1
|
Jiang W, Pang X, Ha P, Li C, Chang GX, Zhang Y, Bossong LA, Ting K, Soo C, Zheng Z. Fibromodulin selectively accelerates myofibroblast apoptosis in cutaneous wounds by enhancing interleukin 1β signaling. Nat Commun 2025; 16:3499. [PMID: 40221432 PMCID: PMC11993684 DOI: 10.1038/s41467-025-58906-z] [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: 04/16/2024] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Activated myofibroblasts deposit extracellular matrix material to facilitate rapid wound closure that can heal scarlessly during fetal development. However, adult myofibroblasts exhibit a relatively long life and persistent function, resulting in scarring. Thus, understanding how fetal and adult tissue regeneration differs may serve to identify factors that promote more optimal wound healing in adults with little or less scarring. We previously found that matricellular proteoglycan fibromodulin is one such factor promoting more optimal repair, but the underlying molecular and cellular mechanisms for these effects have not been fully elucidated. Here, we find that fibromodulin induces myofibroblast apoptosis after wound closure to reduce scarring in small and large animal models. Mechanistically, fibromodulin accelerates and prolongs the formation of the interleukin 1β-interleukin 1 receptor type 1-interleukin 1 receptor accessory protein ternary complex to increase the apoptosis of myofibroblasts and keloid- and hypertrophic scar-derived cells. As the persistence of myofibroblasts during tissue regeneration is a key cause of fibrosis in most organs, fibromodulin represents a promising, broad-spectrum anti-fibrotic therapeutic.
Collapse
Affiliation(s)
- Wenlu Jiang
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xiaoxiao Pang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral, Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, 401147, China
| | - Pin Ha
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Chenshuang Li
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Grace Xinlian Chang
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yuxin Zhang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral, Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, 401147, China
| | - Lawrence A Bossong
- Department of Neuroscience, Princeton University, Princeton, NJ, 08540, USA
| | - Kang Ting
- American Dental Association Forsyth Institute, Cambridge, MA, 02142, USA.
- School of Dentistry, National Yang-Ming Chiao Tung University, Taipei, 30010, Taiwan.
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Zhong Zheng
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
2
|
Song J, Li M, Tao Y, Li Y, Mai C, Zhang J, Yao L, Shi S, Xu J. Enhanced myofibroblast differentiation of eMSCs in intrauterine adhesions. Stem Cell Res Ther 2025; 16:35. [PMID: 39901307 PMCID: PMC11792338 DOI: 10.1186/s13287-025-04183-y] [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: 08/29/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Intrauterine adhesions (IUA) is one of the most common gynecological diseases and main causes of uterine infertility. Among proposed hypotheses on IUA development, the reduced endometrial regeneration resulting from loss of functional stem cells has been proposed as the key factor affecting the IUA prognosis. However, the underlying mechanisms mostly remain unclear. Because the eMSCs (endometrial mesenchymal stem/stromal cells) play a critical role in both supporting the gland development and also preparing the environment for embryo implantation through decidualization, the characteristics and functions were compared between the eMSCs derived from IUA and non-IUA patients, to uncover the important roles of eMSCs in IUA and also the underlying mechanisms. METHODS Endometrium biopsies were collected from IUA patients and controls. The fibrosis features and eMSC distributions were investigated with IHC (immunohistochemistry). Then the eMSCs were isolated and their functions and characteristics were analyzed in vitro. RESULTS Our results indicate that the scar tissues in IUA are characterized with hyper-activation of pro-fibrotic fibroblast and myo-differentiation, along with reduced number of eMSCs. The isolated eMSCs from IUA and controls show similar functions from the perspectives of cell morphology, proliferation, colony formation, exosome secretion, positive ratio of eMSC markers and conventional MSC markers, tri-differentiation efficiency, the ability of suppressing lymphocyte proliferation, cell aging, and promoting vascular tube formation. However, the eMSCs from IUA have reduced levels of decidualization and higher levels of cell migration, invasion, and also myofibroblast differentiation. Further investigations indicate that the TGF-β pathway, which is the major inducer of myofibroblast differentiation, is up-regulated and responsible for the enhanced myofibroblast differentiation potential of eMSCs from IUA. CONCLUSIONS In conclusion, we have demonstrated here that the scar tissues in IUA biopsy are characterized with enhanced differentiation of pro-fibrotic fibroblast and myofibroblast. The number of eMSCs is reduced in IUA tissues, and their myofibroblast differentiation capability is increased.
Collapse
Affiliation(s)
- Jun Song
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150000, People's Republic of China
| | - Meiqi Li
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150000, People's Republic of China
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China
| | - Yuan Tao
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150000, People's Republic of China
| | - Yumeng Li
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China
| | - Canrong Mai
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China
| | - Jingting Zhang
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China
| | - Lan Yao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China
| | - Shaoquan Shi
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China.
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China.
| | - Jianyong Xu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, 518000, People's Republic of China.
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, 518000, People's Republic of China.
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Fuqiang Avenue 1001, Shenzhen, 518060, Guangdong, People's Republic of China.
| |
Collapse
|
3
|
Li R, Wang H, Wang X, Yang Y, Zhong K, Zhang X, Li H. MSC-EVs and UCB-EVs promote skin wound healing and spatial transcriptome analysis. Sci Rep 2025; 15:4006. [PMID: 39893214 PMCID: PMC11787299 DOI: 10.1038/s41598-025-87592-6] [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: 11/13/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025] Open
Abstract
Extracellular vesicles (EVs) are important paracrine mediators derived from various cells and biological fluids, including plasma, that are capable of inducing regenerative effects by transferring bioactive molecules such as microRNAs (miRNAs). This study investigated the effect of mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) isolated from umbilical cord blood and human umbilical cord plasma-derived extracellular vesicles (UCB-EVs) on wound healing and scar formation reduction. Spatial transcriptomics (ST) was used to study the effects of MSC-EVs and UCB-EVs on the heterogeneity of major cell types and wound healing pathways in mouse skin tissue. MSC-EVs and UCB-EVs were isolated using ultracentrifugation and identified using transmission electron microscopy, nanoparticle tracking analysis, and western blot. The effects of MSC-EVs and UCB-EVs on human dermal fibroblast-adult cell (HDF-a) migration and proliferation were evaluated using cell scratch assays, cell migration assays, and cell proliferation assays. In vivo, MSC-EVs and UCB-EVs were injected around full-cut wounds to evaluate their efficacy of wound healing by measuring wound closure rates and scar width and performing histological analysis. ST was performed on skin tissue samples from mice in each group after wound healing to analyze the heterogeneity of major cell types compared with the control group and investigate potential mechanisms affecting wound healing and scar formation. In vitro experiments demonstrated that MSC-EVs and UCB-EVs promoted the proliferation and migration of HDF-a cells. Local injection of MSC-EVs and UCB-EVs into the periphery of a mouse skin wound accelerated re-epithelialization, promoted wound healing, and reduced scar width. ST analysis of skin tissue from each group after wound healing revealed that MSC-EVs and UCB-EVs reduced the relative expression of marker genes in myofibroblasts, regulated wound healing, and decreased scar formation by reducing the expression of the TGF-β signaling pathway and increasing the expression of the Wnt signaling pathway. The results suggest that MSC-EVs and UCB-EVs play a significant role in the activity of cord blood plasma-derived mesenchymal stem cells and cord blood plasma. They can be considered promising new agents for promoting skin wound healing.
Collapse
Affiliation(s)
- Ruonan Li
- Key Laboratory of Animal Biochemistry and Nutrition of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Haotian Wang
- Key Laboratory of Animal Biochemistry and Nutrition of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Xiaolong Wang
- HenanYinfeng Biological Engineering Technology Co., LTD, No. 11 Changchun Road, Zhengzhou High tech Industrial Development Zone, Zhengzhou, 450000, China
| | - Yanbin Yang
- Key Laboratory of Animal Biochemistry and Nutrition of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Kai Zhong
- Key Laboratory of Animal Biochemistry and Nutrition of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China.
| | - Xuemei Zhang
- HenanYinfeng Biological Engineering Technology Co., LTD, No. 11 Changchun Road, Zhengzhou High tech Industrial Development Zone, Zhengzhou, 450000, China.
| | - Heping Li
- Key Laboratory of Animal Biochemistry and Nutrition of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China.
| |
Collapse
|
4
|
Gomes MLNP, Krijnen PAJ, Middelkoop E, Niessen HWM, Boekema BKHL. Fetal Skin Wound Healing: Key Extracellular Matrix Components and Regulators in Scarless Healing. J Invest Dermatol 2025; 145:280-302. [PMID: 39152955 DOI: 10.1016/j.jid.2024.05.027] [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: 09/26/2023] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 08/19/2024]
Abstract
Fetal skin at early gestational stage is able to regenerate and heal rapidly after wounding. The exact mechanisms and molecular pathways involved in this process are however still largely unknown. The numerous differences in the skin of the early fetus versus skin in later developmental stages might provide clues for the mechanisms of scarless healing. This review summarizes the differences between mammalian fetal skin and the skin at later developmental phases in healthy and wounded conditions, focusing on extracellular matrix components, which are crucial factors in the microenvironment that direct cells and tissue functions and hence the wound healing process.
Collapse
Affiliation(s)
- Madalena Lopes Natário Pinto Gomes
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands; Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, The Netherlands; Department of Pathology, Amsterdam UMC Location AMC, Amsterdam, The Netherlands; Tissue Function & Regeneration, Amsterdam Movement Sciences, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands
| | - Paul A J Krijnen
- Department of Pathology, Amsterdam UMC Location AMC, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, Amsterdam, The Netherlands
| | - Esther Middelkoop
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands; Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, The Netherlands; Tissue Function & Regeneration, Amsterdam Movement Sciences, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands; Burn Centre, Red Cross Hospital, Beverwijk, The Netherlands
| | - Hans W M Niessen
- Department of Pathology, Amsterdam UMC Location AMC, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, Amsterdam, The Netherlands; Department of Cardio-thoracic Surgery, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands
| | - Bouke K H L Boekema
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands; Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, The Netherlands.
| |
Collapse
|
5
|
Guo Q, Zhang G, Ren J, Li J, Wang Z, Ba H, Ye Z, Wang Y, Zheng J, Li C. Systemic factors associated with antler growth promote complete wound healing. NPJ Regen Med 2025; 10:4. [PMID: 39833274 PMCID: PMC11756403 DOI: 10.1038/s41536-025-00391-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Deer antlers are the only mammalian appendages that can fully regenerate from periosteum of pedicles (PP). This regeneration process starts from regenerative healing of wounds. Removal of PP abolishes antler regeneration, however, the regenerative cutaneous wound healing proceeds, indicating that some factors in the circulation contribute to this healing. In this study, we produced a wound in the scalp of deer either in antler regeneration period (ARP) (n = 3) or in non-ARP (n = 3). Results showed full regeneration took place only when the wound was created during ARP. Interestingly, topical application of systemic factors from ARP (n = 9) promoted regenerative wound healing in rats. Comparative proteomics analysis (n = 3) revealed that PRG4 and IGF-1 were high during ARP, and topical application of PRG4 + IGF-1 promoted restoration in rat FTE wounds. We believe that, ultimately, incorporating systemic factors into advanced wound care modalities could offer new opportunities for regenerative healing in the clinical setting.
Collapse
Affiliation(s)
- Qianqian Guo
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China
| | - Guokun Zhang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
| | - Jing Ren
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
| | - Jiping Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
| | - Zhen Wang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
| | - Hengxing Ba
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
| | - Zihao Ye
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Junjun Zheng
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin, China.
| | - Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China.
| |
Collapse
|
6
|
Zhao F, Wang L, Zhang Y, Tang S, Ji P, Xiang X, Pang X. MiR-494-3p regulates skin fibroblast activities by mediating fibromodulin production. J Cell Physiol 2024; 239:e31404. [PMID: 39129212 DOI: 10.1002/jcp.31404] [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: 02/07/2024] [Revised: 07/04/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
Skin wound healing is a well-coordinated process in which various cells and factors participate, during which fibroblast exhibits a critical role by exerting its multiple activities, including proliferation, migration, invasion, and differentiation. Previous studies have identified that fibromodulin (FMOD) could enhance dermal wound healing by promoting skin fibroblast activities, but little is known about its upstream regulator. We occasionally found that FMOD expression was downregulated in skin fibroblast by transforming growth factor-β1 treatment. It was hypothesized that microRNAs (miRNA) in skin fibroblast could downregulate FMOD production and blocking them would increase FMOD expression, as well as promote skin wound healing. Here, by utilizing combined analysis of miRNA microarray from the Gene Expression Omnibus database and miRNA targets prediction, we successfully identified a miRNA, termed miR-494-3p, could regulate FMOD production in human skin fibroblast (BJ fibroblast). The functional analysis revealed that miR-494-3p mimics could inhibit BJ fibroblast migration and invasion but not proliferation and differentiation, while miR-494-3p inhibition markedly promotes migration, invasion, and differentiation of BJ fibroblast. Moreover, we established FMOD overexpression (OE) and knockout BJ fibroblast. We found that FMOD OE could rescue the inhibitory effects of miR-494-3p mimics on the migration and invasion of BJ fibroblast. In contrast, the miR-494-3p inhibitor transfection could not enhance migration, invasion, and differentiation of FMOD KO BJ fibroblast. Together, our results suggest that miR-494-3p may be a potential target for skin wound management via regulating FMOD production.
Collapse
Affiliation(s)
- Feng Zhao
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Linshu Wang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxin Zhang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Siqi Tang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Ji
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Xuerong Xiang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoxiao Pang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Yu Y, Xiao H, Tang G, Wang H, Shen J, Sun Y, Wang S, Kong W, Chai Y, Liu X, Wang X, Wen G. Biomimetic hydrogel derived from decellularized dermal matrix facilitates skin wounds healing. Mater Today Bio 2023; 21:100725. [PMID: 37483381 PMCID: PMC10359665 DOI: 10.1016/j.mtbio.2023.100725] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/15/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023] Open
Abstract
Cutaneous wound healing affecting millions of people worldwide represents an unsolvable clinical issue that is frequently challenged by scar formation with dramatical pain, impaired mobility and disfigurement. Herein, we prepared a kind of light-sensitive decellularized dermal extracellular matrix-derived hydrogel with fast gelling performance, biomimetic porous microstructure and abundant bioactive functions. On account of its excellent cell biocompatibility, this ECM-derived hydrogel could induce a marked cellular infiltration and enhance the tube formation of HUVECs. In vivo experiments based upon excisional wound splinting model showed that the hydrogel prominently imparted skin wound healing, as evidenced by notably increased skin appendages and well-organized collagen expression, coupled with significantly enhanced angiogenesis. Moreover, the skin regeneration mediated by this bioactive hydrogel was promoted by an accelerated M1-to-M2 macrophage phenotype transition. Consequently, the decellularized dermal matrix-derived bioactive hydrogel orchestrates the entire skin healing microenvironment to promote wound healing and will be of high value in treatment of cutaneous wound healing. As such, this biomimetic ddECMMA hydrogel provides a promising versatile opinion for the clinical translation.
Collapse
Affiliation(s)
- Yaling Yu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Huimin Xiao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Guoke Tang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, 200080, China
| | - Hongshu Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Junjie Shen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yi Sun
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Shuaiqun Wang
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, China
| | - Wei Kong
- College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, China
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xuanzhe Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Gen Wen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
8
|
Suzdaltseva Y, Kiselev SL. Mesodermal Derivatives of Pluripotent Stem Cells Route to Scarless Healing. Int J Mol Sci 2023; 24:11945. [PMID: 37569321 PMCID: PMC10418846 DOI: 10.3390/ijms241511945] [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: 06/16/2023] [Revised: 07/07/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Scar formation during normal tissue regeneration in adults may result in noticeable cosmetic and functional defects and have a significant impact on the quality of life. In contrast, fetal tissues in the mid-gestation period are known to be capable of complete regeneration with the restitution of the initial architecture, organization, and functional activity. Successful treatments that are targeted to minimize scarring can be realized by understanding the cellular and molecular mechanisms of fetal wound regeneration. However, such experiments are limited by the inaccessibility of fetal material for comparable studies. For this reason, the molecular mechanisms of fetal regeneration remain unknown. Mesenchymal stromal cells (MSCs) are central to tissue repair because the molecules they secrete are involved in the regulation of inflammation, angiogenesis, and remodeling of the extracellular matrix. The mesodermal differentiation of human pluripotent stem cells (hPSCs) recapitulates the sequential steps of embryogenesis in vitro and provides the opportunity to generate the isogenic cell models of MSCs corresponding to different stages of human development. Further investigation of the functional activity of cells from stromal differon in a pro-inflammatory microenvironment will procure the molecular tools to better understand the fundamental mechanisms of fetal tissue regeneration. Herein, we review recent advances in the generation of clonal precursors of primitive mesoderm cells and MSCs from hPSCs and discuss critical factors that determine the functional activity of MSCs-like cells in a pro-inflammatory microenvironment in order to identify therapeutic targets for minimizing scarring.
Collapse
Affiliation(s)
- Yulia Suzdaltseva
- Department of Epigenetics, Vavilov Institute of General Genetics of the Russian Academy of Sciences, 119333 Moscow, Russia;
| | | |
Collapse
|
9
|
Zhao F, Bai Y, Xiang X, Pang X. The role of fibromodulin in inflammatory responses and diseases associated with inflammation. Front Immunol 2023; 14:1191787. [PMID: 37483637 PMCID: PMC10360182 DOI: 10.3389/fimmu.2023.1191787] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/23/2023] [Indexed: 07/25/2023] Open
Abstract
Inflammation is an immune response that the host organism eliminates threats from foreign objects or endogenous signals. It plays a key role in the progression, prognosis as well as therapy of diseases. Chronic inflammatory diseases have been regarded as the main cause of death worldwide at present, which greatly affect a vast number of individuals, producing economic and social burdens. Thus, developing drugs targeting inflammation has become necessary and attractive in the world. Currently, accumulating evidence suggests that small leucine-rich proteoglycans (SLRPs) exhibit essential roles in various inflammatory responses by acting as an anti-inflammatory or pro-inflammatory role in different scenarios of diseases. Of particular interest was a well-studied member, termed fibromodulin (FMOD), which has been largely explored in the role of inflammatory responses in inflammatory-related diseases. In this review, particular focus is given to the role of FMOD in inflammatory response including the relationship of FMOD with the complement system and immune cells, as well as the role of FMOD in the diseases associated with inflammation, such as skin wounding healing, osteoarthritis (OA), tendinopathy, atherosclerosis, and heart failure (HF). By conducting this review, we intend to gain insight into the role of FMOD in inflammation, which may open the way for the development of new anti-inflammation drugs in the scenarios of different inflammatory-related diseases.
Collapse
Affiliation(s)
- Feng Zhao
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Bai
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Xuerong Xiang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoxiao Pang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Gesteira TF, Verma S, Coulson-Thomas VJ. Small leucine rich proteoglycans: Biology, function and their therapeutic potential in the ocular surface. Ocul Surf 2023; 29:521-536. [PMID: 37355022 PMCID: PMC11092928 DOI: 10.1016/j.jtos.2023.06.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/13/2023] [Accepted: 06/21/2023] [Indexed: 06/26/2023]
Abstract
Small leucine rich proteoglycans (SLRPs) are the largest family of proteoglycans, with 18 members that are subdivided into five classes. SLRPs are small in size and can be present in tissues as glycosylated and non-glycosylated proteins, and the most studied SLRPs include decorin, biglycan, lumican, keratocan and fibromodulin. SLRPs specifically bind to collagen fibrils, regulating collagen fibrillogenesis and the biomechanical properties of tissues, and are expressed at particularly high levels in fibrous tissues, such as the cornea. However, SLRPs are also very active components of the ECM, interacting with numerous growth factors, cytokines and cell surface receptors. Therefore, SLRPs regulate major cellular processes and have a central role in major fundamental biological processes, such as maintaining corneal homeostasis and transparency and regulating corneal wound healing. Over the years, mutations and/or altered expression of SLRPs have been associated with various corneal diseases, such as congenital stromal corneal dystrophy and cornea plana. Recently, there has been great interest in harnessing the various functions of SLRPs for therapeutic purposes. In this comprehensive review, we describe the structural features and the related functions of SLRPs, and how these affect the therapeutic potential of SLRPs, with special emphasis on the use of SLRPs for treating ocular surface pathologies.
Collapse
Affiliation(s)
| | - Sudhir Verma
- College of Optometry, University of Houston, USA; Department of Zoology, Deen Dayal Upadhyaya College, University of Delhi, Delhi, India
| | | |
Collapse
|
11
|
Castillo V, Díaz-Astudillo P, Corrales-Orovio R, San Martín S, Egaña JT. Comprehensive Characterization of Tissues Derived from Animals at Different Regenerative Stages: A Comparative Analysis between Fetal and Adult Mouse Skin. Cells 2023; 12:cells12091215. [PMID: 37174615 PMCID: PMC10177150 DOI: 10.3390/cells12091215] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
Tissue regeneration capabilities vary significantly throughout an organism's lifespan. For example, mammals can fully regenerate until they reach specific developmental stages, after which they can only repair the tissue without restoring its original architecture and function. The high regenerative potential of fetal stages has been attributed to various factors, such as stem cells, the immune system, specific growth factors, and the presence of extracellular matrix molecules upon damage. To better understand the local differences between regenerative and reparative tissues, we conducted a comparative analysis of skin derived from mice at regenerative and reparative stages. Our findings show that both types of skin differ in their molecular composition, structure, and functionality. We observed a significant increase in cellular density, nucleic acid content, neutral lipid density, Collagen III, and glycosaminoglycans in regenerative skin compared with reparative skin. Additionally, regenerative skin had significantly higher porosity, metabolic activity, water absorption capacity, and elasticity than reparative skin. Finally, our results also revealed significant differences in lipid distribution, extracellular matrix pore size, and proteoglycans between the two groups. This study provides comprehensive data on the molecular and structural clues that enable full tissue regeneration in fetal stages, which could aid in developing new biomaterials and strategies for tissue engineering and regeneration.
Collapse
Affiliation(s)
- Valentina Castillo
- Institute for Biological and Medical Engineering, Schools of Engineering, Biological Sciences, and Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Pamela Díaz-Astudillo
- Biomedical Research Center, School of Medicine, Universidad de Valparaiso, Valparaiso 2540064, Chile
| | - Rocío Corrales-Orovio
- Institute for Biological and Medical Engineering, Schools of Engineering, Biological Sciences, and Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Division of Hand, Plastic and Aesthetic Surgery, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Sebastián San Martín
- Biomedical Research Center, School of Medicine, Universidad de Valparaiso, Valparaiso 2540064, Chile
| | - José Tomás Egaña
- Institute for Biological and Medical Engineering, Schools of Engineering, Biological Sciences, and Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| |
Collapse
|
12
|
Abstract
Fibromodulin (FMOD) is an archetypal member of the class II small leucine-rich proteoglycan family. By directly binding to extracellular matrix structural components, such as collagen and lysyl oxidase, FMOD regulates collagen cross-linking, packing, assembly, and fibril architecture via a multivalent interaction. Meanwhile, as a pluripotent molecule, FMOD acts as a ligand of various cytokines and growth factors, especially those belonging to the transforming growth factor (TGF) β superfamily, by interacting with the corresponding signaling molecules involved in cell adhesion, spreading, proliferation, migration, invasion, differentiation, and metastasis. Consequently, FMOD exhibits promigratory, proangiogenic, anti-inflammatory, and antifibrogenic properties and plays essential roles in cell fate determination and maturation, progenitor cell recruitment, and tissue regeneration. The multifunctional nature of FMOD thus enables it to be a promising therapeutic agent for a broad repertoire of diseases, including but not limited to arthritis, temporomandibular joint disorders, caries, and fibrotic diseases among different organs, as well as to be a regenerative medicine candidate for skin, muscle, and tendon injuries. Moreover, FMOD is also considered a marker for tumor diagnosis and prognosis prediction and a potential target for cancer treatment. Furthermore, FMOD itself is sufficient to reprogram somatic cells into a multipotent state, creating a safe and efficient cell source for various tissue reconstructions and thus opening a new avenue for regenerative medicine. This review focuses on the recent preclinical efforts bringing FMOD research and therapies to the forefront. In addition, a contemporary understanding of the mechanism underlying FMOD's function, particularly its interaction with TGFβ superfamily members, is also discussed at the molecular level to aid the discovery of novel FMOD-based treatments.
Collapse
Affiliation(s)
- Z. Zheng
- David Geffen School of Medicine,
University of California, Los Angeles, CA, USA
- School of Dentistry, University of
California, Los Angeles, CA, USA
| | - H.S. Granado
- Department of Orthodontics, School of
Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - C. Li
- Department of Orthodontics, School of
Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
13
|
Xu X, Zhang Y, Ha P, Chen Y, Li C, Yen E, Bai Y, Chen R, Wu BM, Da Lio A, Ting K, Soo C, Zheng Z. A novel injectable fibromodulin-releasing granular hydrogel for tendon healing and functional recovery. Bioeng Transl Med 2023; 8:e10355. [PMID: 36684085 PMCID: PMC9842059 DOI: 10.1002/btm2.10355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 01/25/2023] Open
Abstract
A crucial component of the musculoskeletal system, the tendon is one of the most commonly injured tissues in the body. In severe cases, the ruptured tendon leads to permanent dysfunction. Although many efforts have been devoted to seeking a safe and efficient treatment for enhancing tendon healing, currently existing treatments have not yet achieved a major clinical improvement. Here, an injectable granular hyaluronic acid (gHA)-hydrogel is engineered to deliver fibromodulin (FMOD)-a bioactive extracellular matrix (ECM) that enhances tenocyte mobility and optimizes the surrounding ECM assembly for tendon healing. The FMOD-releasing granular HA (FMOD/gHA)-hydrogel exhibits unique characteristics that are desired for both patients and health providers, such as permitting a microinvasive application and displaying a burst-to-sustained two-phase release of FMOD, which leads to a prompt FMOD delivery followed by a constant dose-maintaining period. Importantly, the generated FMOD-releasing granular HA hydrogel significantly augmented tendon-healing in a fully-ruptured rat's Achilles tendon model histologically, mechanically, and functionally. Particularly, the breaking strength of the wounded tendon and the gait performance of treated rats returns to the same normal level as the healthy controls. In summary, a novel effective FMOD/gHA-hydrogel is developed in response to the urgent demand for promoting tendon healing.
Collapse
Affiliation(s)
- Xue Xu
- Department of Oral and Maxillofacial Plastic and Traumatic SurgeryBeijing Stomatological Hospital of Capital Medical UniversityBeijingChina
- Division of Plastic and Reconstructive SurgeryDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
- Division of Growth and DevelopmentSchool of Dentistry, University of CaliforniaLos AngelesCaliforniaUSA
| | - Yulong Zhang
- School of DentistryUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Pin Ha
- Division of Plastic and Reconstructive SurgeryDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
- Division of Growth and DevelopmentSchool of Dentistry, University of CaliforniaLos AngelesCaliforniaUSA
| | - Yao Chen
- School of DentistryUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Chenshuang Li
- Department of OrthodonticsSchool of Dental Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Emily Yen
- Arcadia High SchoolArcadiaCaliforniaUSA
| | - Yuxing Bai
- Department of OrthodonticsBeijing Stomatological Hospital of Capital Medical UniversityBeijingChina
| | - Renji Chen
- Department of Oral and Maxillofacial Plastic and Traumatic SurgeryBeijing Stomatological Hospital of Capital Medical UniversityBeijingChina
| | - Benjamin M. Wu
- School of DentistryUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Andrew Da Lio
- Division of Plastic and Reconstructive SurgeryDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
| | - Kang Ting
- Forsyth Research InstituteHarvard UniversityCambridgeMassachusettsUSA
- Samueli School of EngineeringUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Orthopaedic SurgeryThe Orthopaedic Hospital Research Center, University of CaliforniaLos AngelesCaliforniaUSA
| | - Zhong Zheng
- Division of Plastic and Reconstructive SurgeryDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
- Division of Growth and DevelopmentSchool of Dentistry, University of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
14
|
Cao P, Wang YW, Guan H, Yang YS, Li SH, Chen Y, Zhu C, Wan Y, Ren LY, Yao M. [Effects of mechanical tension on the formation of hypertrophic scars in rabbit ears and transforming growth factor-β 1/Smad signaling pathway]. ZHONGHUA SHAO SHANG YU CHUANG MIAN XIU FU ZA ZHI 2022; 38:1162-1169. [PMID: 36594147 DOI: 10.3760/cma.j.cn501120-20211213-00412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Objective: To explore the effects of mechanical tension on the formation of hypertrophic scars in rabbit ears and transforming growth factor-β1 (TGF-β1)/Smad signaling pathway. Methods: The experimental research method was adopted. Six New Zealand white rabbits, male or female, aged 3-5 months were used and 5 full-thickness skin defect wounds were made on the ventral surface of each rabbit ear. The appearance of all rabbit ear wounds was observed on post surgery day (PSD) 0 (immediately), 7, 14, 21, and 28. On PSD 28, the scar formation rate was calculated. Three mature scars in the left ear of each rabbit were included in tension group and the arch was continuously expanded with a spiral expander. Three mature scars in the right ear of each rabbit were included in sham tension group and only the spiral expander was sutured without expansion. There were 18 scars in each group. After mechanical tension treatment (hereinafter referred to as treatment) for 40 days, the color and texture of scar tissue in the two groups were observed. On treatment day 40, the scar elevation index (SEI) was observed and calculated; the histology was observed after hematoxylin eosin staining, and the collagen morphology was observed after Masson staining; mRNA expressions of TGF-β1, Smad3, collagen Ⅰ, collagen Ⅲ, and α-smooth muscle actin (α-SMA) in scar tissue were detected by real-time fluorescence quantitative reverse transcription polymerase chain reaction; and the protein expressions of TGF-β1, collagen Ⅰ, collagen Ⅲ, and α-SMA, and phosphorylation level of Smad3 in scar tissue were detected by Western blotting. The number of samples of each group in the experiments was 3. Data were statistically analyzed with independent sample t test. Results: On PSD 0, 5 fresh wounds were formed on all the rabbit ears; on PSD 7, the wounds were scabbed; on PSD 14, most of the wounds were epithelialized; on PSD 21, all the wounds were epithelialized; on PSD 28, obvious hypertrophic scars were formed. The scar formation rate was 75% (45/60) on PSD 28. On treatment day 40, the scar tissue of rabbit ears in tension group was more prominent than that in sham tension group, the scar tissue was harder and the color was more ruddy; the SEI of the scar tissue of rabbit ears in tension group (2.02±0.08) was significantly higher than 1.70±0.08 in sham tension group (t=5.07, P<0.01). On treatment day 40, compared with those in sham tension group, the stratum corneum of scar tissue became thicker, and a large number of new capillaries, inflammatory cells, and fibroblasts were observed in the dermis, and collagen was more disordered, with nodular or swirling distribution in the scar tissue of rabbit ears in tension group. On treatment day 40, the mRNA expressions of TGF-β1, Smad3, collagen Ⅰ, collagen Ⅲ, and α-SMA in the scar tissue of rabbit ears in tension group were respectively 1.81±0.25, 5.71±0.82, 7.86±0.56, 4.35±0.28, and 5.89±0.47, which were significantly higher than 1.00±0.08, 1.00±0.12, 1.00±0.13, 1.00±0.14, and 1.00±0.14 in sham tension group (with t values of 5.36, 9.82, 20.60, 18.26, and 17.13, respectively, all P<0.01); the protein expressions of TGF-β1, collagen Ⅰ, collagen Ⅲ, and α-SMA, and phosphorylation level of Smad3 in the scar tissue of rabbit ears in tension group were respectively 0.865±0.050, 0.895±0.042, 0.972±0.027, 1.012±0.057, and 0.968±0.087, which were significantly higher than 0.657±0.050, 0.271±0.029, 0.631±0.027, 0.418±0.023, and 0.511±0.035 in sham tension group (with t values of 5.08, 21.27, 15.55, 16.70, and 8.40, respectively, all P<0.01). Conclusions: Mechanical tension can inhibit the regression of hypertrophic scars in rabbit ears through stimulating the hyperplasia of scars, inhibiting the normal arrangement of dermal collagen fibers, and intensifying the deposition of collagen fibers, and the mechanism may be related to the activation of TGF-β1/Smad signaling pathway by mechanical tension.
Collapse
Affiliation(s)
- P Cao
- Department of Burns and Plastic Surgery, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Y W Wang
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - H Guan
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - Y S Yang
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - S H Li
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - Y Chen
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - C Zhu
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - Y Wan
- Department of Burns and Plastic Surgery, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - L Y Ren
- Department of Burns and Plastic Surgery, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - M Yao
- Department of Burns and Plastic Surgery, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
15
|
Eremenko E, Ding J, Kwan P, Tredget EE. The Biology of Extracellular Matrix Proteins in Hypertrophic Scarring. Adv Wound Care (New Rochelle) 2022; 11:234-254. [PMID: 33913776 DOI: 10.1089/wound.2020.1257] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Significance: Hypertrophic scars (HTS) are a fibroproliferative disorder that occur following deep dermal injury and affect up to 72% of burn patients. These scars result in discomfort, impaired mobility, disruption of normal function and cosmesis, and significant psychological distress. Currently, there are no satisfactory methods to treat or prevent HTS, as the cellular and molecular mechanisms are complex and incompletely understood. This review summarizes the biology of proteins in the dermal extracellular matrix (ECM), which are involved in wound healing and hypertrophic scarring. Recent Advances: New basic research continues toward understanding the diversity of cellular and molecular mechanisms of normal wound healing and hypertrophic scarring. Broadening the understanding of these mechanisms creates insight into novel methods for preventing and treating HTS. Critical Issues: Although there is an abundance of research conducted on collagen in the ECM and its relationship to HTS, there is a significant gap in understanding the role of proteoglycans and their specific isoforms in dermal fibrosis. Future Directions: Exploring the biological roles of ECM proteins and their unique isoforms in HTS, mature scars, and normal skin will further the understanding of abnormal wound healing and create a more robust understanding of what constitutes dermal fibrosis. Research into the biological roles of ECM protein isoforms and their regulation during wound healing warrants a more extensive investigation to identify their distinct biological functions in cellular processes and outcomes.
Collapse
Affiliation(s)
- Elizabeth Eremenko
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
| | - Jie Ding
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
| | - Peter Kwan
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
- Division of Plastic Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| | - Edward E. Tredget
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
- Division of Plastic Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| |
Collapse
|
16
|
Singer AJ. Healing Mechanisms in Cutaneous Wounds: Tipping the Balance. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:1151-1167. [PMID: 34915757 PMCID: PMC9587785 DOI: 10.1089/ten.teb.2021.0114] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute and chronic cutaneous wounds pose a significant health and economic burden. Cutaneous wound healing is a complex process that occurs in four distinct, yet overlapping, highly coordinated stages: hemostasis, inflammation, proliferation, and remodeling. Postnatal wound healing is reparative, which can lead to the formation of scar tissue. Regenerative wound healing occurs during fetal development and in restricted postnatal tissues. This process can restore the wound to an uninjured state by producing new skin cells from stem cell reservoirs, resulting in healing with minimal or no scarring. Focusing on the pathophysiology of acute burn wounds, this review highlights reparative and regenerative healing mechanisms (including the role of cells, signaling molecules, and the extracellular matrix) and discusses how components of regenerative healing are being used to drive the development of novel approaches and therapeutics aimed at improving clinical outcomes. Important components of regenerative healing, such as stem cells, growth factors, and decellularized dermal matrices, are all being evaluated to recapitulate more closely the natural regenerative healing process.
Collapse
Affiliation(s)
- Adam J Singer
- Department of Emergency Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
17
|
Bioflavonoid Galangin Suppresses Hypertrophic Scar Formation by the TGF- β/Smad Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2444839. [PMID: 34518767 PMCID: PMC8434902 DOI: 10.1155/2021/2444839] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/10/2021] [Accepted: 08/20/2021] [Indexed: 11/28/2022]
Abstract
Background Hypertrophic scar (HS) is a benign fibroproliferative skin disease resulting from an aberrant wound healing process and can cause aesthetic and functional damage to patients. Currently, there is no ideal treatment to treat this disease. Galangin, a natural active bioflavonoid compound, is suggested to inhibit fibrosis and proliferation in certain cells. Methods In this study, we found Galangin could attenuate abnormal scar formation in an HS rabbit ear model. Additionally, the HE staining shows Galangin reduced scar elevation index (SEI) and Masson's trichrome staining changed collagen deposition. Results The expressions of type I collagen, type III collagen, and TGF-β1 were much lower under a proper dose of Galangin treatment, and Smad7 expression was also enhanced, which are examined by real-time PCR, immunohistochemistry, and western blot. Conclusion Our data indicated that Galangin can alleviate dermal scarring via the TGF-β/Smad signaling pathway probably by upregulating Smad 7 expression and, thus, suppressing the expression of type I and type III collagens and TGF-β1.
Collapse
|
18
|
Li C, Mills Z, Zheng Z. Novel cell sources for bone regeneration. MedComm (Beijing) 2021; 2:145-174. [PMID: 34766140 PMCID: PMC8491221 DOI: 10.1002/mco2.51] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 01/09/2023] Open
Abstract
A plethora of both acute and chronic conditions, including traumatic, degenerative, malignant, or congenital disorders, commonly induce bone disorders often associated with severe persisting pain and limited mobility. Over 1 million surgical procedures involving bone excision, bone grafting, and fracture repair are performed each year in the U.S. alone, resulting in immense levels of public health challenges and corresponding financial burdens. Unfortunately, the innate self-healing capacity of bone is often inadequate for larger defects over a critical size. Moreover, as direct transplantation of committed osteoblasts is hindered by deficient cell availability, limited cell spreading, and poor survivability, an urgent need for novel cell sources for bone regeneration is concurrent. Thanks to the development in stem cell biology and cell reprogramming technology, many multipotent and pluripotent cells that manifest promising osteogenic potential are considered the regenerative remedy for bone defects. Considering these cells' investigation is still in its relative infancy, each of them offers their own particular challenges that must be conquered before the large-scale clinical application.
Collapse
Affiliation(s)
- Chenshuang Li
- Department of Orthodontics, School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Zane Mills
- College of DentistryUniversity of OklahomaOklahoma CityOklahomaUSA
| | - Zhong Zheng
- Division of Growth and Development, School of DentistryUniversity of CaliforniaLos AngelesCaliforniaUSA
- Department of Surgery, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
19
|
Angioni MM, Floris A, Cangemi I, Congia M, Chessa E, Orrù S, Piga M, Cauli A. Gene Expression Profiling of Monozygotic Twins Affected by Psoriatic Arthritis. Open Access Rheumatol 2021; 13:23-29. [PMID: 33692638 PMCID: PMC7939499 DOI: 10.2147/oarrr.s291391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/23/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction Psoriatic Arthritis (PsA) is a multifactorial disease, where the relative burden of genetic, epigenetic and environmental factors in clinical course and damage accrual is not yet definitively clarified. In clinical practice, there is a real need for useful candidate biomarkers in PsA diagnosis and disease progression, by exploring its underlying transcriptomic and epigenomic mechanisms. This work aims to profile the transcriptome in monozygotic (MZ) twins with psoriatic arthritis (PsA) highly concordant for clinical presentation, but discordant for the radiographic outcomes’ severity. Methods We describe i) the clinical case of two MZ twins; ii) their comparative gene expression profiling (HTA 2.0 Affymetrix) and iii) signal pathways and pathophysiological processes in which differentially expressed genes are involved (in silico analysis by the IPA software, QIAGEN). Results One hundred sixty-three transcripts and 36 coding genes (28 up and 8 down) were differentially expressed between twins, and in the brother with the most erosive form, the transcriptomic profiling highlights the overexpression of genes known to be involved in immunomodulatory processes and on a broad spectrum of PsA manifestations. Discussion Twins’ clinical cases are still a gold mine in medical research: twin brothers are ideal experimental models in estimating the relative importance of genetic versus nongenetic components as determinants of complex phenotypes, non-Mendelian and multifactorial diseases as PsA.
Collapse
Affiliation(s)
- Maria Maddalena Angioni
- Rheumatology Unit, Department of Medical Sciences and Public Health, University of Cagliari, University Clinic AOU, Cagliari, 09042, Italy
| | - Alberto Floris
- Rheumatology Unit, Department of Medical Sciences and Public Health, University of Cagliari, University Clinic AOU, Cagliari, 09042, Italy
| | - Ignazio Cangemi
- Rheumatology Unit, Department of Medical Sciences and Public Health, University of Cagliari, University Clinic AOU, Cagliari, 09042, Italy
| | - Mattia Congia
- Rheumatology Unit, Department of Medical Sciences and Public Health, University of Cagliari, University Clinic AOU, Cagliari, 09042, Italy
| | - Elisabetta Chessa
- Rheumatology Unit, Department of Medical Sciences and Public Health, University of Cagliari, University Clinic AOU, Cagliari, 09042, Italy
| | - Sandro Orrù
- Medical Genetics, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, 09042, Italy
| | - Matteo Piga
- Rheumatology Unit, Department of Medical Sciences and Public Health, University of Cagliari, University Clinic AOU, Cagliari, 09042, Italy
| | - Alberto Cauli
- Rheumatology Unit, Department of Medical Sciences and Public Health, University of Cagliari, University Clinic AOU, Cagliari, 09042, Italy
| |
Collapse
|
20
|
Zeng-Brouwers J, Pandey S, Trebicka J, Wygrecka M, Schaefer L. Communications via the Small Leucine-rich Proteoglycans: Molecular Specificity in Inflammation and Autoimmune Diseases. J Histochem Cytochem 2020; 68:887-906. [PMID: 32623933 PMCID: PMC7708667 DOI: 10.1369/0022155420930303] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/06/2020] [Indexed: 12/15/2022] Open
Abstract
Inflammation is a highly regulated biological response of the immune system that is triggered by assaulting pathogens or endogenous alarmins. It is now well established that some soluble extracellular matrix constituents, such as small leucine-rich proteoglycans (SLRPs), can act as danger signals and trigger aseptic inflammation by interacting with innate immune receptors. SLRP inflammatory signaling cascade goes far beyond its canonical function. By choosing specific innate immune receptors, coreceptors, and adaptor molecules, SLRPs promote a switch between pro- and anti-inflammatory signaling, thereby determining disease resolution or chronification. Moreover, by orchestrating signaling through various receptors, SLRPs fine-tune inflammation and, despite their structural homology, regulate inflammatory processes in a molecule-specific manner. Hence, the overarching theme of this review is to highlight the molecular and functional specificity of biglycan-, decorin-, lumican-, and fibromodulin-mediated signaling in inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Jinyang Zeng-Brouwers
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Sony Pandey
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Jonel Trebicka
- Translational Hepatology, Department of Internal Medicine I, University Clinic Frankfurt, Frankfurt, Germany
| | - Malgorzata Wygrecka
- Department of Biochemistry, Faculty of Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany
- German Center for Lung Research, Giessen, Germany
| | - Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
21
|
Wilgus TA. Inflammation as an orchestrator of cutaneous scar formation: a review of the literature. PLASTIC AND AESTHETIC RESEARCH 2020; 7:54. [PMID: 33123623 PMCID: PMC7592345 DOI: 10.20517/2347-9264.2020.150] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inflammation is a key phase in the cutaneous wound repair process. The activation of inflammatory cells is critical for preventing infection in contaminated wounds and results in the release of an array of mediators, some of which stimulate the activity of keratinocytes, endothelial cells, and fibroblasts to aid in the repair process. However, there is an abundance of data suggesting that the strength of the inflammatory response early in the healing process correlates directly with the amount of scar tissue that will eventually form. This review will summarize the literature related to inflammation and cutaneous scar formation, highlight recent discoveries, and discuss potential treatment modalities that target inflammation to minimize scarring.
Collapse
Affiliation(s)
- Traci A Wilgus
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
22
|
Yin JL, Wu Y, Yuan ZW, Gao XH, Chen HD. Advances in scarless foetal wound healing and prospects for scar reduction in adults. Cell Prolif 2020; 53:e12916. [PMID: 33058377 PMCID: PMC7653265 DOI: 10.1111/cpr.12916] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/25/2020] [Accepted: 09/06/2020] [Indexed: 02/06/2023] Open
Abstract
Healing after mammalian skin injury involves the interaction between numerous cellular constituents and regulatory factors, which together form three overlapping phases: an inflammatory response, a proliferation phase and a remodelling phase. Any slight variation in these three stages can substantially alter the healing process and resultant production of scars. Of particular significance are the mechanisms responsible for the scar‐free phenomenon observed in the foetus. Uncovering such mechanisms would offer great expectations in the treatment of scars and therefore represents an important area of investigation. In this review, we provide a comprehensive summary of studies on injury‐induced skin regeneration within the foetus. The information contained in these studies provides an opportunity for new insights into the treatment of clinical scars based on the cellular and molecular processes involved.
Collapse
Affiliation(s)
- Jia-Li Yin
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning, China.,National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yan Wu
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning, China.,National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zheng-Wei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China
| | - Xing-Hua Gao
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning, China.,National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hong-Duo Chen
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning, China.,National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
23
|
Monavarian M, Kader S, Moeinzadeh S, Jabbari E. Regenerative Scar-Free Skin Wound Healing. TISSUE ENGINEERING PART B-REVIEWS 2020; 25:294-311. [PMID: 30938269 DOI: 10.1089/ten.teb.2018.0350] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
IMPACT STATEMENT Millions of people every year develop scars in response to skin injuries after surgery, trauma, or burns with significant undesired physical and psychological effects. This review provides an update on engineering strategies for scar-free wound healing and discusses the role of different cell types, growth factors, cytokines, and extracellular components in regenerative wound healing. The use of pro-regenerative matrices combined with engineered cells with less intrinsic potential for fibrogenesis is a promising strategy for achieving scar-free skin tissue regeneration.
Collapse
Affiliation(s)
- Mehri Monavarian
- 1Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, South Carolina
| | - Safaa Kader
- 1Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, South Carolina.,2Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina
| | - Seyedsina Moeinzadeh
- 1Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, South Carolina
| | - Esmaiel Jabbari
- 1Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
24
|
Jensen MM, Karring H. The origins and developments of sulfation-prone tyrosine-rich and acidic N- and C-terminal extensions of class ll and lll small leucine-rich repeat proteins shed light on connective tissue evolution in vertebrates. BMC Evol Biol 2020; 20:73. [PMID: 32576155 PMCID: PMC7310474 DOI: 10.1186/s12862-020-01634-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/27/2020] [Indexed: 02/06/2023] Open
Abstract
Background Small leucine-rich repeat protein (SLRP) family members contain conserved leucine-rich repeat motifs flanked by highly variable N- and C-terminal regions. Most class II and III SLRPs have tyrosine-rich N-terminal regions and some of these are sulfated. However, the evolutionary origin and conservation of the tyrosine-rich and acidic terminal regions remain undetermined. In this study, we present the most comprehensive multiple sequence alignment (MSA) analyses of all eight class II and III SLRPs to date. Based on the level of conservation of tyrosine residues and adjacent sequences, we predict which tyrosine residues are most likely to be sulfated in the terminal regions of human class II and III SLRPs. Results Using this novel approach, we predict a total of 22 tyrosine sulfation sites in human SLRPs, of which only 8 sites had been experimentally identified in mammals. Our analyses suggest that sulfation-prone, tyrosine-rich and acidic terminal regions of the class II and III SLRPs emerged via convergent evolution at different stages of vertebrate evolution, coinciding with significant evolutionary events including the development of endochondral bones and articular cartilage, the aquatic to terrestrial transition, and the formation of an amnion. Conclusions Our study suggests that selective pressures due to changes in life conditions led to the formation of sulfotyrosine-rich and acidic terminal regions. We believe the independent emergence and evolution of sulfotyrosine-rich and acidic N- and C-terminal regions have provided each class II and III SLRP member with novel vital functions required to develop new specialized extracellular matrices and tissues in vertebrate species.
Collapse
Affiliation(s)
- Morten M Jensen
- Department of Chemical Engineering, Biotechnology and Environmental Technology, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark
| | - Henrik Karring
- Department of Chemical Engineering, Biotechnology and Environmental Technology, University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark.
| |
Collapse
|
25
|
Paredes J, Marvin JC, Vaughn B, Andarawis-Puri N. Innate tissue properties drive improved tendon healing in MRL/MpJ and harness cues that enhance behavior of canonical healing cells. FASEB J 2020; 34:8341-8356. [PMID: 32350938 DOI: 10.1096/fj.201902825rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/03/2020] [Accepted: 04/10/2020] [Indexed: 12/21/2022]
Abstract
Development of tendon therapeutics has been hindered by the lack of informative adult mammalian models of regeneration. Murphy Roth's Large (MRL/MpJ) mice exhibit improved healing following acute tendon injuries, but the driver of this regenerative healing response remains unknown. The tissue-specific attributes of this healing response, despite a shared systemic environment within the mouse, support the hypothesis of a tissue-driven mechanism for scarless healing. Our objective was to investigate the potential of MRL/MpJ tendon extracellular matrix (ECM)-derived coatings to regulate scar-mediated healing. We found that deviations in the composition of key structural proteins within MRL/MpJ vs C57Bl/6 tendons occur synergistically to mediate the improvements in structure and mechanics following a 1-mm midsubstance injury. Improvement in mechanical properties of healing MRL/MpJ vs C57Bl/6 tendons that were isolated from systemic contributions via organ culture, highlighted the innate tendon environment as the driver of scarless healing. Finally, we established that decellularized coatings derived from early-deposited MRL/MpJ tendon provisional extracellular matrix (provisional-ECM), can modulate canonical healing B6 tendon cell behavior by inducing morphological changes and increasing proliferation in vitro. This study supports that the unique compositional cues in MRL/MpJ provisional-ECM have the therapeutic capability to motivate canonically healing cells toward improved behavior; enhancing our ability to develop effective therapeutics.
Collapse
Affiliation(s)
- Juan Paredes
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Jason C Marvin
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Brenna Vaughn
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Nelly Andarawis-Puri
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA.,Hospital for Special Surgery, New York, NY, USA
| |
Collapse
|
26
|
Pang X, Dong N, Zheng Z. Small Leucine-Rich Proteoglycans in Skin Wound Healing. Front Pharmacol 2020; 10:1649. [PMID: 32063855 PMCID: PMC6997777 DOI: 10.3389/fphar.2019.01649] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Healing of cutaneous wounds is a complex and well-coordinated process requiring cooperation among multiple cells from different lineages and delicately orchestrated signaling transduction of a diversity of growth factors, cytokines, and extracellular matrix (ECM) at the wound site. Most skin wound healing in adults is imperfect, characterized by scar formation which results in significant functional and psychological sequelae. Thus, the reconstruction of the damaged skin to its original state is of concern to doctors and scientists. Beyond the traditional treatments such as corticosteroid injection and radiation therapy, several growth factors or cytokines-based anti-scarring products are being or have been tested in clinical trials to optimize skin wound healing. Unfortunately, all have been unsatisfactory to date. Currently, accumulating evidence suggests that the ECM not only functions as the structural component of the tissue but also actively modulates signal transduction and regulates cellular behaviors, and thus, ECM should be considered as an alternative target for wound management pharmacotherapy. Of particular interest are small leucine-rich proteoglycans (SLRPs), a group of the ECM, which exist in a wide range of connecting tissues, including the skin. This manuscript summarizes the most current knowledge of SLRPs regarding their spatial-temporal expression in the skin, as well as lessons learned from the genetically modified animal models simulating human skin pathologies. In this review, particular focus is given on the diverse roles of SLRP in skin wound healing, such as anti-inflammation, pro-angiogenesis, pro-migration, pro-contraction, and orchestrate transforming growth factor (TGF)β signal transduction, since cumulative investigations have indicated their therapeutic potential on reducing scar formation in cutaneous wounds. By conducting this review, we intend to gain insight into the potential application of SLRPs in cutaneous wound healing management which may pave the way for the development of a new generation of pharmaceuticals to benefit the patients suffering from skin wounds and their sequelae.
Collapse
Affiliation(s)
- Xiaoxiao Pang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nuo Dong
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Zhong Zheng
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
27
|
Li LJ, Wang MZ, Yuan TJ, Xu XH, Dad HA, Yu CL, Hou J, Peng LH. The crude ethanol extract of Periplaneta americana L. stimulates wound healing in vitro & in vivo. Chin Med 2019; 14:33. [PMID: 31548851 PMCID: PMC6751902 DOI: 10.1186/s13020-019-0259-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/11/2019] [Indexed: 12/11/2022] Open
Abstract
Periplaneta americana L. is a Traditional Chinese Medicine that has been used in clinic treatment of various diseases for a long history. However, the therapeutic potential and the underlying mechanism of Periplaneta americana L. in the skin wound therapy was not investigated comprehensively yet. This study aims to investigate the influence of the crude ethanol extract of PAL in the different wound stages including: (1) the migration and chemotaxis to skin cells in the first stage; (2) proliferation and cells cycle of skin cells in the second stage; (3) remodeling effect and secretion of growth factors, collagens in the third stage; (4) as well as the influence in the blood vessels regeneration in the late stage. The crude ethanol extract of PAL was shown to (1) promote the keratinocytes proliferation and regulate the cells cycle of fibroblasts significantly; (2) stimulate the migration of keratinocytes and fibroblasts obviously; (3) enhance the EGF and VEGF secretion both in vitro & in vivo; (4) accelerate the wound healing, collagen synthesis and angiogenesis. The crude ethanol extract of KFX was shown a promising therapeutic agent for the wound therapy with great efficacy to accelerate the wound healing with improved quality.
Collapse
Affiliation(s)
- Long-Jian Li
- 1College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058 Zhejiang People's Republic of China.,Jiaxing Lvy Geriatric Hospital, Jiaxing, Zhejiang People's Republic of China
| | - Mao-Ze Wang
- 1College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058 Zhejiang People's Republic of China
| | - Tie-Jun Yuan
- 1College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058 Zhejiang People's Republic of China
| | - Xue-Han Xu
- 1College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058 Zhejiang People's Republic of China
| | - Haseeb Anwar Dad
- 1College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058 Zhejiang People's Republic of China
| | - Chui-Liang Yu
- Zhejiangjingxin Pharmaceutical Co., Ltd, XinChang, 312500 Zhejiang People's Republic of China
| | - Jian Hou
- Zhejiangjingxin Pharmaceutical Co., Ltd, XinChang, 312500 Zhejiang People's Republic of China
| | - Li-Hua Peng
- 1College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058 Zhejiang People's Republic of China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, People's Republic of China
| |
Collapse
|
28
|
Zheng Z, Li C, Ha P, Chang GX, Yang P, Zhang X, Kim JK, Jiang W, Pang X, Berthiaume EA, Mills Z, Haveles CS, Chen E, Ting K, Soo C. CDKN2B upregulation prevents teratoma formation in multipotent fibromodulin-reprogrammed cells. J Clin Invest 2019; 129:3236-3251. [PMID: 31305260 PMCID: PMC6668700 DOI: 10.1172/jci125015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 05/21/2019] [Indexed: 12/11/2022] Open
Abstract
Tumorigenicity is a well-documented risk to overcome for pluripotent or multipotent cell applications in regenerative medicine. To address the emerging demand for safe cell sources in tissue regeneration, we established a novel, protein-based reprogramming method that does not require genome integration or oncogene activation to yield multipotent fibromodulin (FMOD)-reprogrammed (FReP) cells from dermal fibroblasts. When compared with induced pluripotent stem cells (iPSCs), FReP cells exhibited a superior capability for bone and skeletal muscle regeneration with markedly less tumorigenic risk. Moreover, we showed that the decreased tumorigenicity of FReP cells was directly related to an upregulation of cyclin-dependent kinase inhibitor 2B (CDKN2B) expression during the FMOD reprogramming process. Indeed, sustained suppression of CDKN2B resulted in tumorigenic, pluripotent FReP cells that formed teratomas in vivo that were indistinguishable from iPSC-derived teratomas. These results highlight the pivotal role of CDKN2B in cell fate determination and tumorigenic regulation and reveal an alternative pluripotent/multipotent cell reprogramming strategy that solely uses FMOD protein.
Collapse
Affiliation(s)
- Zhong Zheng
- Division of Growth and Development, School of Dentistry, and
| | - Chenshuang Li
- Division of Growth and Development, School of Dentistry, and
| | - Pin Ha
- Division of Growth and Development, School of Dentistry, and
| | - Grace X. Chang
- David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Pu Yang
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xinli Zhang
- Division of Growth and Development, School of Dentistry, and
| | - Jong Kil Kim
- Division of Growth and Development, School of Dentistry, and
| | - Wenlu Jiang
- Division of Growth and Development, School of Dentistry, and
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoxiao Pang
- Division of Growth and Development, School of Dentistry, and
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatology Hospital of Chongqing Medical University, Chongqing, China
| | | | - Zane Mills
- Department of Ecology and Evolutionary Biology, and
| | | | - Eric Chen
- Division of Growth and Development, School of Dentistry, and
| | - Kang Ting
- Division of Growth and Development, School of Dentistry, and
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA, Los Angeles, California, USA
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA, Los Angeles, California, USA
| |
Collapse
|
29
|
Korntner S, Lehner C, Gehwolf R, Wagner A, Grütz M, Kunkel N, Tempfer H, Traweger A. Limiting angiogenesis to modulate scar formation. Adv Drug Deliv Rev 2019; 146:170-189. [PMID: 29501628 DOI: 10.1016/j.addr.2018.02.010] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 01/22/2018] [Accepted: 02/26/2018] [Indexed: 02/06/2023]
Abstract
Angiogenesis, the process of new blood vessel formation from existing blood vessels, is a key aspect of virtually every repair process. During wound healing an extensive, but immature and leaky vascular plexus forms which is subsequently reduced by regression of non-functional vessels. More recent studies indicate that uncontrolled vessel growth or impaired vessel regression as a consequence of an excessive inflammatory response can impair wound healing, resulting in scarring and dysfunction. However, in order to elucidate targetable factors to promote functional tissue regeneration we need to understand the molecular and cellular underpinnings of physiological angiogenesis, ranging from induction to resolution of blood vessels. Especially for avascular tissues (e.g. cornea, tendon, ligament, cartilage, etc.), limiting rather than boosting vessel growth during wound repair potentially is beneficial to restore full tissue function and may result in favourable long-term healing outcomes.
Collapse
|
30
|
Theocharis AD, Manou D, Karamanos NK. The extracellular matrix as a multitasking player in disease. FEBS J 2019; 286:2830-2869. [PMID: 30908868 DOI: 10.1111/febs.14818] [Citation(s) in RCA: 277] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/06/2019] [Accepted: 03/22/2019] [Indexed: 12/12/2022]
Abstract
Extracellular matrices (ECMs) are highly specialized and dynamic three-dimensional (3D) scaffolds into which cells reside in tissues. ECM is composed of a variety of fibrillar components, such as collagens, fibronectin, and elastin, and non-fibrillar molecules as proteoglycans, hyaluronan, and glycoproteins including matricellular proteins. These macromolecular components are interconnected forming complex networks that actively communicate with cells through binding to cell surface receptors and/or matrix effectors. ECMs exert diverse roles, either providing tissues with structural integrity and mechanical properties essential for tissue functions or regulating cell phenotype and functions to maintain tissue homeostasis. ECM molecular composition and structure vary among tissues, and is markedly modified during normal tissue repair as well as during the progression of various diseases. Actually, abnormal ECM remodeling occurring in pathologic circumstances drives disease progression by regulating cell-matrix interactions. The importance of matrix molecules to normal tissue functions is also highlighted by mutations in matrix genes that give rise to genetic disorders with diverse clinical phenotypes. In this review, we present critical and emerging issues related to matrix assembly in tissues and the multitasking roles for ECM in diseases such as osteoarthritis, fibrosis, cancer, and genetic diseases. The mechanisms underlying the various matrix-based diseases are also discussed. Research focused on the highly dynamic 3D ECM networks will help to discover matrix-related causative abnormalities of diseases as well as novel diagnostic tools and therapeutic targets.
Collapse
Affiliation(s)
- Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiochemistry Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| |
Collapse
|
31
|
Kim T, Ahmad K, Shaikh S, Jan AT, Seo MG, Lee EJ, Choi I. Dermatopontin in Skeletal Muscle Extracellular Matrix Regulates Myogenesis. Cells 2019; 8:cells8040332. [PMID: 30970625 PMCID: PMC6523808 DOI: 10.3390/cells8040332] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/06/2019] [Accepted: 04/09/2019] [Indexed: 12/31/2022] Open
Abstract
Dermatopontin (DPT) is an extensively distributed non-collagenous component of the extracellular matrix predominantly found in the dermis of the skin, and consequently expressed in several tissues. In this study, we explored the role of DPT in myogenesis and perceived that it enhances the cell adhesion, reduces the cell proliferation and promotes the myoblast differentiation in C2C12 cells. Our results reveal an inhibitory effect with fibronectin (FN) in myoblast differentiation. We also observed that DPT and fibromodulin (FMOD) regulate positively to each other and promote myogenic differentiation. We further predicted the 3D structure of DPT, which is as yet unknown, and validated it using state-of-the-art in silico tools. Furthermore, we explored the in-silico protein-protein interaction between DPT-FMOD, DPT-FN, and FMOD-FN, and perceived that the interaction between FMOD-FN is more robust than DPT-FMOD and DPT-FN. Taken together, our findings have determined the role of DPT at different stages of the myogenic process.
Collapse
Affiliation(s)
- Taeyeon Kim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea.
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea.
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea.
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185236, India.
| | - Myung-Gi Seo
- Department of Veterinary Histology, College of Veterinary Medicine, Kyungpook National University, Daegu 702-701, Korea.
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea.
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea.
| |
Collapse
|
32
|
Khaghani SAB, Akbarova G, Soon CF, Dilbazi G. Effect of transforming growth factor-β2 on biological regulation of multilayer primary chondrocyte culture. Cell Tissue Bank 2018; 19:763-775. [PMID: 30377863 PMCID: PMC6280861 DOI: 10.1007/s10561-018-9732-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 10/15/2018] [Indexed: 12/22/2022]
Abstract
Cytokines are extremely potent biomolecules that regulate cellular functions and play multiple roles in initiation and inhibition of disease. These highly specialised macromolecules are actively involved in control of cellular proliferation, apoptosis, cell migration and adhesion. This work, investigates the effect of transforming growth factor-beta2 (TGF-β2) on the biological regulation of chondrocyte and the repair of a created model wound on a multilayer culture system. Also the effect of this cytokine on cell length, proliferation, and cell adhesion has been investigated. Chondrocytes isolated from knee joint of rats and cultured at 4 layers. Each layer consisted of 2 × 105 cells/ml with and without TGF-β2. The expression of mRNA and protein levels of TGF-β receptors and Smad1, 3, 4, and 7 have been analysed by RT-PCR and western blot analysis. The effect of different supplementations in chondrocyte cell proliferation, cell length, adhesion, and wound repair was statistically analysed by One-way ANOVA test. Our results showed that the TGFβ2 regulates mRNA levels of its own receptors, and of Smad3 and Smad7. Also the TGF-β2 caused an increase in chondrocyte cell length, but decreased its proliferation rate and the wound healing process. TGF-β2 also decreased cell adhesion ability to the surface of the culture flask. Since, TGF-β2 increased the cell size, but showed negative effect on cell proliferation and adhesion of CHC, the effect of manipulated TGF-β2 with other growth factors and/or proteins needs to be investigated to finalize the utilization of this growth factor and design of scaffolding in treatment of different types of arthritis.
Collapse
Affiliation(s)
| | - Gunay Akbarova
- Department of Genetics and Theory of Evolution, Faculty of Biology, Baku State University, Baku, Azerbaijan.
| | - Chin Fhong Soon
- Biosensor and Bioengineering Laboratory, MiNT-SRC Research Center, Tun Hussein Onn University of Malaysia, Batu Pahat, Johor, Malaysia
| | - Gulrukh Dilbazi
- The Laboratory of Veterinary Preparations, The Veterinary Scientific-Research Institute, Baku, Azerbaijan
| |
Collapse
|
33
|
Andenæs K, Lunde IG, Mohammadzadeh N, Dahl CP, Aronsen JM, Strand ME, Palmero S, Sjaastad I, Christensen G, Engebretsen KVT, Tønnessen T. The extracellular matrix proteoglycan fibromodulin is upregulated in clinical and experimental heart failure and affects cardiac remodeling. PLoS One 2018; 13:e0201422. [PMID: 30052659 PMCID: PMC6063439 DOI: 10.1371/journal.pone.0201422] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/13/2018] [Indexed: 01/01/2023] Open
Abstract
Pressure overload of the heart leads to cardiac remodeling that may progress into heart failure, a common, morbid and mortal condition. Increased mechanistic insight into remodeling is instrumental for development of novel heart failure treatment. Cardiac remodeling comprises cardiomyocyte hypertrophic growth, extracellular matrix alterations including fibrosis, and inflammation. Fibromodulin is a small leucine-rich proteoglycan that regulates collagen fibrillogenesis. Fibromodulin is expressed in the cardiac extracellular matrix, however its role in the heart remains largely unknown. We investigated fibromodulin levels in myocardial biopsies from heart failure patients and mice, subjected fibromodulin knock-out (FMOD-KO) mice to pressure overload by aortic banding, and overexpressed fibromodulin in cultured cardiomyocytes and cardiac fibroblasts using adenovirus. Fibromodulin was 3-10-fold upregulated in hearts of heart failure patients and mice. Both cardiomyocytes and cardiac fibroblasts expressed fibromodulin, and its expression was increased by pro-inflammatory stimuli. Without stress, FMOD-KO mice showed no cardiac phenotype. Upon aortic banding, left ventricles of FMOD-KO mice developed mildly exacerbated hypertrophic remodeling compared to wild-type mice, with increased cardiomyocyte size and altered infiltration of leukocytes. There were no differences in mortality, left ventricle dilatation, dysfunction or expression of heart failure markers. Although collagen amount and cross-linking were comparable in FMOD-KO and wild-type, overexpression of fibromodulin in cardiac fibroblasts in vitro decreased their migratory capacity and expression of fibrosis-associated molecules, i.e. the collagen-cross linking enzyme lysyl oxidase, transglutaminase 2 and periostin. In conclusion, despite a robust fibromodulin upregulation in clinical and experimental heart failure, FMOD-KO mice showed a relatively mild hypertrophic phenotype. In cultured cardiac fibroblasts, fibromodulin has anti-fibrotic effects.
Collapse
Affiliation(s)
- Kine Andenæs
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Ida G. Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Centre for Molecular Medicine Norway, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Naiyereh Mohammadzadeh
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Christen P. Dahl
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Department of Cardiology, Oslo University Hospital, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Bjørknes College, Oslo, Norway
| | - Mari E. Strand
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Sheryl Palmero
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Kristin V. T. Engebretsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Department of Surgery, Vestre Viken Hospital, Drammen, Norway
| | - Theis Tønnessen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
34
|
Larouche J, Sheoran S, Maruyama K, Martino MM. Immune Regulation of Skin Wound Healing: Mechanisms and Novel Therapeutic Targets. Adv Wound Care (New Rochelle) 2018; 7:209-231. [PMID: 29984112 PMCID: PMC6032665 DOI: 10.1089/wound.2017.0761] [Citation(s) in RCA: 362] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/12/2017] [Indexed: 12/17/2022] Open
Abstract
Significance: The immune system plays a central role in orchestrating the tissue healing process. Hence, controlling the immune system to promote tissue repair and regeneration is an attractive approach when designing regenerative strategies. This review discusses the pathophysiology of both acute and chronic wounds and possible strategies to control the immune system to accelerate chronic wound closure and promote skin regeneration (scar-less healing) of acute wounds. Recent Advances: Recent studies have revealed the key roles of various immune cells and immune mediators in skin repair. Thus, immune components have been targeted to promote chronic wound repair or skin regeneration and several growth factors, cytokines, and biomaterials have shown promising results in animal models. However, these novel strategies are often struggling to meet efficacy standards in clinical trials, partly due to inadequate drug delivery systems and safety concerns. Critical Issues: Excess inflammation is a major culprit in the dysregulation of normal wound healing, and further limiting inflammation effectively reduces scarring. However, current knowledge is insufficient to efficiently control inflammation and specific immune cells. This is further complicated by inadequate drug delivery methods. Future Directions: Improving our understanding of the molecular pathways through which the immune system controls the wound healing process could facilitate the design of novel regenerative therapies. Additionally, better delivery systems may make current and future therapies more effective. To promote the entry of current regenerative strategies into clinical trials, more evidence on their safety, efficacy, and cost-effectiveness is also needed.
Collapse
Affiliation(s)
- Jacqueline Larouche
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Victoria, Australia
| | - Sumit Sheoran
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Victoria, Australia
| | - Kenta Maruyama
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Mikaël M. Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Victoria, Australia
| |
Collapse
|
35
|
Jiang W, Ting K, Lee S, Zara JN, Song R, Li C, Chen E, Zhang X, Zhao Z, Soo C, Zheng Z. Fibromodulin reduces scar size and increases scar tensile strength in normal and excessive-mechanical-loading porcine cutaneous wounds. J Cell Mol Med 2018; 22:2510-2513. [PMID: 29392829 PMCID: PMC5867110 DOI: 10.1111/jcmm.13516] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 11/20/2017] [Indexed: 02/05/2023] Open
Abstract
Hypertrophic scarring is a major postoperative complication which leads to severe disfigurement and dysfunction in patients and usually requires multiple surgical revisions due to its high recurrence rates. Excessive-mechanical-loading across wounds is an important initiator of hypertrophic scarring formation. In this study, we demonstrate that intradermal administration of a single extracellular matrix (ECM) molecule-fibromodulin (FMOD) protein-can significantly reduce scar size, increase tensile strength, and improve dermal collagen architecture organization in the normal and even excessive-mechanical-loading red Duroc pig wound models. Since pig skin is recognized by the Food and Drug Administration as the closest animal equivalent to human skin, and because red Duroc pigs show scarring that closely resembles human proliferative scarring and hypertrophic scarring, FMOD-based technologies hold high translational potential and applicability to human patients suffering from scarring-especially hypertrophic scarring.
Collapse
Affiliation(s)
- Wenlu Jiang
- State Key Laboratory of Oral DiseasesDepartment of OrthodonticsWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
- Division of Growth and DevelopmentSection of OrthodonticsSchool of DentistryUniversity of California, Los AngelesLos AngelesCAUSA
| | - Kang Ting
- Division of Growth and DevelopmentSection of OrthodonticsSchool of DentistryUniversity of California, Los AngelesLos AngelesCAUSA
| | - Soonchul Lee
- Division of Growth and DevelopmentSection of OrthodonticsSchool of DentistryUniversity of California, Los AngelesLos AngelesCAUSA
- Department of Orthopaedic SurgeryCHA Bundang Medical CenterCHA UniversityGyeonggi‐doSouth Korea
| | - Janette N. Zara
- Department of BioengineeringSchool of EngineeringUniversity of California, Los AngelesLos AngelesCAUSA
| | - Richard Song
- Division of Growth and DevelopmentSection of OrthodonticsSchool of DentistryUniversity of California, Los AngelesLos AngelesCAUSA
| | - Chenshuang Li
- Division of Growth and DevelopmentSection of OrthodonticsSchool of DentistryUniversity of California, Los AngelesLos AngelesCAUSA
| | - Eric Chen
- Division of Growth and DevelopmentSection of OrthodonticsSchool of DentistryUniversity of California, Los AngelesLos AngelesCAUSA
| | - Xinli Zhang
- Division of Growth and DevelopmentSection of OrthodonticsSchool of DentistryUniversity of California, Los AngelesLos AngelesCAUSA
| | - Zhihe Zhao
- State Key Laboratory of Oral DiseasesDepartment of OrthodonticsWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Chia Soo
- UCLA Division of Plastic and Reconstructive SurgeryDepartment of Orthopaedic SurgeryThe Orthopaedic Hospital Research CenterUniversity of California, Los AngelesLos AngelesCAUSA
| | - Zhong Zheng
- Division of Growth and DevelopmentSection of OrthodonticsSchool of DentistryUniversity of California, Los AngelesLos AngelesCAUSA
| |
Collapse
|
36
|
Yang Z, He C, He J, Chu J, Liu H, Deng X. Curcumin-mediated bone marrow mesenchymal stem cell sheets create a favorable immune microenvironment for adult full-thickness cutaneous wound healing. Stem Cell Res Ther 2018; 9:21. [PMID: 29386050 PMCID: PMC5793416 DOI: 10.1186/s13287-018-0768-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/13/2017] [Accepted: 01/02/2018] [Indexed: 02/06/2023] Open
Abstract
Background Adult full-thickness cutaneous wound repair suffers from an imbalanced immune response, leading to nonfunctional reconstructed tissue and fibrosis. Although various treatments have been reported, the immune-mediated tissue regeneration driven by biomaterial offers an attractive regenerative strategy for damaged tissue repair. Methods In this research, we investigated a specific bone marrow-derived mesenchymal stem cell (BMSC) sheet that was induced by the Traditional Chinese Medicine curcumin (CS-C) and its immunomodulatory effects on wound repair. Comparisons were made with the BMSC sheet induced without curcumin (CS-N) and control (saline). Results In vitro cultured BMSC sheets (CS-C) showed that curcumin promoted the proliferation of BMSCs and modified the features of produced extracellular matrix (ECM) secreted by BMSCs, especially the contents of ECM structural proteins such as fibronectin (FN) and collagen I and III, as well as the ratio of collagen III/I. Two-photon fluorescence (TPF) and second-harmonic generation (SHG) imaging of mouse implantation revealed superior engraftment of BMSCs, maintained for 35 days in the CS-C group. Most importantly, CS-C created a favorable immune microenvironment. The chemokine stromal cell-derived factor 1 (SDF1) was abundantly produced by CS-C, thus facilitating a mass migration of leukocytes from which significantly increased expression of signature TH1 cells (interferon gamma) and M1 macrophages (tumor necrosis factor alpha) genes were confirmed at 7 days post-operation. The number of TH1 cells and associated pro-inflammatory M1 macrophages subsequently decreased sharply after 14 days post-operation, suggesting a rapid type I immune regression. Furthermore, the CS-C group showed an increased trend towards M2 macrophage polarization in the early phase. CS-C led to an epidermal thickness and collagen deposition that was closer to that of normal skin. Conclusions Curcumin has a good regulatory effect on BMSCs and this promising CS-C biomaterial creates a pro-regenerative immune microenvironment for cutaneous wound healing.
Collapse
Affiliation(s)
- Zhi Yang
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China
| | - Chengmin He
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China
| | - Jinyang He
- Tropical Medicine Institute, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jing Chu
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China
| | - Hanping Liu
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China.
| | - Xiaoyuan Deng
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, No. 55 Zhongshan Avenue West, Tianhe District, Guangzhou, 510631, China.
| |
Collapse
|
37
|
Lee EJ, Jan AT, Baig MH, Ahmad K, Malik A, Rabbani G, Kim T, Lee IK, Lee YH, Park SY, Choi I. Fibromodulin and regulation of the intricate balance between myoblast differentiation to myocytes or adipocyte-like cells. FASEB J 2018; 32:768-781. [PMID: 28974563 DOI: 10.1096/fj.201700665r] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Interactions between myoblasts and the surrounding microenvironment led us to explore the role of fibromodulin (FMOD), an extracellular matrix protein, in the maintenance of myoblast stemness and function. Microarray analysis of FMODkd myoblasts and in silico studies were used to identify the top most differentially expressed genes in FMODkd, and helped establish that FMOD-based regulations of integral membrane protein 2a and clusterin are essential components of the myogenic program. Studies in knockout, obese, and diabetic mouse models helped characterize the operation of a novel FMOD-based regulatory circuit that controls myoblast switching from a myogenic to a lipid accumulation fate. FMOD regulation of myoblasts is an essential part of the myogenic program, and it offers opportunities for the development of therapeutics for the treatment of different muscle diseases.-Lee, E. J., Jan, A. T., Baig, M. H., Ahmad, K., Malik, A., Rabbani, G., Kim, T., Lee, I.-K., Lee, Y. H., Park, S.-Y., Choi, I. Fibromodulin and regulation of the intricate balance between myoblast differentiation to myocytes or adipocyte-like cells.
Collapse
Affiliation(s)
- Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Arif Tasleem Jan
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Mohammad Hassan Baig
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Adeel Malik
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, South Korea
| | - Gulam Rabbani
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Taeyeon Kim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Yong Ho Lee
- Department of Biomedical Science, Catholic University of Daegu, Gyeongsan, South Korea; and
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, South Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
38
|
Hall C, Hardin C, Corkins CJ, Jiwani AZ, Fletcher J, Carlsson A, Chan R. Pathophysiologic Mechanisms and Current Treatments for Cutaneous Sequelae of Burn Wounds. Compr Physiol 2017; 8:371-405. [PMID: 29357133 DOI: 10.1002/cphy.c170016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Burn injuries are a pervasive clinical problem. Extensive thermal trauma can be life-threatening or result in long-lasting complications, generating a significant impact on quality of life for patients as well as a cost burden to the healthcare system. The importance of addressing global or systemic issues such as resuscitation and management of inhalation injuries is not disputed but is beyond the scope of this review, which focuses on cutaneous pathophysiologic mechanisms for current treatments, both in the acute and long-term settings. Pathophysiological mechanisms of burn progression and wound healing are mediated by highly complex cascades of cellular and biochemical events, which become dysregulated in slow-healing wounds such as burns. Burns can result in fibroproliferative scarring, skin contractures, or chronic wounds that take weeks or months to heal. Burn injuries are highly individualized owing to wound-specific differences such as burn depth and surface area, in addition to patient-specific factors including genetics, immune competency, and age. Other extrinsic complications such as microbial infection can complicate wound healing, resulting in prolonged inflammation and delayed re-epithelialization. Although mortality is decreasing with advancements in burn care, morbidity from postburn deformities continues to be a challenge. Optimizing specialized acute care and late burn outcome intervention on a patient-by-patient basis is critical for successful management of burn wounds and the associated pathological scar outcome. Understanding the fundamentals of integument physiology and the cellular processes involved in wound healing is essential for designing effective treatment strategies for burn wound care as well as development of future therapies. Published 2018. Compr Physiol 8:371-405, 2018.
Collapse
Affiliation(s)
- Caroline Hall
- Quality Skin Collaborative for Advanced Reconstruction and Regeneration (Q-SCARRTM), Dental Craniofacial Trauma Research Division, US Army Institute of Surgical Research, Ft. Sam Houston, TX, USA
| | - Carolyn Hardin
- Quality Skin Collaborative for Advanced Reconstruction and Regeneration (Q-SCARRTM), Dental Craniofacial Trauma Research Division, US Army Institute of Surgical Research, Ft. Sam Houston, TX, USA
| | - Christopher J Corkins
- Quality Skin Collaborative for Advanced Reconstruction and Regeneration (Q-SCARRTM), Dental Craniofacial Trauma Research Division, US Army Institute of Surgical Research, Ft. Sam Houston, TX, USA.,Clinical Division and Burn Center, US Army Institute of Surgical Research, Ft. Sam Houston, TX, USA
| | - Alisha Z Jiwani
- Quality Skin Collaborative for Advanced Reconstruction and Regeneration (Q-SCARRTM), Dental Craniofacial Trauma Research Division, US Army Institute of Surgical Research, Ft. Sam Houston, TX, USA.,Clinical Division and Burn Center, US Army Institute of Surgical Research, Ft. Sam Houston, TX, USA
| | - John Fletcher
- Quality Skin Collaborative for Advanced Reconstruction and Regeneration (Q-SCARRTM), Dental Craniofacial Trauma Research Division, US Army Institute of Surgical Research, Ft. Sam Houston, TX, USA.,Clinical Division and Burn Center, US Army Institute of Surgical Research, Ft. Sam Houston, TX, USA
| | - Anders Carlsson
- Quality Skin Collaborative for Advanced Reconstruction and Regeneration (Q-SCARRTM), Dental Craniofacial Trauma Research Division, US Army Institute of Surgical Research, Ft. Sam Houston, TX, USA.,Clinical Division and Burn Center, US Army Institute of Surgical Research, Ft. Sam Houston, TX, USA
| | - Rodney Chan
- Quality Skin Collaborative for Advanced Reconstruction and Regeneration (Q-SCARRTM), Dental Craniofacial Trauma Research Division, US Army Institute of Surgical Research, Ft. Sam Houston, TX, USA.,Clinical Division and Burn Center, US Army Institute of Surgical Research, Ft. Sam Houston, TX, USA
| |
Collapse
|
39
|
Zheng Z, James AW, Li C, Jiang W, Wang JZ, Chang GX, Lee KS, Chen F, Berthiaume EA, Chen Y, Pan HC, Chen EC, Li W, Zhao Z, Zhang X, Ting K, Soo C. Fibromodulin reduces scar formation in adult cutaneous wounds by eliciting a fetal-like phenotype. Signal Transduct Target Ther 2017; 2:17050-. [PMID: 29201497 PMCID: PMC5661627 DOI: 10.1038/sigtrans.2017.50] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 05/28/2017] [Accepted: 08/18/2017] [Indexed: 02/02/2023] Open
Abstract
Blocking transforming growth factor (TGF)β1 signal transduction has been a central strategy for scar reduction; however, this approach appears to be minimally effective. Here, we show that fibromodulin (FMOD), a 59-kD small leucine-rich proteoglycan critical for normal collagen fibrillogenesis, significantly reduces scar formation while simultaneously increasing scar strength in both adult rodent models and porcine wounds, which simulate human cutaneous scar repair. Mechanistically, FMOD uncouples pro-migration/contraction cellular signals from pro-fibrotic signaling by selectively enhancing SMAD3-mediated signal transduction, while reducing AP-1-mediated TGFβ1 auto-induction and fibrotic extracellular matrix accumulation. Consequently, FMOD accelerates TGFβ1-responsive adult fibroblast migration, myofibroblast conversion, and function. Furthermore, our findings strongly indicate that, by delicately orchestrating TGFβ1 activities rather than indiscriminately blocking TGFβ1, FMOD elicits fetal-like cellular and molecular phenotypes in adult dermal fibroblasts in vitro and adult cutaneous wounds in vivo, which is a unique response of living system undescribed previously. Taken together, this study illuminates the signal modulating activities of FMOD beyond its structural support functions, and highlights the potential for FMOD-based therapies to be used in cutaneous wound repair.
Collapse
Affiliation(s)
- Zhong Zheng
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, The Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aaron W James
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, The Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Chenshuang Li
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Wenlu Jiang
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Joyce Z Wang
- Department of Emergency Medicine, Highland General Hospital, Oakland, CA 94602, USA
| | - Grace X Chang
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kevin S Lee
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Feng Chen
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Central Laboratory, School of Stomatology, Peking University, Beijing 100081, China
| | - Emily A Berthiaume
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yao Chen
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hsin Chuan Pan
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Eric C Chen
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Weiming Li
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Orthopaedics, the First Clinical Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xinli Zhang
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kang Ting
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, The Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chia Soo
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, The Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
40
|
Mescher AL. Macrophages and fibroblasts during inflammation and tissue repair in models of organ regeneration. ACTA ACUST UNITED AC 2017; 4:39-53. [PMID: 28616244 PMCID: PMC5469729 DOI: 10.1002/reg2.77] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/30/2017] [Accepted: 04/05/2017] [Indexed: 12/15/2022]
Abstract
This review provides a concise summary of the changing phenotypes of macrophages and fibroblastic cells during the local inflammatory response, the onset of tissue repair, and the resolution of inflammation which follow injury to an organ. Both cell populations respond directly to damage and present coordinated sequences of activation states which determine the reparative outcome, ranging from true regeneration of the organ to fibrosis and variable functional deficits. Recent work with mammalian models of organ regeneration, including regeneration of full‐thickness skin, hair follicles, ear punch tissues, and digit tips, is summarized and the roles of local immune cells in these systems are discussed. New investigations of the early phase of amphibian limb and tail regeneration, including the effects of pro‐inflammatory and anti‐inflammatory agents, are then briefly discussed, focusing on the transition from the normally covert inflammatory response to the initiation of the regeneration blastema by migrating fibroblasts and the expression of genes for limb patterning.
Collapse
Affiliation(s)
- Anthony L Mescher
- Department of Anatomy and Cell Biology, Indiana University School of Medicine - Bloomington Indiana University Center for Developmental and Regenerative Biology Bloomington IN 47405 USA
| |
Collapse
|
41
|
Li LJ, Gao SQ, Peng LH, Wang XR, Zhang Y, Hu ZJ, Gao JQ. Evaluation of efficacy of aloin in treating acute trauma in vitro and in vivo. Biomed Pharmacother 2017. [DOI: 10.1016/j.biopha.2017.01.174] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|