1
|
Byun KA, Kim HM, Oh S, Batsukh S, Son KH, Byun K. Radiofrequency Treatment Attenuates Age-Related Changes in Dermal-Epidermal Junctions of Animal Skin. Int J Mol Sci 2024; 25:5178. [PMID: 38791217 PMCID: PMC11120932 DOI: 10.3390/ijms25105178] [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] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
The dermal-epidermal junction (DEJ) is essential for maintaining skin structural integrity and regulating cell survival and proliferation. Thus, DEJ rejuvenation is key for skin revitalization, particularly in age-related DEJ deterioration. Radiofrequency (RF) treatment, known for its ability to enhance collagen fiber production through thermal mechanisms and increase heat shock protein (HSP) expression, has emerged as a promising method for skin rejuvenation. Additionally, RF activates Piezo1, an ion channel implicated in macrophage polarization toward an M2 phenotype and enhanced TGF-β production. This study investigated the impact of RF treatment on HSP47 and HSP90 expression, known stimulators of DEJ protein expression. Furthermore, using in vitro and aged animal skin models, we assessed whether RF-induced Piezo1 activation and the subsequent M2 polarization could counter age-related DEJ changes. The RF treatment of H2O2-induced senescent keratinocytes upregulated the expression of HSP47, HSP90, TGF-β, and DEJ proteins, including collagen XVII. Similarly, the RF treatment of senescent macrophages increased Piezo1 and CD206 (M2 marker) expression. Conditioned media from RF-treated senescent macrophages enhanced the expression of TGF-β and DEJ proteins, such as nidogen and collagen IV, in senescent fibroblasts. In aged animal skin, RF treatment increased the expression of HSP47, HSP90, Piezo1, markers associated with M2 polarization, IL-10, and TGF-β. Additionally, RF treatment enhanced DEJ protein expression. Moreover, RF reduced lamina densa replication, disrupted lesions, promoted hemidesmosome formation, and increased epidermal thickness. Overall, RF treatment effectively enhanced DEJ protein expression and mitigated age-related DEJ structural changes by increasing HSP levels and activating Piezo1.
Collapse
Affiliation(s)
- Kyung-A Byun
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- LIBON Inc., Incheon 22006, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Hyoung Moon Kim
- Maylin Anti-Aging Center Ilsan, Goyang 10391, Republic of Korea
| | - Seyeon Oh
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Sosorburam Batsukh
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Kyunghee Byun
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
2
|
Liu C, Xu Y, Lu Y, Du P, Li X, Wang C, Guo P, Diao L, Lu G. Mesenchymal stromal cells pretreated with proinflammatory cytokines enhance skin wound healing via IL-6-dependent M2 polarization. Stem Cell Res Ther 2022; 13:414. [PMID: 35964139 PMCID: PMC9375394 DOI: 10.1186/s13287-022-02934-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/23/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Numerous studies have shown that mesenchymal stromal cells (MSCs) promote cutaneous wound healing via paracrine signaling. Our previous study found that the secretome of MSCs was significantly amplified by treatment with IFN-γ and TNF-α (IT). It has been known that macrophages are involved in the initiation and termination of inflammation, secretion of growth factors, phagocytosis, cell proliferation, and collagen deposition in wound, which is the key factor during wound healing. In this study, we aim to test whether the supernatant of MSCs pretreated with IT (S-IT MSCs) possesses a more pronounced effect on improving wound healing and describe the interplay between S-IT MSCs and macrophages as well as the potential mechanism in skin wound healing. METHODS In the present study, we used a unique supernatant of MSCs from human umbilical cord-derived MSCs (UC-MSCs) pretreated with IT, designated S-IT MSCs, subcutaneously injected into a mice total skin excision. We evaluated the effect of S-IT MSCs on the speed and quality of wound repair via IT MSCs-derived IL-6-dependent M2 polarization in vivo by hematoxylin-eosin staining (H&E), immunohistochemistry (IHC), immunofluorescence (IF), Masson's trichrome staining, Sirius red staining, quantitative real-time PCR (qPCR). In addition, the effect of S-IT MSCs on the polarization of macrophages toward M2 phenotype and the potential mechanism of it were also investigated in vitro by flow cytometry (FCM), enzyme-linked immunosorbent assay (ELISA), tube formation assay, and western blot analysis. RESULTS Compared with control supernatant (S-MSCs), our H&E and IF results showed that S-IT MSCs were more effectively in promoting macrophages convert to the M2 phenotype and enhancing phagocytosis of M2 macrophages. Meanwhile, the results of tube formation assay, IHC, Masson's trichrome staining, Sirius red staining showed that the abilities of M2 phenotype to promote vascularization and collagen deposition were significantly enhanced by S-IT MSCs-treated, thereby accelerating higher quality wound healing. Further, our ELISA, FCM, qPCR and western blot results showed that IL-6 was highly enriched in S-IT MSCs and acted as a key regulator to induce macrophages convert to the M2 phenotype through IL-6-dependent signaling pathways, ultimately achieving the above function of promoting wound repair. CONCLUSIONS These findings provide the first evidence that the S-IT MSCs is more capable of eliciting M2 polarization of macrophages via IL-6-dependent signaling pathways and accelerating wound healing, which may represent a new strategy for optimizing the therapeutic effect of MSCs on wound healing.
Collapse
Affiliation(s)
- Chenyang Liu
- Nanjng University of Traditional Chinese Medcine, Nanjng, Jiangsu, China.,Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, The Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Yan Xu
- Nanjng University of Traditional Chinese Medcine, Nanjng, Jiangsu, China.,Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, The Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Yichi Lu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Pan Du
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaoxiao Li
- Nanjng University of Traditional Chinese Medcine, Nanjng, Jiangsu, China
| | - Chengchun Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Peng Guo
- Nantong University, Nantong, Jiangsu, China
| | - Ling Diao
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, The Affiliated Hospital of Jiangnan University, Jiangsu, China.
| | - Guozhong Lu
- Nanjng University of Traditional Chinese Medcine, Nanjng, Jiangsu, China. .,Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, The Affiliated Hospital of Jiangnan University, Jiangsu, China.
| |
Collapse
|
3
|
Li J, Ding Z, Zheng X, Lu G, Lu Q, Kaplan DL. Injectable silk nanofiber hydrogels as stem cell carriers to accelerate wound healing. J Mater Chem B 2021; 9:7771-7781. [PMID: 34586152 PMCID: PMC8486307 DOI: 10.1039/d1tb01320c] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stem cells have potential utility in wound therapy, however the benefits are often limited due to cell injury from shear stress during injection and poor retention at the wound site. Here, shear-thinning silk nanofiber hydrogels were used to load bone marrow derived mesenchymal stem cells (BMSCs) and inject into wound sites to optimize cell retention and accelerate wound healing. The BMSCs in the silk nanofiber hydrogels maintained stemness better than the cells cultured on plates, and the expression of wound healing-related genes was significantly higher in the hydrogels with higher silk concentrations (2 wt%). The silk nanofibers physically prevented migration of BMSCs from the deposition site in the wound bed. In addition to faster wound healing, these BMSC-loaded hydrogels mediated angiogenesis and inflammation and improved collagen deposition and hair follicle regeneration in vivo in rats. Considering that these silk nanofiber hydrogels were successfully used here as carriers for stem cells to accelerate wound healing, further study for skin regeneration may be warranted.
Collapse
Affiliation(s)
- Jiadai Li
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, P. R. China.
- Nanjng University of Chinese Medicine, Nanjng 210000, P. R. China
| | - Zhaozhao Ding
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, P. R. China.
| | - Xin Zheng
- Department of Orthopedics, Taizhou Municipal Hospital, Taizhou 318000, P. R. China
| | - Guozhong Lu
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, The Affiliated Hospital of Jiangnan University, Wuxi 214041, P. R. China.
| | - Qiang Lu
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, P. R. China.
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, USA
| |
Collapse
|
4
|
Zhu Z, Chen B, Peng L, Gao S, Guo J, Zhu X. Blockade of LINC01605-enriched exosome generation in M2 macrophages impairs M2 macrophage-induced proliferation, migration, and invasion of human dermal fibroblasts. Int J Immunopathol Pharmacol 2021; 35:20587384211016724. [PMID: 34011185 PMCID: PMC8150463 DOI: 10.1177/20587384211016724] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Activated M2 macrophages are involved in hypertrophic scar (HS) formation via manipulating the differentiation of fibroblasts to myofibroblasts having the proliferative capacity and biological function. However, the function of exosomes derived from M2 macrophages in HS formation is unclear. Thus, this study aims to investigate the role of exosomes derived by M2 in the formation of HS. To understand the effect of exosomes derived from M2 macrophages on formation of HS, M2 macrophages were co-cultured with human dermal fibroblast (HDF) cells. Cell Counting Kit-8 assay was performed to evaluate HDF proliferation. To evaluate the migration and invasion of HDFs, wound-healing and transwell invasion assays were performed, respectively. To investigate the interaction between LINC01605 and miR-493-3p, a dual-luciferase reporter gene assay was adopted; consequently, an interaction between miR-493-3p and AKT1 was detected. Our results demonstrated that exosomes derived from M2 macrophages promoted the proliferation, migration, and invasion of HDFs. Additionally, we found that long noncoding RNA LINC01605, enriched in exosomes derived from M2 macrophages, promoted fibrosis of HDFs and that GW4869, an inhibitor of exosomes, could revert this effect. Mechanistically, LINC01605 promoted fibrosis of HDFs by directly inhibiting the secretion of miR-493-3p, and miR-493-3p down-regulated the expression of AKT1. Exosomes derived from M2 macrophages promote the proliferation and migration of HDFs by transmitting LINC01605, which may activate the AKT signaling pathway by sponging miR-493-3p. Our results provide a novel approach and basis for further investigation of the function of M2 macrophages in HS formation.
Collapse
Affiliation(s)
- Zhensen Zhu
- Department of Plastic and Burn Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Bo Chen
- Department of Plastic and Burn Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Liang Peng
- Department of Plastic and Burn Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Songying Gao
- Department of Plastic and Burn Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Jingdong Guo
- Department of Plastic and Burn Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Xiongxiang Zhu
- Department of Plastic and Burn Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| |
Collapse
|
5
|
Wang G, Sweren E, Liu H, Wier E, Alphonse MP, Chen R, Islam N, Li A, Xue Y, Chen J, Park S, Chen Y, Lee S, Wang Y, Wang S, Archer NK, Andrews W, Kane MA, Dare E, Reddy SK, Hu Z, Grice EA, Miller LS, Garza LA. Bacteria induce skin regeneration via IL-1β signaling. Cell Host Microbe 2021; 29:777-791.e6. [PMID: 33798492 PMCID: PMC8122070 DOI: 10.1016/j.chom.2021.03.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/03/2021] [Accepted: 02/24/2021] [Indexed: 02/08/2023]
Abstract
Environmental factors that enhance regeneration are largely unknown. The immune system and microbiome are attributed roles in repairing and regenerating structure but their precise interplay is unclear. Here, we assessed the function of skin bacteria in wound healing and wound-induced hair follicle neogenesis (WIHN), a rare adult organogenesis model. WIHN levels and stem cell markers correlate with bacterial counts, being lowest in germ-free (GF), intermediate in conventional specific pathogen-free (SPF), and highest in wild-type mice, even those infected with pathogenic Staphylococcus aureus. Reducing skin microbiota via cage changes or topical antibiotics decreased WIHN. Inflammatory cytokine IL-1β and keratinocyte-dependent IL-1R-MyD88 signaling are necessary and sufficient for bacteria to promote regeneration. Finally, in a small trial, a topical broad-spectrum antibiotic also slowed skin wound healing in adult volunteers. These results demonstrate a role for IL-1β to control morphogenesis and support the need to reconsider routine applications of topical prophylactic antibiotics.
Collapse
Affiliation(s)
- Gaofeng Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Evan Sweren
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Eric Wier
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Ruosi Chen
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Nasif Islam
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Ang Li
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Yingchao Xue
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Junjie Chen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21210, USA
| | - Seungman Park
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21210, USA
| | - Yun Chen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21210, USA
| | - Sam Lee
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Saifeng Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Nate K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - William Andrews
- Department of Pharmaceutical Sciences, School of Pharmacy Mass Spectrometry Center, University of Maryland, MD 21201, USA
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, School of Pharmacy Mass Spectrometry Center, University of Maryland, MD 21201, USA
| | - Erika Dare
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Sashank K Reddy
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Elizabeth A Grice
- Department of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA
| | - Luis A Garza
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA.
| |
Collapse
|
6
|
Desai K, Kale A, Angadi P, Datar U, Belaldavar C, Arany P. Role of programmed cell death 4 in myofibroblast differentiation in oral submucous fibrosis. J Oral Maxillofac Pathol 2021; 25:430-436. [PMID: 35281179 PMCID: PMC8859592 DOI: 10.4103/jomfp.jomfp_86_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/30/2021] [Accepted: 05/31/2021] [Indexed: 11/04/2022] Open
|
7
|
Bryja A, Sujka-Kordowska P, Konwerska A, Ciesiółka S, Wieczorkiewicz M, Bukowska D, Antosik P, Bryl R, Skowroński MT, Jaśkowski JM, Mozdziak P, Angelova Volponi A, Shibli JA, Kempisty B, Dyszkiewicz-Konwińska M. New Gene Markers Involved in Molecular Processes of Tissue Repair, Response to Wounding and Regeneration Are Differently Expressed in Fibroblasts from Porcine Oral Mucosa during Long-Term Primary Culture. Animals (Basel) 2020; 10:ani10111938. [PMID: 33105567 PMCID: PMC7690285 DOI: 10.3390/ani10111938] [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: 08/24/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Wound healing and vascularization mechanisms are key steps in the complex morphological process of tissue reconstruction. Additionally, these processes in the oral cavity are more rapid than in the skin and result in less scar formation. Epithelial cells and fibroblasts play an important role in the process of wound healing. In our study, we focused on fibroblasts and monitored changes in gene expression during their in vitro culture. Based on the analysis, we distinguished three groups of processes that play important roles in tissue regeneration: response to wounding, wound healing and vascularization. We identified genes that were involved in all three processes. These genes could be selected as tissue specific repair markers for oral fibroblasts. Abstract The mechanisms of wound healing and vascularization are crucial steps of the complex morphological process of tissue reconstruction. In addition to epithelial cells, fibroblasts play an important role in this process. They are characterized by dynamic proliferation and they form the stroma for epithelial cells. In this study, we have used primary cultures of oral fibroblasts, obtained from porcine buccal mucosa. Cells were maintained long-term in in vitro conditions, in order to investigate the expression profile of the molecular markers involved in wound healing and vascularization. Based on the Affymetrix assays, we have observed three ontological groups of markers as wound healing group, response to wounding group and vascularization group, represented by different genes characterized by their expression profile during long-term primary in vitro culture (IVC) of porcine oral fibroblasts. Following the analysis of gene expression in three previously identified groups of genes, we have identified that transforming growth factor beta 1 (TGFB1), ITGB3, PDPN, and ETS1 are involved in all three processes, suggesting that these genes could be recognized as markers of repair specific for oral fibroblasts within the porcine mucosal tissue.
Collapse
Affiliation(s)
- Artur Bryja
- Department of Anatomy, Poznan University of Medical Science, 60-781 Poznań, Poland; (A.B.); (R.B.); (M.D.-K.)
| | - Patrycja Sujka-Kordowska
- Department of Histology and Embryology, Poznan University of Medical Science, 60-781 Poznań, Poland; (P.S.-K.); (A.K.); (S.C.)
- Department of Anatomy and Histology, University of Zielona Gora, 65-046 Zielona Góra, Poland
| | - Aneta Konwerska
- Department of Histology and Embryology, Poznan University of Medical Science, 60-781 Poznań, Poland; (P.S.-K.); (A.K.); (S.C.)
| | - Sylwia Ciesiółka
- Department of Histology and Embryology, Poznan University of Medical Science, 60-781 Poznań, Poland; (P.S.-K.); (A.K.); (S.C.)
| | - Maria Wieczorkiewicz
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, 87-100 Toruń, Poland; (M.W.); (M.T.S.)
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Nicolaus Copernicus University in Torun, 87-100 Toruń, Poland; (D.B.); (J.M.J.)
| | - Paweł Antosik
- Department of Veterinary Surgery, Nicolaus Copernicus University in Torun, 87-100 Toruń, Poland;
| | - Rut Bryl
- Department of Anatomy, Poznan University of Medical Science, 60-781 Poznań, Poland; (A.B.); (R.B.); (M.D.-K.)
| | - Mariusz T. Skowroński
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Torun, 87-100 Toruń, Poland; (M.W.); (M.T.S.)
| | - Jędrzej M. Jaśkowski
- Department of Diagnostics and Clinical Sciences, Nicolaus Copernicus University in Torun, 87-100 Toruń, Poland; (D.B.); (J.M.J.)
| | - Paul Mozdziak
- Physiology Graduate Program, North Carolina State University, Raleigh, NC 27695, USA;
| | - Ana Angelova Volponi
- Department of Craniofacial Development and Stem Cell Biology, King’s College University of London, London WC2R 2LS, UK;
| | - Jamil A. Shibli
- Department of Periodontology and Oral Implantology, Dental Research Division, University of Guarulhos, Guarulhos SP 07030-010, Brazil;
| | - Bartosz Kempisty
- Department of Anatomy, Poznan University of Medical Science, 60-781 Poznań, Poland; (A.B.); (R.B.); (M.D.-K.)
- Department of Histology and Embryology, Poznan University of Medical Science, 60-781 Poznań, Poland; (P.S.-K.); (A.K.); (S.C.)
- Department of Veterinary Surgery, Nicolaus Copernicus University in Torun, 87-100 Toruń, Poland;
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 601 77 Brno, Czech Republic
- Correspondence: ; Tel.: +48-61-8546418
| | - Marta Dyszkiewicz-Konwińska
- Department of Anatomy, Poznan University of Medical Science, 60-781 Poznań, Poland; (A.B.); (R.B.); (M.D.-K.)
- Department of Biomaterials and Experimental Dentistry, Poznan University of Medical Sciences, 61-701 Poznań, Poland
| |
Collapse
|
8
|
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: 3.3] [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
|
9
|
Sun ZL, Feng Y, Zou ML, Zhao BH, Liu SY, Du Y, Yu S, Yang ML, Wu JJ, Yuan ZD, Lv GZ, Zhang JR, Yuan FL. Emerging Role of IL-10 in Hypertrophic Scars. Front Med (Lausanne) 2020; 7:438. [PMID: 32974363 PMCID: PMC7481393 DOI: 10.3389/fmed.2020.00438] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/06/2020] [Indexed: 12/31/2022] Open
Abstract
Hypertrophic scars (HS) arise from traumatic or surgical injuries and the subsequent abnormal wound healing, which is characterized by continuous and histologically localized inflammation. Therefore, inhibiting local inflammation is an effective method of treating HS. Recent insight into the role of interleukin-10 (IL-10), an important anti-inflammatory cytokine, in fibrosis has increased our understanding of the pathophysiology of HS and has suggested new therapeutic targets. This review summarizes the recent progress in elucidating the role of IL-10 in the formation of HS and its therapeutic potential based on current research. This knowledge will enhance our understanding of the role of IL-10 in scar formation and shed new light on the regulation and potential treatment of HS.
Collapse
Affiliation(s)
- Zi-Li Sun
- Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, China
| | - Yi Feng
- Department of Pharmacology, Medical School, Yangzhou University, Yangzhou, China
| | - Ming-Li Zou
- Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, China
| | - Bin-Hong Zhao
- Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, China
| | - Si-Yu Liu
- Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, China
| | - Yong Du
- Department of Burns and Plastic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Shun Yu
- Department of Burns and Plastic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Min-Lie Yang
- Department of Burns and Plastic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jun-Jie Wu
- Department of Burns and Plastic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Zheng-Dong Yuan
- Department of Burns and Plastic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Guo-Zhong Lv
- Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, China.,Department of Burns and Plastic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Ji-Ru Zhang
- Department of Burns and Plastic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Feng-Lai Yuan
- Wuxi Clinical Medicine School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Wuxi, China.,Department of Burns and Plastic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
10
|
Singampalli KL, Balaji S, Wang X, Parikh UM, Kaul A, Gilley J, Birla RK, Bollyky PL, Keswani SG. The Role of an IL-10/Hyaluronan Axis in Dermal Wound Healing. Front Cell Dev Biol 2020; 8:636. [PMID: 32850791 PMCID: PMC7396613 DOI: 10.3389/fcell.2020.00636] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022] Open
Abstract
Scar formation is the typical endpoint of postnatal dermal wound healing, which affects more than 100 million individuals annually. Not only do scars cause a functional burden by reducing the biomechanical strength of skin at the site of injury, but they also significantly increase healthcare costs and impose psychosocial challenges. Though the mechanisms that dictate how dermal wounds heal are still not completely understood, they are regulated by extracellular matrix (ECM) remodeling, neovascularization, and inflammatory responses. The cytokine interleukin (IL)-10 has emerged as a key mediator of the pro- to anti-inflammatory transition that counters collagen deposition in scarring. In parallel, the high molecular weight (HMW) glycosaminoglycan hyaluronan (HA) is present in the ECM and acts in concert with IL-10 to block pro-inflammatory signals and attenuate fibrotic responses. Notably, high concentrations of both IL-10 and HMW HA are produced in early gestational fetal skin, which heals scarlessly. Since fibroblasts are responsible for collagen deposition, it is critical to determine how the concerted actions of IL-10 and HA drive their function to potentially control fibrogenesis. Beyond their independent actions, an auto-regulatory IL-10/HA axis may exist to modulate the magnitude of CD4+ effector T lymphocyte activation and enhance T regulatory cell function in order to reduce scarring. This review underscores the pathophysiological impact of the IL-10/HA axis as a multifaceted molecular mechanism to direct primary cell responders and regulators toward either regenerative dermal tissue repair or scarring.
Collapse
Affiliation(s)
- Kavya L Singampalli
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States.,Department of Bioengineering, Rice University, Houston, TX, United States.,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Swathi Balaji
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | - Xinyi Wang
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | - Umang M Parikh
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | - Aditya Kaul
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | - Jamie Gilley
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States.,Division of Neonatology, Department of Pediatrics, Texas Children's Hospital, Houston, TX, United States
| | | | - Paul L Bollyky
- Division of Infectious Diseases, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Sundeep G Keswani
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| |
Collapse
|
11
|
Chitosan/LiCl composite scaffolds promote skin regeneration in full-thickness loss. SCIENCE CHINA-LIFE SCIENCES 2019; 63:552-562. [DOI: 10.1007/s11427-018-9389-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 11/24/2018] [Indexed: 12/21/2022]
|
12
|
Human Novel MicroRNA Seq-915_x4024 in Keratinocytes Contributes to Skin Regeneration by Suppressing Scar Formation. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 14:410-423. [PMID: 30731322 PMCID: PMC6365370 DOI: 10.1016/j.omtn.2018.12.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/28/2018] [Accepted: 12/28/2018] [Indexed: 12/23/2022]
Abstract
Early in gestation, wounds in fetal skin heal by regeneration, in which microRNAs play key roles. Seq-915_x4024 is a novel microRNA candidate confirmed by deep sequencing and mirTools 2.0. It is highly expressed in fetal keratinocytes during early gestation. Using an in vitro wound-healing assay, Transwell cell migration assay, and MTS proliferation assay, we demonstrated that keratinocytes overexpressing seq-915_x4024 exhibited higher proliferative activity and the ability to promote fibroblast migration and fibroblast proliferation. These characteristics of keratinocytes are the same biological behaviors as those of fetal keratinocytes, which contribute to skin regeneration. In addition, seq-915_x4024 suppressed the expression of the pro-inflammatory markers TNF-α, IL-6, and IL-8 and the pro-inflammatory chemokines CXCL1 and CXCL5. We also demonstrated that seq-915_x4024 regulates TGF-β isoforms and the extracellular matrix. Moreover, using an in vivo wound-healing model, we demonstrated that overexpression of seq-915_x4024 in keratinocytes suppresses inflammatory cell infiltration and scar formation. Using bioinformatics analyses, luciferase reporter assays, and western blotting, we further demonstrated that Sar1A, Smad2, TNF-α, and IL-8 are direct targets of seq-915_x4024. Furthermore, the expression of phosphorylated Smad2 and Smad3 was reduced by seq-915_x4024. Seq-915_x4024 could be used as an anti-fibrotic factor for the treatment of wound healing.
Collapse
|
13
|
Abstract
After a myocardial infarction, heart tissue becomes irreversibly damaged, leading to scar formation and inevitably ischemic heart failure. Of the many available interventions after a myocardial infarction, such as percutaneous intervention or pharmacological optimization, none can reverse the ischemic insult on the heart and restore cardiac function. Thus, the only available cure for patients with scarred myocardium is allogeneic heart transplantation, which comes with extensive costs, risks, and complications. However, multiple studies have shown that the heart is, in fact, not an end-stage organ and that there are endogenous mechanisms in place that have the potential to spark regeneration. Stem cell therapy has emerged as a potential tool to tap into and activate this endogenous framework. Particularly promising are stem cells derived from cardiac tissue itself, referred to as cardiosphere-derived cells (CDCs). CDCs can be extracted and isolated from the patient's myocardium and then administered by intramyocardial injection or intracoronary infusion. After early success in the animal model, multiple clinical trials have demonstrated the safety and efficacy of autologous CDC therapy in humans. Clinical trials with allogeneic CDCs showed early promising results and pose a potential "off-the-shelf" therapy for patients in the acute setting after a myocardial infarction. The mechanism responsible for CDC-induced cardiac regeneration seems to be a combination of triggering native cardiomyocyte proliferation and recruitment of endogenous progenitor cells, which most prominently occurs via paracrine effects. A further understanding of the mediators involved in paracrine signaling can help with the development of a stem cell-free therapy, with all the benefits and none of the associated complications.
Collapse
|
14
|
Pericytes reduce inflammation and collagen deposition in acute wounds. Cytotherapy 2018; 20:1046-1060. [PMID: 30093323 DOI: 10.1016/j.jcyt.2018.06.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 05/22/2018] [Accepted: 06/22/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Pericytes have been shown to have mesenchymal stromal cell-like properties and play a role in tissue regeneration. The goal of this study was to determine whether the addition of a pericyte sheet to a full-thickness dermal wound would enhance the healing of an acute wound. METHODS Human muscle-derived pericytes and human dermal fibroblasts were formed into cell sheets, then applied to full-thickness excisional wounds on the dorsum of nu/nu mice. Histology was performed to evaluate epidermal and dermal reformation, inflammation and fibrosis. In addition, real-time reverse transcriptase-polymerase chain reaction (RT-PCR) was used to determine cytokine response. RESULTS Pericytes were detected in the wounds until day 16 but not fibroblasts. Decrease in wound size was noted in pericyte sheet-treated wounds. Enhanced neo-vascularization and healthy granulation tissue formation were noted in the pericyte-treated wounds. Expression of type I collagen messenger RNA (mRNA) was significantly higher in the fibroblast-treated group, whereas Type III collagen mRNA showed significant increase in the pericyte group at days 3, 6 and 9 compared with the fibroblast and no-cell groups. Trichrome staining revealed thick unorganized collagen fibrils in the fibroblast-treated wounds, whereas pericyte-treated wounds contained thinner and more alligned collagen fibrils. Tumor necrosis factor (TNF)-α mRNA levels were increased in the fibroblast-treated wounds compared with pericyte-treated wounds. DISCUSSION The addition of pericytes may confer beneficial effects to wound healing resulting in reduced recruitment of inflammatory cells and collagen I deposition, potential to enhance wound closure and better collagen alignment promoting stronger tissue.
Collapse
|
15
|
Gao ZH, Deng CJ, Xie YY, Guo XL, Wang QQ, Liu LZ, Lee WH, Li SA, Zhang Y. Pore‐forming toxin‐like protein complex expressed by frog promotes tissue repair. FASEB J 2018; 33:782-795. [DOI: 10.1096/fj.201800087r] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Zhen-Hua Gao
- Key Laboratory of Animal Models and Human Disease Mechanisms Kunming China
- Key Laboratory of Bioactive Peptides of Yunnan ProvinceKunming Institute of Zoology Kunming China
- Kunming College of Life ScienceUniversity of Chinese Academy of Sciences Kunming China
- First Affiliated Hospital of Kunming Medical University Kunming China
| | - Cheng-Jie Deng
- Key Laboratory of Animal Models and Human Disease Mechanisms Kunming China
- Key Laboratory of Bioactive Peptides of Yunnan ProvinceKunming Institute of Zoology Kunming China
- Kunming College of Life ScienceUniversity of Chinese Academy of Sciences Kunming China
| | - Yue-Ying Xie
- Key Laboratory of Animal Models and Human Disease Mechanisms Kunming China
- Kunming College of Life ScienceUniversity of Chinese Academy of Sciences Kunming China
| | - Xiao-Long Guo
- Key Laboratory of Animal Models and Human Disease Mechanisms Kunming China
- Kunming College of Life ScienceUniversity of Chinese Academy of Sciences Kunming China
| | - Qi-Quan Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms Kunming China
- Key Laboratory of Bioactive Peptides of Yunnan ProvinceKunming Institute of Zoology Kunming China
- Kunming College of Life ScienceUniversity of Chinese Academy of Sciences Kunming China
| | - Ling-Zhen Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms Kunming China
- Key Laboratory of Bioactive Peptides of Yunnan ProvinceKunming Institute of Zoology Kunming China
- Kunming College of Life ScienceUniversity of Chinese Academy of Sciences Kunming China
| | - Wen-Hui Lee
- Key Laboratory of Animal Models and Human Disease Mechanisms Kunming China
- Key Laboratory of Bioactive Peptides of Yunnan ProvinceKunming Institute of Zoology Kunming China
| | - Sheng-An Li
- Key Laboratory of Animal Models and Human Disease Mechanisms Kunming China
- Key Laboratory of Bioactive Peptides of Yunnan ProvinceKunming Institute of Zoology Kunming China
| | - Yun Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms Kunming China
- Key Laboratory of Bioactive Peptides of Yunnan ProvinceKunming Institute of Zoology Kunming China
- Center for Excellence in Animal Evolution and GeneticsChinese Academy of Sciences Kunming China
| |
Collapse
|
16
|
Li Y, Zhang J, Zhang W, Liu Y, Li Y, Wang K, Zhang Y, Yang C, Li X, Shi J, Su L, Hu D. MicroRNA-192 regulates hypertrophic scar fibrosis by targeting SIP1. J Mol Histol 2017; 48:357-366. [PMID: 28884252 DOI: 10.1007/s10735-017-9734-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/04/2017] [Indexed: 02/08/2023]
Abstract
Hypertrophic scar (HS) is a fibro-proliferative disorder which is characterized by excessive deposition of collagen and accumulative activity of myofibroblasts. Increasing evidences have demonstrated miRNAs play a pivotal role in the pathogenesis of HS. MiR-192 is closely associated with renal fibrosis, but its effect on HS formation and skin fibrosis remains unknown. In the study, we presented that miR-192 was up-regulated in HS and HS derived fibroblasts (HSFs) compared to normal skin (NS) and NS derived fibroblasts (NSFs), accompanied by the reduction of smad interacting protein 1 (SIP1) expression and the increase of Col1, Col3 and α-SMA levels. Furthermore, we confirmed SIP1 was a direct target of miR-192 by using luciferase reporter assays. Meanwhile, the overexpression of miR-192 increased the levels of Col1, Col3 and α-SMA. The synthesis of collagen and more positive α-SMA staining were also observed in bleomycin-induced dermal fibrosis model of BALB/c mice treated with subcutaneous miR-192 mimics injection, whereas the inhibition of miR-192 decreased the expression of Col1, Col3 and α-SMA. Moreover, SIP1 siRNA could enhance the levels of Col1, Col3 and α-SMA, showing that the effect of knockdown SIP1 was similar to miR-192 mimics, and the phenomenon manifested miR-192 regulated HS fibrosis by targeting SIP1. Together, our results indicated that miR-192 was a critical factor of HS formation and facilitated skin fibrosis by targeting directly SIP1.
Collapse
Affiliation(s)
- Yan Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 West Chang-le Road, Xi'an, 710032, China
| | - Julei Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 West Chang-le Road, Xi'an, 710032, China
| | - Wei Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 West Chang-le Road, Xi'an, 710032, China
| | - Yang Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 West Chang-le Road, Xi'an, 710032, China
| | - Yuehua Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 West Chang-le Road, Xi'an, 710032, China
| | - Kejia Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 West Chang-le Road, Xi'an, 710032, China
| | - Yijie Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 West Chang-le Road, Xi'an, 710032, China
| | - Chen Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 West Chang-le Road, Xi'an, 710032, China
| | - Xiaoqiang Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 West Chang-le Road, Xi'an, 710032, China
| | - Jihong Shi
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 West Chang-le Road, Xi'an, 710032, China
| | - Linlin Su
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 West Chang-le Road, Xi'an, 710032, China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 West Chang-le Road, Xi'an, 710032, China.
| |
Collapse
|
17
|
Li M, Zhao Y, Hao H, Han W, Fu X. Theoretical and practical aspects of using fetal fibroblasts for skin regeneration. Ageing Res Rev 2017; 36:32-41. [PMID: 28238941 DOI: 10.1016/j.arr.2017.02.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 02/21/2017] [Accepted: 02/21/2017] [Indexed: 12/17/2022]
Abstract
Cutaneous wounding in late-gestational fetal or postnatal humans results in scar formation without any skin appendages. Early or mid- gestational skin healing in humans is characterized by the absence of scaring in a process resembling regeneration. Tremendous cellular and molecular mechanisms contribute to this distinction, and fibroblasts play critical roles in scar or scarless wound healing. This review discussed the different repair mechanisms involved in wound healing of fibroblasts at different developmental stages and further confirmed that fetal fibroblast transplantation resulted in reduced scar healing in vivo. We also discussed the possible problem in fetal fibroblast transplantation for wound repair. We proposed the use of small molecules to improve the regenerative potential of repairing cells in the wound given that remodeling of the wound microenvironment into a regenerative microenvironment in adults might improve skin regeneration.
Collapse
|
18
|
Xiao K, Luo X, Wang X, Gao Z. MicroRNA‑185 regulates transforming growth factor‑β1 and collagen‑1 in hypertrophic scar fibroblasts. Mol Med Rep 2017; 15:1489-1496. [PMID: 28259900 PMCID: PMC5364971 DOI: 10.3892/mmr.2017.6179] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 11/30/2016] [Indexed: 12/22/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β1) and collagen type I (Col-1) serve a critical role in the development and progression of hypertrophic scarring (HS). The present study hypothesized that a post‑translational mechanism of microRNAs (miR) regulated the expression of TGF‑β1 and Col‑1 in HS fibroblasts (HSFBs). A collection of 20 HS tissues was compared with corresponding normal tissues from clinical patients, and the expression of miR‑185 was measured. Using PicTar, TargetScan and miRBase databases, it was identified that miR‑185 may be a regulator of TGF‑β1 and Col‑1 in humans. Based on these hypotheses, the expression of miR‑185, TGF‑β1 and Col‑1 in HS tissues was investigated. The results demonstrated that the expression of miR‑185 was markedly suppressed, and TGF‑β1 and Col‑1 levels were increased, in HS tissues. The expression levels of endogenous miR‑185 negatively correlated with the TGF‑β1 and Col‑1 mRNA levels (Pearson's correlation coefficient r=‑0.674, P<0.01 and r=‑0.590, P<0.01, respectively). In vitro, miR‑185 can regulate TGF‑β1 and Col‑1 through the predicted binding sites in its 3'‑untranslated region. miR‑185 had an effect on cell proliferation and apoptosis, thereby regulating HSFBs growth. In addition, miR‑185 gain‑of‑function decreased TGF‑β1 and Col‑1 protein expression, and miR‑185 loss‑of‑function increased TGF‑β1 and Col‑1 protein expression in HSFBs. In conclusion, overexpressed miR‑185 could inhibit HSFBs growth, and the underlying mechanism was mediated, at least partly, through the suppression of TGF‑β1 and Col‑1 expression. However, above all, miR‑185 might serve as a potential therapeutic approach for the treatment of HS.
Collapse
Affiliation(s)
- Kaiyan Xiao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Xusong Luo
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Xiuxia Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Zhen Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
19
|
Zhou H, You C, Wang X, Jin R, Wu P, Li Q, Han C. The progress and challenges for dermal regeneration in tissue engineering. J Biomed Mater Res A 2017; 105:1208-1218. [PMID: 28063210 DOI: 10.1002/jbm.a.35996] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 12/30/2016] [Accepted: 01/03/2017] [Indexed: 01/17/2023]
Affiliation(s)
- Hanlei Zhou
- Department of Burns; 2nd Affiliated Hospital of Zhejiang University, College of Medicine; Hangzhou 310009 China
| | - Chuangang You
- Department of Burns; 2nd Affiliated Hospital of Zhejiang University, College of Medicine; Hangzhou 310009 China
| | - Xingang Wang
- Department of Burns; 2nd Affiliated Hospital of Zhejiang University, College of Medicine; Hangzhou 310009 China
| | - Ronghua Jin
- Department of Burns; 2nd Affiliated Hospital of Zhejiang University, College of Medicine; Hangzhou 310009 China
| | - Pan Wu
- Department of Burns; 2nd Affiliated Hospital of Zhejiang University, College of Medicine; Hangzhou 310009 China
| | - Qiong Li
- Department of Burns; 2nd Affiliated Hospital of Zhejiang University, College of Medicine; Hangzhou 310009 China
| | - Chunmao Han
- Department of Burns; 2nd Affiliated Hospital of Zhejiang University, College of Medicine; Hangzhou 310009 China
| |
Collapse
|
20
|
Administration of adipose-derived stem cells enhances vascularity, induces collagen deposition, and dermal adipogenesis in burn wounds. Burns 2016; 42:1212-22. [PMID: 27211359 DOI: 10.1016/j.burns.2015.12.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 11/20/2015] [Accepted: 12/07/2015] [Indexed: 01/09/2023]
Abstract
Current treatment options for severe burn wounds are often insufficient in reconstructing skin and soft tissue defects. Adipose-derived stem cells (ASCs), a readily available source of multipotent stem cells, represent a promising therapy for the treatment of full-thickness burn wounds. Full-thickness burn wounds were created on the paraspinal region of athymic mice. A one-time, sub-eschar injection of 6.8×10(6) ASCs in PBS or PBS alone was administered at 24-h postoperatively. Time to healing was quantified using Image J analysis. At days 4, 7, 14, and 21, mice were sacrificed and tissues were excised for molecular and histological analysis. ASCs were able to survive in burn wounds as determined by the presence of PKH labeling and human PPARγ expression within the wounds. CD-31 staining demonstrated increased vascularity in ASC-treated wounds at POD 4 (p<0.05). Molecular studies showed enhanced adipogenesis, as well as type III and type I collagen deposition in the ASC treated group (p<0.05). An increase in the mRNA expression ratio of type III to type I collagen was also observed following ASC treatment (p<0.05). By enhancing vascularity, collagen deposition, and adipogenesis, ASCs show promise as an adjunctive therapy for the current treatment of full thickness burn wounds.
Collapse
|
21
|
Gene Signature of Human Oral Mucosa Fibroblasts: Comparison with Dermal Fibroblasts and Induced Pluripotent Stem Cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:121575. [PMID: 26339586 PMCID: PMC4538314 DOI: 10.1155/2015/121575] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 04/03/2015] [Accepted: 04/10/2015] [Indexed: 01/27/2023]
Abstract
Oral mucosa is a useful material for regeneration therapy with the advantages of its accessibility and versatility regardless of age and gender. However, little is known about the molecular characteristics of oral mucosa. Here we report the first comparative profiles of the gene signatures of human oral mucosa fibroblasts (hOFs), human dermal fibroblasts (hDFs), and hOF-derived induced pluripotent stem cells (hOF-iPSCs), linking these with biological roles by functional annotation and pathway analyses. As a common feature of fibroblasts, both hOFs and hDFs expressed glycolipid metabolism-related genes at higher levels compared with hOF-iPSCs. Distinct characteristics of hOFs compared with hDFs included a high expression of glycoprotein genes, involved in signaling, extracellular matrix, membrane, and receptor proteins, besides a low expression of HOX genes, the hDFs-markers. The results of the pathway analyses indicated that tissue-reconstructive, proliferative, and signaling pathways are active, whereas senescence-related genes in p53 pathway are inactive in hOFs. Furthermore, more than half of hOF-specific genes were similarly expressed to those of hOF-iPSC genes and might be controlled by WNT signaling. Our findings demonstrated that hOFs have unique cellular characteristics in specificity and plasticity. These data may provide useful insight into application of oral fibroblasts for direct reprograming.
Collapse
|
22
|
Gras C, Ratuszny D, Hadamitzky C, Zhang H, Blasczyk R, Figueiredo C. miR-145 Contributes to Hypertrophic Scarring of the Skin by Inducing Myofibroblast Activity. Mol Med 2015; 21:296-304. [PMID: 25876136 DOI: 10.2119/molmed.2014.00172] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 04/08/2015] [Indexed: 12/21/2022] Open
Abstract
Hyperthrophic scarring of the skin is caused by excessive activity of skin myofibroblasts after wound healing and often leads to functional and/or aesthetic disturbance with significant impairment of patient quality of life. MicroRNA (miRNA) gene therapies have recently been proposed for complex processes such as fibrosis and scarring. In this study, we focused on the role of miR-145 in skin scarring and its influence in myofibroblast function. Our data showed not only a threefold increase of miR-145 levels in skin hypertrophic scar tissue but also in transforming growth factor β1 (TGF-β1)-induced skin myofibroblasts compared with healthy skin or nontreated fibroblasts (p < 0.001). Consistent with the upregulation of miR-145 induced by TGF-β1 stimulation of fibroblasts, the expression of Kruppel-like factor 4 (KLF4) was decreased by 50% and α-smooth muscle actin (α-SMA) protein expression showed a threefold increase. Both could be reversed by miR-145 inhibition (p < 0.05). Restoration of KLF4 levels equally abrogated TGF-β1-induced α-SMA expression. These data demonstrate that TGF-β1 induces miR-145 expression in fibroblasts, which in turn inhibits KLF4, a known inhibitor of α-SMA, hence upregulating α-SMA expression. Furthermore, treatment of myofibroblasts with a miR-145 inhibitor strongly decreased their α-1 type I collagen expression, TGF-β1 secretion, contractile force generation and migration. These data demonstrate that upregulation of miR-145 plays an important role in the differentiation and function of skin myofibroblasts. Additionally, inhibition of miR-145 significantly reduces skin myofibroblast activity. Taken together, these results suggest that miR-145 is a promising therapeutic target to prevent or reduce hypertrophic scarring of the skin.
Collapse
Affiliation(s)
- Christiane Gras
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Dominica Ratuszny
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Catarina Hadamitzky
- Department of Plastic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Haijiao Zhang
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Rainer Blasczyk
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
23
|
Wang Z, Liu X, Zhang D, Wang X, Zhao F, Zhang T, Wang R, Lin X, Shi P, Pang X. Phenotypic and functional modulation of 20-30 year old dermal fibroblasts by mid- and late-gestational keratinocytes in vitro. Burns 2015; 41:1064-75. [PMID: 25599870 DOI: 10.1016/j.burns.2014.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 12/16/2014] [Accepted: 12/18/2014] [Indexed: 12/24/2022]
Abstract
Fetal wound healing occurs rapidly and without scar formation early in gestation, but the mechanisms underlying this scarless healing are poorly understood. This study explores the phenotypic and functional modulation of 20-30 year old dermal fibroblasts by mid- and late-gestational keratinocytes (KCs) in vitro. Human KCs of different gestational ages were isolated, characterized, and co-cultured with human 20-30 year old fibroblasts. Gene expression and protein levels of TGF-β family members, precollagen, collagen, matrix metalloproteinases (MMPs), and the tissue inhibitors of metalloproteinases (TIMPs) were measured in the fibroblasts. Mid-gestational KCs promoted faster proliferation and migration of fibroblasts than late-gestational KCs. Additionally, significant differences in gene expression and protein levels of some markers were observed in fibroblasts co-cultured with mid- or late-gestational KCs. Fibroblasts co-cultured with mid-gestational KCs for 48 h exhibited downregulated gene expression of precollagen 1, collagen 1, TGF-β1, TGF-β2, TIMP-2 and TIMP-3, while precollagen 3, collagen 3, TGF-β3, and MMP-1, -2, -3, -9 and -14 were upregulated. In contrast, late-gestational KCs exhibited downregulated TIMP-1, TIMP-2 and TIMP-3 levels, while collagen 1, TGF-β2, TGF-β3, and MMP-2, -3, -9 and -14 were upregulated. Moreover, statistically significant differences in expression levels of precollagen 1, precollagen 3, collagen 1, TGF-β1, -β2, and -β3, MMP-1, -3 and MMP-14, TIMP-1 and TIMP-2 were found between fibroblasts co-cultured with mid- and late-gestational KCs. Furthermore, cytokine levels of IL-1a and HB-EGF were found to be statistically different between conditioned medium from mid- and late-gestational KCs. Therefore, the gestational age of KCs appears to have an important effect on scarless wound healing in the human fetus.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China; Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoyu Liu
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Dianbao Zhang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Xiliang Wang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Feng Zhao
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Tao Zhang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Rui Wang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Xuewen Lin
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Ping Shi
- Department of General Practice, The First Affiliated Hospital of China Medical, Shenyang, China
| | - Xining Pang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.
| |
Collapse
|
24
|
Morales K, Rowehl L, Smith J, Cole R, Liu F, Beyer B, Herron BJ. Mapping Novel Subcutaneous Angiogenesis Quantitative Trait Loci in [B6×MRL]F2 Mice. Adv Wound Care (New Rochelle) 2014; 3:563-572. [PMID: 25207199 DOI: 10.1089/wound.2013.0501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 12/30/2013] [Indexed: 12/14/2022] Open
Abstract
Objective: MRL/MpJ mice are known for enhanced healing, but mechanistic details or how specific aspects of wounding (e.g., angiogenesis) contribute to healing are unknown. While previous studies investigated the systemic effects of immunity in MRL/MpJ healing, few have focused on tissue-intrinsic effects. Approach:Ex vivo skin biopsies from MRL/MpJ and C57BL/6J mice were cultured in ex vivo conditions that favor endothelial cell growth to compare their angiogenic potential. We localized enhanced angiogenesis quantitative trait loci (QTL) in an F2 intercross. We then performed an expression analysis in cultured skin biopsies from MRL/MpJ and C57BL/6J mice to determine the pathways that are associated with the capacity for differential growth. Results: MRL/MpJ biopsies have a two- to threefold greater growth potential than C57BL/6J mice, supporting the hypothesis that angiogenesis may contribute to enhanced healing in MRL/MpJ skin. We mapped two QTLs that are unique from previously mapped MRL/MpJ wound healing QTLs and detected interactions between wound healing QTLs and loci in this cross. Additionally, we found that pathways previously implicated in MRL/MpJ healing are also enriched in skin biopsies. Innovation: We have developed a novel approach to determine how specific aspects of tissue development contribute to wound healing that will ultimately lead to the discovery of unidentified genes that contribute to enhanced healing. Conclusion: We have shown that, consistent with previous studies following wound closure in MRL/MpJ mice, vessel growth during healing is also influenced by genetic background. Our ongoing work will identify the genetic factors that should be useful biomarkers or as therapeutic targets for enhanced wound healing.
Collapse
Affiliation(s)
- Krista Morales
- Wadsworth Center, NYS Department of Health, Albany, New York
| | - Leahana Rowehl
- Forensic Biology, State University of New York at Albany, Albany, New York
| | - Jason Smith
- Wadsworth Center, NYS Department of Health, Albany, New York
| | - Rich Cole
- Wadsworth Center, NYS Department of Health, Albany, New York
- School of Public Health, State University of New York at Albany, Albany, New York
| | - Fang Liu
- School of Public Health, State University of New York at Albany, Albany, New York
| | - Barb Beyer
- Wadsworth Center, NYS Department of Health, Albany, New York
| | - Bruce J. Herron
- Wadsworth Center, NYS Department of Health, Albany, New York
- School of Public Health, State University of New York at Albany, Albany, New York
| |
Collapse
|
25
|
Regulating inflammation using acid-responsive electrospun fibrous scaffolds for skin scarless healing. Mediators Inflamm 2014; 2014:858045. [PMID: 24795507 PMCID: PMC3984856 DOI: 10.1155/2014/858045] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 02/18/2014] [Accepted: 02/18/2014] [Indexed: 12/26/2022] Open
Abstract
Skin injury in adult mammals brings about a series of events and inflammation in the wounded area is initiated first and provides lots of inflammatory factors, which is critical for the final scar formation. While the postinjured skin of fetus and nude mice heals scarlessly owing to the absence of inflammation or immunodeficient, we designed a feasible acid-responsive ibuprofen-loaded poly(L-lactide) (PLLA) fibrous scaffolds via doping sodium bicarbonate to prevent excessive inflammation and achieve scarless healing finally. The morphological results of in vivo experiments revealed that animals treated with acid-responsive ibuprofen-loaded PLLA fibrous scaffolds exhibited alleviative inflammation, accelerated healing process, and regulated collagen deposition via interference in the collagen distribution, the α-smooth muscle actin (α-SMA), and the basic fibroblast growth factor (bFGF) expression. The lower ratios of collagen I/collagen III and TGF-β1/TGF-β3 and higher ratio of matrix metalloproteinase-1 (MMP-1)/tissue inhibitor of metalloproteinase-1 (TIMP-1) in acid-responsive ibuprofen-loaded PLLA fibrous scaffolds group were confirmed by real-time qPCR as well. These results suggest that inhibiting the excessive inflammation will result in regular collagen distribution and appropriate ratio between the factors, which promote or suppress the scar formation, then decrease the scar area, and finally achieve the scarless healing.
Collapse
|
26
|
Hoang M, Potter JA, Gysler SM, Han CS, Guller S, Norwitz ER, Abrahams VM. Human fetal membranes generate distinct cytokine profiles in response to bacterial Toll-like receptor and nod-like receptor agonists. Biol Reprod 2014; 90:39. [PMID: 24429216 DOI: 10.1095/biolreprod.113.115428] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bacterial infection-associated inflammation is thought to be a major cause of preterm premature rupture of membranes. Proinflammatory cytokines, such as interleukin 1B (IL1B), can weaken fetal membranes (FM) by upregulating matrix metalloproteinases and inducing apoptosis. The mechanism by which infection leads to inflammation at the maternal-fetal interface and subsequent preterm birth is thought to involve innate immune pattern recognition receptors (PRR), such as the Toll-like receptors (TLR) and Nod-like receptors (NLR), which recognize pathogen-associated molecular patterns (PAMPs). The objective of this study was to determine the cytokine profile generated by FMs in response to the bacterial TLR and NLR agonists peptidoglycan (PDG; TLR2), lipopolysaccharide (LPS; TLR4), flagellin (TLR5), CpG ODN (TLR9), iE-DAP (Nod1), and MDP (Nod2). PDG, LPS, flagellin, iE-DAP, and MDP triggered FMs to generate an inflammatory response, but the cytokine profiles were distinct for each TLR and NLR agonist, and only IL1B and RANTES were commonly upregulated in response to all five PAMPs. CpG ODN, in contrast, had a mild stimulatory effect only on MCP-1 and primarily downregulated basal FM cytokine production. IL1B secretion induced by PDG, LPS, flagellin, iE-DAP, and MDP was associated with its processing. Furthermore, FM IL1B secretion in response to TLR2, TLR4, and TLR5 activation was caspase 1-dependent, whereas Nod1 and Nod2 induced IL1B secretion independent of caspase 1. These findings demonstrate that FMs respond to different bacterial TLR and NLR PAMPs by generating distinct inflammatory cytokine profiles through distinct mechanisms that are specific to the innate immune PRR activated.
Collapse
Affiliation(s)
- Mai Hoang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | | | | | | | | | | | | |
Collapse
|
27
|
Seifert AW, Maden M. New insights into vertebrate skin regeneration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 310:129-69. [PMID: 24725426 DOI: 10.1016/b978-0-12-800180-6.00004-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Regeneration biology has experienced a renaissance as clinicians, scientists, and engineers have combined forces to drive the field of regenerative medicine. Studies investigating the mechanisms that regulate wound healing in adult mammals have led to a good understanding of the stereotypical processes that lead to scarring. Despite comparative studies of fetal wound healing in which no scar is produced, the fact remains that insights from this work have failed to produce therapies that can regenerate adult human skin. In this review, we analyze past and contemporary accounts of wound healing in a variety of vertebrates, namely, fish, amphibians, and mammals, in order to demonstrate how examples of skin regeneration in adult organisms can impact traditional wound-healing research. When considered together, these studies suggest that inflammation and reepithelialization are necessary events preceding both scarring and regeneration. However, the extent to which these processes may direct one outcome over another is likely weaker than currently accepted. In contrast, the extent to which newly deposited extracellular matrix in the wound bed can be remodeled into new skin, and the intrinsic ability of new epidermis to regenerate appendages, appears to underlie the divergence between scar-free healing and the persistence of a scar. We discuss several ideas that may offer areas of overlap between researchers using these different model organisms and which may be of benefit to the ultimate goal of scar-free human wound healing.
Collapse
Affiliation(s)
- Ashley W Seifert
- Department of Biology, University of Kentucky, Lexington, Kentucky, USA
| | - Malcolm Maden
- Department of Biology and UF Genetics Institute, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
28
|
Maguire G. Stem cell therapy without the cells. Commun Integr Biol 2013; 6:e26631. [PMID: 24567776 PMCID: PMC3925653 DOI: 10.4161/cib.26631] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 09/25/2013] [Indexed: 12/14/2022] Open
Abstract
As an example of the burgeoning importance of stem cell therapy, this past month the California Institute for Regenerative Medicine (CIRM) has approved $70 million to create a new network of stem cell clinical trial centers. Much work in the last decade has been devoted to developing the use of autologous and allogeneic adult stem cell transplants to treat a number of conditions, including heart attack, dementia, wounds, and immune system-related diseases. The standard model teaches us that adult stem cells exists throughout most of the body and provide a means to regenerate and repair most tissues through replication and differentiation. Although we have often witnessed the medical cart placed in front of the scientific horse in the development of stem cell therapies outside of academic circles, great strides have been made, such as the use of purified stem cells1 instead of whole bone marrow transplants in cancer patients, where physicians avoid re-injecting the patients with their own cancer cells.2 We most often think of stem cell therapy acting to regenerate tissue through replication and then differentiation, but recent studies point to the dramatic effects adult stem cells exert in the repair of various tissues through the release of paracrine and autocrine substances, and not simply through differentiation. Indeed, up to 80% of the therapeutic effect of adult stem cells has been shown to be through paracrine mediated actions.3 That is, the collected types of molecules released by the stem cells, called the secretome, or stem cell released molecules (SRM), number in the 100s, including proteins, microRNA, growth factors, antioxidants, proteasomes, and exosomes, and target a multitude of biological pathways through paracrine actions. The composition of the different molecule types in SRM is state dependent, and varies with cell type and conditions such as age and environment.
Collapse
Affiliation(s)
- Greg Maguire
- BioRegenerative Sciences, Inc; The SRM Molecular Foundry; San Diego, CA USA
| |
Collapse
|