upregulation of Smad7 in human hypertrophic scar fibroblasts. Mol Med Rep 2020; 22:2573-2582. [PMID: 32705254 PMCID: PMC7411334 DOI: 10.3892/mmr.2020.11313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 05/20/2020] [Indexed: 12/18/2022] Open
Abstract
The aims of the present study were to investigate the role of sphingosine kinase 2 (Sphk2) in hypertrophic scar (HS) formation and its underlying mechanisms. The expression levels of Sphk2 and Smad7 in HS tissues and healthy skin tissues of patients undergoing plastic surgery were determined using immunohistochemical staining. Subsequently, the expression levels of Sphk2 and collagen I in human embryonic skin fibroblasts (control) and human HS fibroblasts (HSF) were detected using western blot analysis and immunofluorescence assay, respectively. Following Sphk2 silencing, Smad7 overexpression or both Sphk2 and Smad7 silencing, HSF proliferative ability was assessed using Cell Counting Kit‑8 assay and proliferation‑associated proteins were evaluated using western blot analysis. In addition, the level of apoptosis in HSF was assessed using flow cytometry and expression levels of apoptotic‑associated proteins were determined using western blotting. Furthermore, the expression levels of collagen I and proteins in the TGF‑β1/Smad signaling pathway were detected using western blot analysis. The results indicated that the expression of Sphk2 was significantly increased, while Smad7 expression was decreased in HS tissue. Moreover, the upregulation of Sphk2 and collagen I expression levels was identified in HSF. The present results also indicated that Sphk2 silencing or Smad7 overexpression inhibited proliferation, but promoted apoptosis of HSF, coupled with changes in the expression levels of proliferation‑associated proteins, with an increase in p21 and a decrease in cyclin D1 expression levels, and apoptosis‑associated proteins, with an increase in Bax and cleaved caspase‑3, and a decrease in Bcl‑2, which were reversed following transfection with both Sphk2 and Smad7 using small interfering RNA in HSF. In addition, the expression levels of transforming growth factor‑β1, phosphorylated (p)‑Smad2, p‑Smad3 and collagen I were reduced following Sphk2 silencing or Smad7 overexpression, which were abolished by silencing both Sphk2 and Smad7. Collectively, the present results indicated that inhibition of Sphk2 attenuated HS formation via upregulation of Smad7 expression, thus Sphk2 may serve as a potential therapeutic target for the treatment of HS.
Collapse
Affiliation(s)
- Jian Zeng
- Department of Medical Cosmetology, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Bin Jiang
- Department of Medical Cosmetology, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xia Xiao
- Department of Medical Cosmetology, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Rou Zhang
- Department of Medical Cosmetology, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
13
|
Kruglikov IL, Scherer PE. Caveolin-1 as a target in prevention and treatment of hypertrophic scarring. NPJ Regen Med 2019; 4:9. [PMID: 31044089 PMCID: PMC6486604 DOI: 10.1038/s41536-019-0071-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 04/02/2019] [Indexed: 12/11/2022] Open
Abstract
Reduced expression of caveolin-1 (Cav-1) is an important pathogenic factor in hypertrophic scarring (HTS). Such a reduction can be found in connection with the main known risk factors for HTS, including dark skin, female gender, young age, burn site and severity of the injury. The degree of overexpression of Cav-1 associated with different therapeutic options for HTS correlates with clinical improvements in HTS. This makes endo- or exogenous induction of Cav-1 not only an important therapeutic target for HTS, but also highlights its use as a preventive target to reduce or avoid HTS formation.
Collapse
Affiliation(s)
| | - Philipp E. Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX 75390-8549 USA
| |
Collapse
|
14
|
Liu B, Guo Z, Gao W. miR-181b-5p promotes proliferation and inhibits apoptosis of hypertrophic scar fibroblasts through regulating the MEK/ERK/p21 pathway. Exp Ther Med 2019; 17:1537-1544. [PMID: 30783419 PMCID: PMC6364240 DOI: 10.3892/etm.2019.7159] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/26/2018] [Indexed: 12/22/2022] Open
Abstract
Hypertrophic scar (HS) is a common skin disorder occurring during the wound healing process, and the pathogenesis of HS remains unclear. Previous studies indicated that miRNAs may be involved in the onset and progression of HS. In the present study, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting were used to investigate the expression of miR-181b-5p and decorin in HS tissues. Direct interaction between miR-181b-5p and decorin was confirmed using a dual-luciferase assay. Human HS fibroblasts (HSFbs) were cultured and transfected with miR-181b-5p mimics, and MTT assay and Annexin V fluorescein isothiocyanate/propidium iodide staining were performed to investigate the role of miR-181b-5p in the proliferation and apoptosis of HSFbs. Subsequently, the expression levels of mitogen-activated protein kinase kinase (MEK), phospho (p)-extracellular signal-regulated kinase (ERK) and p21 were determined in HSFbs transfected with miR-181b-5p mimics and untransfected cells using RT-qPCR and western blotting. The results indicated upregulation of miR-181b-5p and downregulation of decorin expression in HS tissues compared with normal skin samples. miR-181b-5p may regulate the expression of decorin through direct binding to the 3′-untranslated region, as demonstrated by the results of the dual-luciferase assay. Transfection with miR-181b-5p mimics in HSFbs enhanced cell proliferation, reduced apoptosis and increased the expression of MEK, p-ERK and p21. Furthermore, treatment with MEK inhibitor in HSFbs transfected with miR-181b-5p mimics partially inhibited miR-181b-5p-induced antiapoptotic effects. Taken together, increased expression of miR-181b-5p may serve important roles in the pathogenesis of HS through regulating the MEK/ERK/p21 pathway, suggesting that miR-181b-5p may be a therapeutic target for the treatment of HS.
Collapse
Affiliation(s)
- Bo Liu
- Department of Medical Cosmetology, Eastern Liaoning University, Dandong, Liaoning 118003, P.R. China
| | - Zhe Guo
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Weiming Gao
- Department of Medicine, Eastern Liaoning University, Dandong, Liaoning 118003, P.R. China
| |
Collapse
|
15
|
Monsuur HN, van den Broek LJ, Koolwijk P, Niessen FB, Gibbs S. Endothelial cells enhance adipose mesenchymal stromal cell-mediated matrix contraction via ALK receptors and reduced follistatin: Potential role of endothelial cells in skin fibrosis. J Cell Physiol 2018; 233:6714-6722. [PMID: 29345319 PMCID: PMC6056025 DOI: 10.1002/jcp.26494] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/17/2018] [Indexed: 12/18/2022]
Abstract
Abnormal cutaneous wound healing can lead to formation of fibrotic hypertrophic scars. Although several clinical risk factors have been described, the cross‐talk between different cell types resulting in hypertrophic scar formation is still poorly understood. The aim of this in vitro study was to investigate whether endothelial cells (EC) may play a role in skin fibrosis, for example, hypertrophic scar formation after full‐thickness skin trauma. Using a collagen/elastin matrix, we developed an in vitro fibrosis model to study the interaction between EC and dermal fibroblasts or adipose tissue‐derived mesenchymal stromal cells (ASC). Tissue equivalents containing dermal fibroblasts and EC displayed a normal phenotype. In contrast, tissue equivalents containing ASC and EC displayed a fibrotic phenotype indicated by contraction of the matrix, higher gene expression of ACTA2, COL1A, COL3A, and less secretion of follistatin. The contraction was in part mediated via the TGF‐β pathway, as both inhibition of the ALK4/5/7 receptors and the addition of recombinant follistatin resulted in decreased matrix contraction (75 ± 11% and 24 ± 8%, respectively). In conclusion, our study shows that EC may play a critical role in fibrotic events, as seen in hypertrophic scars, by stimulating ASC‐mediated matrix contraction via regulation of fibrosis‐related proteins.
Collapse
Affiliation(s)
- Hanneke N Monsuur
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Lenie J van den Broek
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Pieter Koolwijk
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Frank B Niessen
- Department of Plastic, Reconstructive and Hand Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Dermatology, VU University Medical Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands.,Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| |
Collapse
|