1
|
徐 双, 龚 政, 韩 雨, 汪 佳, 俞 璐, 徐 刚. [Role and mechanism of Prussian blue nanoparticles in the apoptosis of mouse adipose-derived mesenchymal stem cells treated with hydrogen peroxide]. ZHONGHUA SHAO SHANG YU CHUANG MIAN XIU FU ZA ZHI 2025; 41:481-490. [PMID: 40419362 PMCID: PMC12123590 DOI: 10.3760/cma.j.cn501225-20240525-00197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Indexed: 05/28/2025]
Abstract
Objective: To explore the role and mechanism of Prussian blue nanoparticle (PBNP) in the apoptosis of mouse adipose-derived stem cells (ADSCs) treated with hydrogen peroxide, providing a reference for chronic wound treatment. Methods: This research was an experimental research. PBNP with a cubic micromorphology was synthesized via the hydrothermal method. ADSCs were isolated from 6 male 6-8 weeks old Institute of Cancer Research mice using enzymatic digestion. ADSCs were divided into control group with normal culture, hydrogen peroxide group treated with hydrogen peroxide at final molarity of 200 μmol/L, and low PBNP group and high PBNP group pretreated with PBNP at final mass concentration of 10 and 20 μg/mL respectively and then treated as that in hydrogen peroxide group. After 24 h of culture, the reactive oxygen species (ROS) level was detected by fluorescence probe method, the superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), catalase (CAT), malondialdehyde (MDA) levels, and lactate dehydrogenase (LDH) release rate were measured by colorimetric method, the cell survival rate was assessed by cell counting kit-8, and the protein expression levels of B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X protein (Bax), cytochrome C (Cyt-C), cleaved cysteinyl aspartate specific protease-3 (caspase-3), cleaved caspase-9, phosphatidylinositide 3-kinase (PI3K), phospho-PI3K (p-PI3K), protein kinase B (Akt), and phospho-Akt (p-Akt) were detected by Western blotting, with ratios of p-PI3K/PI3K and p-Akt/Akt being calculated. Another batch of ADSCs were divided into control group, hydrogen peroxide group, high PBNP group, which were treated as before, and N-acetyl-L-cysteine (NAC) group, high PBNP+LY294002 group, and high PBNP+MK-2206 group pretreated with NAC at final molarity of 5 mmol/L, PBNP at final mass concentration of 20 μg/mL and LY294002 at final molarity of 10 μmol/L, and PBNP at final mass concentration of 20 μg/mL and MK-2206 at final molarity of 100 μmol/L, respectively, and then treated as that in hydrogen peroxide group. After 24 h of culture, the p-PI3K/PI3K and p-Akt/Akt ratios were detected and calculated, and protein expression levels of Bcl-2, Bax, Cyt-C, cleaved caspase-3, and cleaved caspase-9 were measured as before. There were 3 samples in all experiments. Results: After 24 h of culture, the ROS level in cells in hydrogen peroxide group was 29.0±1.1, which was significantly higher than 2.6±1.1 in control group, 16.5±0.9 in low PBNP group, and 5.3±0.9 in high PBNP group (with P values all <0.05). Compared with those in hydrogen peroxide group, the levels of SOD, CAT, and GSH-Px, the cell survival rate, the Bcl-2 protein expression level, and the ratios of p-PI3K/PI3K and p-Akt/Akt were markedly increased in cells in control group, low PBNP group, and high PBNP group (P<0.05), the MDA level and LDH release rate in cells in control group and high PBNP group and the LDH release rate in cells in low PBNP group were significantly decreased (P<0.05), and the protein expression levels of Bax, Cyt-C, cleaved caspase-9, and cleaved caspase-3 in cells in control group, low PBNP group, and high PBNP group were significantly decreased (P<0.05). After 24 h of culture, compared with those in hydrogen peroxide group, the ratios of p-PI3K/PI3K and p-Akt/Akt, as well as Bcl-2 protein expression level were significantly increased in cells in control group, NAC group, and high PBNP group (P<0.05), while the protein expression levels of Bax, Cyt-C, cleaved caspase-9, and cleaved caspase-3 were significantly decreased (P<0.05). Compared with those in high PBNP group, the ratios of p-PI3K/PI3K and p-Akt/Akt, as well as Bcl-2 protein expression level were significantly decreased in cells in high PBNP+LY-294002 group and high PBNP+MK-2206 group (P<0.05), while the protein expression levels of Bax, Cyt-C, cleaved caspase-9, and cleaved caspase-3 were significantly increased (P<0.05). Conclusions: PBNP can inhibit apoptosis of mouse ADSCs caused by oxidative stress through activating the PI3K/Akt signaling pathway and reducing the expression level of apoptosis-related proteins in cells.
Collapse
Affiliation(s)
- 双依 徐
- />扬州大学附属苏北人民医院/苏北人民医院整形烧伤外科, 扬州 225009Department of Plastic and Burn Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Northern Jiangsu People's Hospital, Yangzhou 225009, China
| | - 政 龚
- />扬州大学附属苏北人民医院/苏北人民医院整形烧伤外科, 扬州 225009Department of Plastic and Burn Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Northern Jiangsu People's Hospital, Yangzhou 225009, China
| | - 雨佳 韩
- />扬州大学附属苏北人民医院/苏北人民医院整形烧伤外科, 扬州 225009Department of Plastic and Burn Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Northern Jiangsu People's Hospital, Yangzhou 225009, China
| | - 佳希 汪
- />扬州大学附属苏北人民医院/苏北人民医院整形烧伤外科, 扬州 225009Department of Plastic and Burn Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Northern Jiangsu People's Hospital, Yangzhou 225009, China
| | - 璐 俞
- />扬州大学附属苏北人民医院/苏北人民医院整形烧伤外科, 扬州 225009Department of Plastic and Burn Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Northern Jiangsu People's Hospital, Yangzhou 225009, China
| | - 刚 徐
- />扬州大学附属苏北人民医院/苏北人民医院整形烧伤外科, 扬州 225009Department of Plastic and Burn Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Northern Jiangsu People's Hospital, Yangzhou 225009, China
| |
Collapse
|
2
|
Yan R, Liu Z, Wang S, Fan D. 1α,25-Dihydroxyvitamin D3 accelerates skin wound re-epithelialization by promoting epidermal stem cell proliferation and differentiation through PI3K activation: an in vitro and in vivo study. Braz J Med Biol Res 2025; 58:e14121. [PMID: 40053036 PMCID: PMC11884782 DOI: 10.1590/1414-431x2025e14121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 01/28/2025] [Indexed: 03/10/2025] Open
Abstract
1α,25-Dihydroxyvitamin D3 (VD3), the active form of vitamin D, plays a crucial role in wound healing. In this study, we aimed to investigate the effect of VD3 on the proliferation and differentiation of epidermal stem cells (EpSCs) and monitor its impact on re-epithelialization. We established a murine full-thickness skin defect model and applied four doses of VD3 (0, 5, 50, and 250 ng/mouse/day) to the wounds topically for three days. Immunostaining and flow cytometry confirmed the effect of VD3 on the proliferation and differentiation of EpSCs in wounds. This effect of VD3 (0, 1, 10, and 50 nM) on EpSCs and its possible mechanism were further confirmed in vitro by CCK8, westen blot, immunostaining, and flow cytometry. We found that on day five post-wounding, the means±SD length of the neo-epidermis was 195.88±11.57, 231.84±16.45, 385.80±17.50, and 268.00±8.22 μm in the control, 5, 50, and 250 ng groups, respectively, with a significant difference from the control (all P<0.05). Immunostaining and flow cytometry showed that VD3 improved the proliferation and differentiation of K15+ EpSC (vs control, all P<0.05), K14+ epidermal progenitor cells (vs control, all P<0.05), and K10+ epidermal terminal cells (vs control, all P<0.05) in vivo and in vitro. The PI3K signaling pathway appeared to underlie this response because significant inhibition of the response was found when inhibitors were used to inhibit PI3K. Our study demonstrated that VD3 is a potent promoter of cutaneous wound healing by stimulating EpSC proliferation and differentiation through PI3K activation.
Collapse
Affiliation(s)
- Rongshuai Yan
- Department of Plastic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhihui Liu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Song Wang
- Department of Burn and Plastic Surgery, General Hospital of Central Theater Command, Wuhan, China
| | - Dongli Fan
- Department of Plastic Surgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
3
|
Nirban S, Pandey M, Kumar S. Topical formulation for wound management in animals: A paradigm shifts from conventional to advance. ANNALES PHARMACEUTIQUES FRANÇAISES 2025:S0003-4509(25)00039-2. [PMID: 40020871 DOI: 10.1016/j.pharma.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/20/2025] [Accepted: 02/23/2025] [Indexed: 03/03/2025]
Abstract
Chronic wounds are a significant clinical problem for the healthcare system and require several intensive efforts to improve alternative drug delivery systems for wound care. Sometimes, this is insufficient to support the healing process. There are several obstacles to effective wound therapy, such as poor healing, off-targeting, non-compliance, and frequent dosage. Animal healthcare is a much-needed service for pet owners, farmers, and government agencies. However, the varied physiology of animals presents a challenge in producing effective medicines for animal health. To improve drug therapeutic performance, reduce animal stress, and minimize adverse effects, the industry focuses on current developments in technology for wound management in animals. The literature search utilized various reviews, research articles, clinical trials, case reports, etc. Search engines like Google Scholar, PubMed, and ScienceDirect were used to retrieve data. Various keywords such as "wound dressing and animals", "antimicrobials and animal wounds", "wound healing and animals" etc. were used to search the literature. Different formulation avenues are being explored, including hydrogels, wafers, and nanoemulsions. Therefore, further research is necessary to explore wound healing techniques. Utilising various advanced drug delivery systems for veterinary medicine has significantly reduced adverse effects, the frequency of doses, and stress on animals, resulting in increased profits for the industry. However, this also requires investment in research to ensure the safety, quality, and effectiveness of drugs, and delivery systems. This article reviews the various phases involved in wound healing, multiple factors affecting wound healing, different wound healing approaches, clinical trials, and case studies.
Collapse
Affiliation(s)
- Sunita Nirban
- Central University of Haryana, Jant-Pali, Mahendergarh, Haryana, India 123031
| | - Manisha Pandey
- Central University of Haryana, Jant-Pali, Mahendergarh, Haryana, India 123031.
| | - Sumit Kumar
- Central University of Haryana, Jant-Pali, Mahendergarh, Haryana, India 123031.
| |
Collapse
|
4
|
Pontiggia L, Yosef H, Biedermann T, Moehrlen U. Characterization of Human Skin Derived Cells by Raman Micro-Spectroscopy. Methods Mol Biol 2025; 2922:209-227. [PMID: 40208538 DOI: 10.1007/978-1-0716-4510-9_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Studying human skin biology can aid in understanding the pathophysiology of skin diseases and developing novel therapies, including tissue engineering approaches. Among various optical and spectroscopic techniques, Raman micro-spectroscopy stands out. This non-invasive method requires minimal sample preparation and provides molecular-level information on healthy and diseased cell types. The following chapter aims to provide a comprehensive guide to the basic methods of Raman micro-spectroscopy analysis of human epidermal samples from the perspective of their biochemical-molecular relationship, without the need for a deep and specialized knowledge in the field of spectroscopy. We do this based on two distinct examples: (1) Unstained cell subpopulations identified by hierarchical cluster analysis are compared with antibody treated sorted cell populations, and (2) skin cell type-specific molecular features are determined, and the composition of unknown cellular mixtures is revealed.
Collapse
Affiliation(s)
- Luca Pontiggia
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children's Hospital Zurich, Zurich, Switzerland.
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.
| | | | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Ueli Moehrlen
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
- Spina Bifida Center, University Children's Hospital Zurich, Zurich, Switzerland
- Zurich Center for Fetal Diagnosis and Treatment, Zurich, Switzerland
| |
Collapse
|
5
|
Phiboonchaiyanan PP, Harikarnpakdee S, Songsak T, Chowjarean V. In Vitro Evaluation of Wound Healing, Stemness Potentiation, Antioxidant Activity, and Phytochemical Profile of Cucurbita moschata Duchesne Fruit Pulp Ethanolic Extract. Adv Pharmacol Pharm Sci 2024; 2024:9288481. [PMID: 39502575 PMCID: PMC11535185 DOI: 10.1155/2024/9288481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/03/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Wound healing comprises an intricate process to repair damaged tissue. Research on plant extracts with properties to expedite wound healing has been of interest, particularly their ability to enhance the stemness of keratinocyte stem cells. Hence, the present study aims to determine the wound healing and stemness potentiation properties of an ethanolic extract derived from Cucurbita moschata fruit pulp (PKE). Human keratinocytes (HaCaT) and primary skin fibroblast cells were used in this study. The migration of the cells was examined by using a scratch wound healing assay, and spheroid behavior was determined by using a spheroid formation assay. The proteins related to migration and stemness were further measured by using Western blotting to explore the mechanism of action of PKE. The methods used to evaluate PKE's antioxidant properties were 2,2-diphenyl-2-picrylhydrazyl (DPPH) scavenging, ABTS radical scavenging activity, and superoxide anion radical scavenging (SOSA) assays. The phytochemistry of the PKE was investigated using phytochemical screening and high-performance liquid chromatography (HPLC) analysis. The results of this study indicate that nontoxic concentrations of PKE increase the rate of migration and spheroid formation. Mechanistically, PKE increased the expression of the migratory-related protein active FAK (phosphorylated FAK), and the subsequence increased the level of p-AKT. The expression of stem cell marker CD133, upstream protein signaling β-catenin, and self-renewal transcription factor Nanog was increased. The PKE also possessed scavenging properties against DPPH, ABTS, and SOSA. The phytochemistry analyses exhibited the presence of alkaloids, glycosides, xanthones, triterpenes, and steroids. Additionally, bioactive compounds such as ɑ-tocopherol, riboflavin, protocatechuic acid, β-carotene, and luteolin were detected. The presence of these chemicals in PKE may contribute to its antioxidant, stem cell potentiation, and wound-healing effects. The findings could be beneficial in the identification of valuable natural resources that possess the capacity to be used in the process of wound healing through the potentiation of stemness via a readily detectable molecular mechanism.
Collapse
Affiliation(s)
| | - Saraporn Harikarnpakdee
- Department of Industrial Pharmacy, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand
| | - Thanapat Songsak
- Department of Pharmacognosy, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand
| | - Verisa Chowjarean
- Department of Pharmaceutical Technology, College of Pharmacy, Rangsit University, Pathum Thani 12000, Thailand
| |
Collapse
|
6
|
Pontiggia L, Klar AS, Michalak-Micka K, Moehrlen U, Biedermann T. Isolation, Characterization, and Utilization of Human Skin Basal and Suprabasal Epidermal Stem Cells. Methods Mol Biol 2024; 2849:1-15. [PMID: 38904915 DOI: 10.1007/7651_2024_551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Studying human skin biology can aid in comprehending the pathophysiology of skin diseases and developing novel cell-based therapies, including tissue engineering approaches. This chapter provides a comprehensive guide of methods to determine human skin samples from the perspective of their cellular compositions. We describe as useful technique the histological analysis of tissue sections. We further illustrate the biological characterization of isolated and cultured basal and suprabasal interfollicular keratinocytes by cell sorting, cytospin immunostaining, colony forming efficiency, and long-term dermo-epidermal organotypic cultures.
Collapse
Affiliation(s)
- Luca Pontiggia
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children's Hospital Zurich, Schlieren, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Agnes S Klar
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children's Hospital Zurich, Schlieren, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Katarzyna Michalak-Micka
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children's Hospital Zurich, Schlieren, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Ueli Moehrlen
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children's Hospital Zurich, Schlieren, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
- Spina Bifida Center, University Children's Hospital Zurich, Zurich, Switzerland
- Zurich Center for Fetal Diagnosis and Treatment, Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children's Hospital Zurich, Schlieren, Switzerland.
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.
- University of Zurich, Zurich, Switzerland.
| |
Collapse
|
7
|
Martin‐Piedra MA, Carmona G, Campos F, Carriel V, Fernández‐González A, Campos A, Cuende N, Garzón I, Gacto P, Alaminos M. Histological assessment of nanostructured fibrin-agarose skin substitutes grafted in burnt patients. A time-course study. Bioeng Transl Med 2023; 8:e10572. [PMID: 38023713 PMCID: PMC10658487 DOI: 10.1002/btm2.10572] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 12/01/2023] Open
Abstract
A previously developed fibrin-agarose skin model-UGRSKIN-showed promising clinical results in severely burnt patients. To determine the histological parameters associated to the biocompatibility and therapeutic effects of this model, we carried out a comprehensive structural and ultrastructural study of UGRSKIN grafted in severely burnt patients after 3 months of follow-up. The grafted epidermis was analogue to native human skin from day 30th onward, revealing well-structured strata with well-differentiated keratinocytes expressing CK5, CK8, CK10, claudin, plakoglobin, filaggrin, and involucrin in a similar way to controls, suggesting that the epidermis was able to mature and differentiate very early. Melanocytes and Langerhans cells were found from day 30th onward, together with a basement membrane, abundant hemidesmosomes and lack of rete ridges. At the dermal layer, we found an interface between the grafted skin and the host tissue at day 30th, which tended to disappear with time. The grafted superficial dermis showed a progressive increase in properly-oriented collagen fibers, elastic fibers and proteoglycans, including decorin, similarly to control dermis at day 60-90th of in vivo follow-up. Blood vessels determined by CD31 and SMA expression were more abundant in grafted skin than controls, whereas lymphatic vessels were more abundant at day 90th. These results contribute to shed light on the histological parameters associated to biocompatibility and therapeutic effect of the UGRSKIN model grafted in patients and demonstrate that the bioengineered skin grafted in patients is able to mature and differentiate very early at the epithelial level and after 60-90 days at the dermal level.
Collapse
Affiliation(s)
- Miguel Angel Martin‐Piedra
- Tissue Engineering Group, Department of HistologyUniversity of GranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
| | - Gloria Carmona
- Andalusian Network for the Design and Translation of Advanced Therapies (former Andalusian Initiative for Advanced Therapies) ‐ Fundación Andaluza Progreso y Salud, Junta de Andalucía, Seville, Spain; Andalusian Transplant Coordination, Servicio Andaluz de SaludSevilleSpain
- Doctoral program in BiomedicineUniversity of GranadaGranadaSpain
| | - Fernando Campos
- Tissue Engineering Group, Department of HistologyUniversity of GranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
| | - Víctor Carriel
- Tissue Engineering Group, Department of HistologyUniversity of GranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
| | - Ana Fernández‐González
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
- Andalusian Network for the Design and Translation of Advanced Therapies (former Andalusian Initiative for Advanced Therapies) ‐ Fundación Andaluza Progreso y Salud, Junta de Andalucía, Seville, Spain; Andalusian Transplant Coordination, Servicio Andaluz de SaludSevilleSpain
- Unidad de Producción Celular e Ingeniería TisularHospital Universitario Virgen de las NievesGranadaSpain
| | - Antonio Campos
- Tissue Engineering Group, Department of HistologyUniversity of GranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
| | - Natividad Cuende
- Andalusian Network for the Design and Translation of Advanced Therapies (former Andalusian Initiative for Advanced Therapies) ‐ Fundación Andaluza Progreso y Salud, Junta de Andalucía, Seville, Spain; Andalusian Transplant Coordination, Servicio Andaluz de SaludSevilleSpain
| | - Ingrid Garzón
- Tissue Engineering Group, Department of HistologyUniversity of GranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
| | | | - Miguel Alaminos
- Tissue Engineering Group, Department of HistologyUniversity of GranadaSpain
- Instituto de Investigación Biosanitaria ibs.GRANADAGranadaSpain
| |
Collapse
|
8
|
Tang Y, Jia Z, Li X, Zhao X, Zhang S, Luo L, Xia L, Fang Z, Zhang Y, Chen M. Mechanism of wound repair in diabetic rats using nanosilver-free alginate dressing. J Wound Care 2023; 32:cli-clx. [PMID: 37561702 DOI: 10.12968/jowc.2023.32.sup8.cli] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
OBJECTIVE Nanosilver-alginate dressing can effectively promote the healing of diabetic wounds in rats. However, due to the potential toxicity of nanosilver, its widespread application in hard-to-heal wound healing is limited. In the present study, the role and potential mechanism of nanosilver-free alginate gel (NSFAG) in the healing process of diabetic wounds were explored. METHOD A diabetic rat skin wound model was established, and wounds were treated with saline (NC group), nanosilver gel (NSG group) or nanosilver-free alginate gel (NSFAG group) for seven consecutive days. RESULTS NSFAG significantly promoted wound healing and increased the content of protein and hydroxyproline in granulation tissues, and was superior to NSG (p<0.05). Immunohistochemical analyses revealed that the skin wound tissue structure of the NSFAG group was intact, and the number of skin appendages in the dermis layer was significantly higher compared with the NC group and the NSG group (p<0.05). Western blot analysis found that the protein expression of the epidermal stem cell marker molecules CK19 and CK14 as well the proliferation marker of keratinocytes Ki67 in the NSFAG group was significantly higher compared with the NC group or NSG group (p<0.05). Additionally, the proliferation marker of keratinocytes Ki67 in the NSFAG group was significantly higher compared with the NC or NSG group (p<0.05). Immunofluorescence staining analyses indicated that the CK19- and CK14-positive cells were mainly distributed around the epidermis and the newly formed appendages in the NSFAG group, and this result was not observed in the NC or NSG groups. CONCLUSION The present findings demonstrate that NSFAG can effectively accelerate wound healing in diabetic rats by promoting epidermal stem cell proliferation and differentiation into skin cells, as well as formation of granulation tissue, suggesting that it can be a potential dressing for diabetic wounds.
Collapse
Affiliation(s)
- Ying Tang
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei 230032, People's Republic of China
| | - Zeguo Jia
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei 230032, People's Republic of China
| | - Xueting Li
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei 230032, People's Republic of China
| | - Xiaotong Zhao
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei 230032, People's Republic of China
| | - Shiqi Zhang
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei 230032, People's Republic of China
| | - Li Luo
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei 230032, People's Republic of China
| | - Li Xia
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei 230032, People's Republic of China
| | - Zhaohui Fang
- Institute of Traditional Chinese Medicine Diabetes Prevention, Anhui Academy of Traditional Chinese Medicine, People's Republic of China
| | - Yuanzhi Zhang
- Hefei Institute of Physical Science, Chinese Academy of Sciences, People's Republic of China
| | - Mingwei Chen
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei 230032, People's Republic of China
- Institute of Traditional Chinese Medicine Diabetes Prevention, Anhui Academy of Traditional Chinese Medicine, People's Republic of China
| |
Collapse
|
9
|
Cheng D, Zhu X, Yan S, Shi L, Liu Z, Zhou X, Bi X. New insights into inflammatory memory of epidermal stem cells. Front Immunol 2023; 14:1188559. [PMID: 37325632 PMCID: PMC10264694 DOI: 10.3389/fimmu.2023.1188559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/16/2023] [Indexed: 06/17/2023] Open
Abstract
Inflammatory memory, as one form of innate immune memory, has a wide range of manifestations, and its occurrence is related to cell epigenetic modification or metabolic transformation. When re-encountering similar stimuli, executing cells with inflammatory memory function show enhanced or tolerated inflammatory response. Studies have identified that not only hematopoietic stem cells and fibroblasts have immune memory effects, but also stem cells from various barrier epithelial tissues generate and maintain inflammatory memory. Epidermal stem cells, especially hair follicle stem cells, play an essential role in wound healing, immune-related skin diseases, and skin cancer development. In recent years, it has been found that epidermal stem cells from hair follicle can remember the inflammatory response and implement a more rapid response to subsequent stimuli. This review updates the advances of inflammatory memory and focuses on its mechanisms in epidermal stem cells. We are finally looking forward to further research on inflammatory memory, which will allow for the development of precise strategies to manipulate host responses to infection, injury, and inflammatory skin disease.
Collapse
Affiliation(s)
- Dapeng Cheng
- Department of Dermatology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaochen Zhu
- Department of Dermatology, Changhai Hospital, Naval Medical University, Shanghai, China
- Department of Dermatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Shaochen Yan
- Department of Dermatology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Linli Shi
- Department of Dermatology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhi Liu
- Department of Dermatology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xin Zhou
- Department of Dermatology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xinling Bi
- Department of Dermatology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
10
|
Bhatt T, Dey R, Hegde A, Ketkar AA, Pulianmackal AJ, Deb AP, Rampalli S, Jamora C. Initiation of wound healing is regulated by the convergence of mechanical and epigenetic cues. PLoS Biol 2022; 20:e3001777. [PMID: 36112666 PMCID: PMC9522318 DOI: 10.1371/journal.pbio.3001777] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 09/29/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022] Open
Abstract
Wound healing in the skin is a complex physiological process that is a product of a cell state transition from homeostasis to repair. Mechanical cues are increasingly being recognized as important regulators of cellular reprogramming, but the mechanism by which it is translated to changes in gene expression and ultimately cellular behavior remains largely a mystery. To probe the molecular underpinnings of this phenomenon further, we used the down-regulation of caspase-8 as a biomarker of a cell entering the wound healing program. We found that the wound-induced release of tension within the epidermis leads to the alteration of gene expression via the nuclear translocation of the DNA methyltransferase 3A (DNMT3a). This enzyme then methylates promoters of genes that are known to be down-regulated in response to wound stimuli as well as potentially novel players in the repair program. Overall, these findings illuminate the convergence of mechanical and epigenetic signaling modules that are important regulators of the transcriptome landscape required to initiate the tissue repair process in the differentiated layers of the epidermis. Wound healing in the skin is a complex physiological process that entails a cell state transition from homeostasis to repair. This study reveals a mechanism involving nuclear translocation of DNA methyltransferase 3A (DNMT3a) that initiates the wound-healing process and is perturbed in skin diseases such as psoriasis.
Collapse
Affiliation(s)
- Tanay Bhatt
- IFOM-inStem Joint Research Laboratory, Center for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
- National Centre for Biological Sciences, Bangalore, India
| | - Rakesh Dey
- IFOM-inStem Joint Research Laboratory, Center for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Akshay Hegde
- IFOM-inStem Joint Research Laboratory, Center for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
- School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | - Alhad Ashok Ketkar
- Center for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Ajai J. Pulianmackal
- IFOM-inStem Joint Research Laboratory, Center for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Ashim P. Deb
- IFOM-inStem Joint Research Laboratory, Center for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Shravanti Rampalli
- Center for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Colin Jamora
- IFOM-inStem Joint Research Laboratory, Center for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
- * E-mail:
| |
Collapse
|
11
|
Jazaeri M, Torkzaban P, Afshar S, Najafi-Vosough R, Arany P, Gholami L. Comparison of Pulsed and Continuous Wave Diode Laser at 940 nm on the Viability and Migration of Gingival Fibroblasts. Photochem Photobiol 2022; 99:1003-1009. [PMID: 36086909 DOI: 10.1111/php.13711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/07/2022] [Indexed: 11/28/2022]
Abstract
Gingival fibroblasts have critical roles in oral wound healing. Photobiomodulation (PBM) has been shown to promote mucosal healing and is now recommended for managing oncotherapy-associated oral mucositis. This study examined the effects of the emission mode of a 940 nm diode laser on the viability and migration of human gingival fibroblasts. Cells were cultured in a routine growth media and treated with PBM (average power 0.1 W/cm2 , average fluence 3 J/cm2 , every 12h for 6 sessions) in one continuous wave (CW) and two pulsing settings with 20 % and 50 % duty cycles. Cell viability was assessed using MTT, and digital imaging quantified cell migration. After 48 and 72 hours, all treatment groups had significantly higher viability (n = 6, p < 0.05) compared to the control. The highest viability was seen in the pulsed (20% duty cycle) group at the 72-hour time point. PBM improved fibroblast migration in all PBM-treated groups, but differences were not statistically significant (n = 2, p > 0.05). PBM treatments can promote cell viability in both continuous and pulsed modes. Further studies are needed to elucidate the optimal setting for PBM-evoked responses for its rationalized use in promoting specific phases of oral wound healing.
Collapse
Affiliation(s)
- Marzieh Jazaeri
- Dental Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Parviz Torkzaban
- Department of Periodontics, School of Dentistry, Dental Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saeid Afshar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Roya Najafi-Vosough
- Department of Biostatistics, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Praveen Arany
- Oral Biology, Surgery, Biomedical Engineering, University at Buffalo, NY, USA
| | - Leila Gholami
- Department of Periodontics, School of Dentistry, Dental Implants Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| |
Collapse
|
12
|
Pontiggia L, Ahuja AK, Yosef HK, Rütsche D, Reichmann E, Moehrlen U, Biedermann T. Human Basal and Suprabasal Keratinocytes Are Both Able to Generate and Maintain Dermo-Epidermal Skin Substitutes in Long-Term In Vivo Experiments. Cells 2022; 11:2156. [PMID: 35883599 PMCID: PMC9319791 DOI: 10.3390/cells11142156] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 11/16/2022] Open
Abstract
The basal layer of human interfollicular epidermis has been described to harbour both quiescent keratinocyte stem cells and a transit amplifying cell population that maintains the suprabasal epidermal layers. We performed immunofluorescence analyses and revealed that the main proliferative keratinocyte pool in vivo resides suprabasally. We isolated from the human epidermis two distinct cell populations, the basal and the suprabasal keratinocytes, according to the expression of integrin β4 (iβ4). We compared basal iβ4+ or suprabasal iβ4- keratinocytes with respect to their proliferation and colony-forming ability and their Raman spectral properties. In addition, we generated dermo-epidermal substitutes using freshly isolated and sorted basal iβ4+ or suprabasal iβ4- keratinocytes and transplanted them on immuno-compromised rats. We show that suprabasal iβ4- keratinocytes acquire a similar proliferative capacity as basal iβ4+ keratinocytes after two weeks of culture in vitro, with expression of high levels of iβ4 and downregulation of K10 expression. In addition, both basal iβ4+ and suprabasal iβ4- keratinocytes acquire authentic self-renewing properties during the in vitro 3D-culture phase and are able to generate and maintain a fully stratified epidermis for 16 weeks in vivo. Therefore, against the leading dogma, we propose that human suprabasal keratinocytes can retro-differentiate into true basal stem cells in a wound situation and/or when in contact with the basement membrane.
Collapse
Affiliation(s)
- Luca Pontiggia
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children’s Hospital Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (A.K.A.); (D.R.); (E.R.); (U.M.); (T.B.)
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
| | - Akshay Kumar Ahuja
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children’s Hospital Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (A.K.A.); (D.R.); (E.R.); (U.M.); (T.B.)
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
| | | | - Dominic Rütsche
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children’s Hospital Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (A.K.A.); (D.R.); (E.R.); (U.M.); (T.B.)
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
| | - Ernst Reichmann
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children’s Hospital Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (A.K.A.); (D.R.); (E.R.); (U.M.); (T.B.)
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
| | - Ueli Moehrlen
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children’s Hospital Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (A.K.A.); (D.R.); (E.R.); (U.M.); (T.B.)
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
- Department of Pediatric Surgery, University Children’s Hospital Zurich, University of Zurich, 8032 Zurich, Switzerland
- Zurich Center for Fetal Diagnosis and Treatment, 8032 Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Pediatric Surgery, University Children’s Hospital Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland; (A.K.A.); (D.R.); (E.R.); (U.M.); (T.B.)
- Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
13
|
Aleemardani M, Trikić MZ, Green NH, Claeyssens F. The Importance of Mimicking Dermal-Epidermal Junction for Skin Tissue Engineering: A Review. Bioengineering (Basel) 2021; 8:bioengineering8110148. [PMID: 34821714 PMCID: PMC8614934 DOI: 10.3390/bioengineering8110148] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/14/2021] [Accepted: 10/16/2021] [Indexed: 12/11/2022] Open
Abstract
There is a distinct boundary between the dermis and epidermis in the human skin called the basement membrane, a dense collagen network that creates undulations of the dermal-epidermal junction (DEJ). The DEJ plays multiple roles in skin homeostasis and function, namely, enhancing the adhesion and physical interlock of the layers, creating niches for epidermal stem cells, regulating the cellular microenvironment, and providing a physical boundary layer between fibroblasts and keratinocytes. However, the primary role of the DEJ has been determined as skin integrity; there are still aspects of it that are poorly investigated. Tissue engineering (TE) has evolved promising skin regeneration strategies and already developed TE scaffolds for clinical use. However, the currently available skin TE equivalents neglect to replicate the DEJ anatomical structures. The emergent ability to produce increasingly complex scaffolds for skin TE will enable the development of closer physical and physiological mimics to natural skin; it also allows researchers to study the DEJ effect on cell function. Few studies have created patterned substrates that could mimic the human DEJ to explore their significance. Here, we first review the DEJ roles and then critically discuss the TE strategies to create the DEJ undulating structure and their effects. New approaches in this field could be instrumental for improving bioengineered skin substitutes, creating 3D engineered skin, identifying pathological mechanisms, and producing and screening drugs.
Collapse
Affiliation(s)
- Mina Aleemardani
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, The University of Sheffield, Sheffield S3 7HQ, UK; (M.A.); (M.Z.T.); (N.H.G.)
| | - Michael Zivojin Trikić
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, The University of Sheffield, Sheffield S3 7HQ, UK; (M.A.); (M.Z.T.); (N.H.G.)
| | - Nicola Helen Green
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, The University of Sheffield, Sheffield S3 7HQ, UK; (M.A.); (M.Z.T.); (N.H.G.)
- Insigneo Institute for in Silico Medicine, The Pam Liversidge Building, Sir Robert Hadfield Building, Mappin Street, Sheffield S1 3JD, UK
| | - Frederik Claeyssens
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, The University of Sheffield, Sheffield S3 7HQ, UK; (M.A.); (M.Z.T.); (N.H.G.)
- Correspondence:
| |
Collapse
|
14
|
Yang Z, Hu X, Zhou L, He Y, Zhang X, Yang J, Ju Z, Liou YC, Shen HM, Luo G, Hamblin MR, He W, Yin R. Photodynamic therapy accelerates skin wound healing through promoting re-epithelialization. BURNS & TRAUMA 2021; 9:tkab008. [PMID: 34514005 PMCID: PMC8420953 DOI: 10.1093/burnst/tkab008] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/01/2021] [Indexed: 11/16/2022]
Abstract
Background Epidermal stem cells (EpSCs) that reside in cutaneous hair follicles and the basal layer of the epidermis are indispensable for wound healing and skin homeostasis. Little is known about the effects of photochemical activation on EpSC differentiation, proliferation and migration during wound healing. The present study aimed to determine the effects of photodynamic therapy (PDT) on wound healing in vivo and in vitro. Methods We created mouse full-thickness skin resection models and applied 5-aminolevulinic acid (ALA) for PDT to the wound beds. Wound healing was analysed by gross evaluation and haematoxylin–eosin staining in vivo. In cultured EpSCs, protein expression was measured using flow cytometry and immunohistochemistry. Cell migration was examined using a scratch model; apoptosis and differentiation were measured using flow cytometry. Results PDT accelerated wound closure by enhancing EpSC differentiation, proliferation and migration, thereby promoting re-epithelialization and angiogenesis. PDT inhibited inflammatory infiltration and expression of proinflammatory cytokines, whereas the secretion of growth factors was greater than in other groups. The proportion of transient amplifying cells was significantly greater in vivo and in vitro in the PDT groups. EpSC migration was markedly enhanced after ALA-induced PDT. Conclusions Topical ALA-induced PDT stimulates wound healing by enhancing re-epithelialization, promoting angiogenesis as well as modulating skin homeostasis. This work provides a preliminary theoretical foundation for the clinical administration of topical ALA-induced PDT in skin wound healing.
Collapse
Affiliation(s)
- Zengjun Yang
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xiaohong Hu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Lina Zhou
- Department of Endocrinology, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yaxiong He
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xiaorong Zhang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Jiacai Yang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, No. 601 Huangpu Street, Tianhe District, Guangzhou, Guangdong Province, 510632, China
| | - Yih-Cherng Liou
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, 117543, Singapore
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 14 Science Drive 4, 117543, Singapore
| | - Gaoxing Luo
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, 40 Blossom Street, Boston, MA, 02114, USA
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Rui Yin
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| |
Collapse
|
15
|
Abstract
The skin is a passive and active barrier which protects the body from the environment. Its health is essential for the accomplishment of this role. Since several decades, the skin has aroused a strong interest in various fields (for e.g. cell biology, medicine, toxicology, cosmetology, and pharmacology). In contrast to other organs, 3D models were mostly and directly elaborated in humans due to its architectural simplicity and easy accessibility. The development of these models benefited from the societal pressure to reduce animal experiments. In this review, we first describe human and mouse skin structure and the major differences with other mammals and birds. Next, we describe the different 3D human skin models and their main applications. Finally, we review the available models for domestic animals and discuss the current and potential applications.
Collapse
Affiliation(s)
- Laurent Souci
- ISP, INRAE, Université de Tours, Equipe BioVA, Centre Val de Loire, 37380, Nouzilly, France
| | - Caroline Denesvre
- ISP, INRAE, Université de Tours, Equipe BioVA, Centre Val de Loire, 37380, Nouzilly, France.
| |
Collapse
|
16
|
Chou YT, Lai FJ, Chang NS, Hsu LJ. Wwox Deficiency Causes Downregulation of Prosurvival ERK Signaling and Abnormal Homeostatic Responses in Mouse Skin. Front Cell Dev Biol 2020; 8:558432. [PMID: 33195192 PMCID: PMC7652735 DOI: 10.3389/fcell.2020.558432] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 10/09/2020] [Indexed: 11/24/2022] Open
Abstract
Deficiency of tumor suppressor WW domain-containing oxidoreductase (WWOX) in humans and animals leads to growth retardation and premature death during postnatal developmental stages. Skin integrity is essential for organism survival due to its protection against dehydration and hypothermia. Our previous report demonstrated that human epidermal suprabasal cells express WWOX protein, and the expression is gradually increased toward the superficial differentiated cells prior to cornification. Here, we investigated whether abnormal skin development and homeostasis occur under Wwox deficiency that may correlate with early death. We determined that keratinocyte proliferation and differentiation were decreased, while apoptosis was increased in Wwox–/– mouse epidermis and primary keratinocyte cultures and WWOX-knockdown human HaCaT cells. Without WWOX, progenitor cells in hair follicle junctional zone underwent massive proliferation in early postnatal developmental stages and the stem/progenitor cell pools were depleted at postnatal day 21. These events lead to significantly decreased epidermal thickness, dehydration state, and delayed hair development in Wwox–/– mouse skin, which is associated with downregulation of prosurvival MEK/ERK signaling in Wwox–/– keratinocytes. Moreover, Wwox depletion results in substantial downregulation of dermal collagen contents in mice. Notably, Wwox–/– mice exhibit severe loss of subcutaneous adipose tissue and significant hypothermia. Collectively, our knockout mouse model supports the validity of WWOX in assisting epidermal and adipose homeostasis, and the involvement of prosurvival ERK pathway in the homeostatic responses regulated by WWOX.
Collapse
Affiliation(s)
- Ying-Tsen Chou
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Feng-Jie Lai
- Department of Dermatology, Chimei Medical Center, Tainan, Taiwan.,Center for General Education, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Nan-Shan Chang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
| | - Li-Jin Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
17
|
Rangel-Huerta E, Guzman A, Maldonado E. The dynamics of epidermal stratification during post-larval development in zebrafish. Dev Dyn 2020; 250:175-190. [PMID: 32877571 DOI: 10.1002/dvdy.249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/08/2020] [Accepted: 08/22/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The epidermis, as a defensive barrier, is a consistent trait throughout animal evolution. During post-larval development, the zebrafish epidermis thickens by stratification or addition of new cell layers. Epidermal basal stem cells, expressing the transcription factor p63, are known to be involved in this process. Zebrafish post-larval epidermal stratification is a tractable system to study how stem cells participate in organ growth. METHODS We used immunohistochemistry, in combination with EdU cell proliferation detection, to study zebrafish epidermal stratification. For this procedure, we selected a window of post-larval stages (5-8 mm of standard length or SL, which normalizes age by size). Simultaneously, we used markers for asymmetric cell division and the Notch signaling pathway. RESULTS We found that epidermal stratification is the consequence of several events, including changes in cell shape, active cell proliferation and asymmetrical cell divisions. We identified a subset of highly proliferative epidermal cells with reduced levels of p63, which differed from the basal stem cells with high levels of p63. Additionally, we described different mechanisms that participate in the stratification process, including the phosphorylation of p63, asymmetric cell division regulated by the Par3 and LGN proteins, and expression of Notch genes.
Collapse
Affiliation(s)
- Emma Rangel-Huerta
- EvoDevo Research Group, Unidad de Sistemas Arrecifales, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México (UNAM), Puerto Morelos, Quintana Roo, Mexico.,Posgrado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, UNAM, Puerto Morelos, Quintana Roo, Mexico
| | - Aida Guzman
- EvoDevo Research Group, Unidad de Sistemas Arrecifales, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México (UNAM), Puerto Morelos, Quintana Roo, Mexico.,Estudio Técnico Especializado en Histopatología, Escuela Nacional Preparatoria, ENP, Universidad Nacional Autónoma de México, UNAM, Ciudad de México, Mexico
| | - Ernesto Maldonado
- EvoDevo Research Group, Unidad de Sistemas Arrecifales, Instituto de Ciencias del Mar y Limnología, Universidad Nacional Autónoma de México (UNAM), Puerto Morelos, Quintana Roo, Mexico
| |
Collapse
|
18
|
Abstract
p63 (also known as TP63) is a transcription factor of the p53 family, along with p73. Multiple isoforms of p63 have been discovered and these have diverse functions encompassing a wide array of cell biology. p63 isoforms are implicated in lineage specification, proliferative potential, differentiation, cell death and survival, DNA damage response and metabolism. Furthermore, p63 is linked to human disease states including cancer. p63 is critical to many aspects of cell signaling, and in this Cell science at a glance article and the accompanying poster, we focus on the signaling cascades regulating TAp63 and ΔNp63 isoforms and those that are regulated by TAp63 and ΔNp63, as well the role of p63 in disease.
Collapse
Affiliation(s)
- Matthew L Fisher
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Seamus Balinth
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA.,Stony Brook University, Department of Molecular and Cell Biology, Stony Brook, NY, 11794, USA
| | - Alea A Mills
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| |
Collapse
|
19
|
Yamada T, Hasegawa S, Iwata Y, Arima M, Kobayashi T, Numata S, Nakata S, Sugiura K, Akamatsu H. UV irradiation-induced DNA hypomethylation around WNT1 gene: Implications for solar lentigines. Exp Dermatol 2020; 28:723-729. [PMID: 31020703 DOI: 10.1111/exd.13949] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 03/13/2019] [Accepted: 04/17/2019] [Indexed: 12/13/2022]
Abstract
Wnt/β-catenin signalling promotes melanogenesis in melanocytes and also induces melanocytogenesis from melanocyte stem cells (McSCs). Previous study reported that WNT1, a ligand which activates Wnt/β-catenin signalling pathway, was more highly expressed in the epidermis at SLs than in normal skin areas, suggesting that WNT1 causes hyperpigmentation. To elucidate the mechanism by which WNT1 expression is increased in SLs, we examined the methylation of 5-carbon of cytosine (5mC), that is 5-methylcytosine (5mC) level, in a region within the WNT1 promoter; the methylation of the region was known to negatively regulate WNT1 gene expression. We used an immortalized cell line of human interfollicular epidermal stem cells to analyse the effect of UVB irradiation on DNA methylation level of WNT1 promoter and found that UVB irradiation caused demethylation of WNT1 promoter and promoted WNT1 mRNA expression. It was also found that UVB irradiation reduced the expression of DNA methyltransferase 1 (DNMT1), an enzyme responsible for maintaining methylation patterns during cell division. Pathological analysis of SLs and non-SL regions in the human skin revealed that both DNMT1 expression and 5mC level were decreased at SLs compared to non-SL skins. Furthermore, bisulphite sequencing showed that the methylated CpG level in WNT1 promoter was also lower at SLs than in non-SL skins. Thus, in the skin exposed to a high amount of UV rays, excessive expression of WNT1 is thought to be caused by the demethylation of WNT1 promoter, and the upregulated WNT1 promotes melanocytogenesis and melanogenesis, then resulting in SL formation.
Collapse
Affiliation(s)
- Takaaki Yamada
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd., Nagoya, Japan.,Department of Applied Cell and Regenerative Medicine, Fujita Health University School of Medicine, Toyoake, Japan.,Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Seiji Hasegawa
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd., Nagoya, Japan.,Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan.,Nagoya University-MENARD Collaborative Chair, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yohei Iwata
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Masaru Arima
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Tsukane Kobayashi
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Shigeki Numata
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Satoru Nakata
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd., Nagoya, Japan
| | - Kazumitsu Sugiura
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hirohiko Akamatsu
- Department of Applied Cell and Regenerative Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| |
Collapse
|
20
|
Xu P, Wu Y, Zhou L, Yang Z, Zhang X, Hu X, Yang J, Wang M, Wang B, Luo G, He W, Cheng B. Platelet-rich plasma accelerates skin wound healing by promoting re-epithelialization. BURNS & TRAUMA 2020; 8:tkaa028. [PMID: 32821743 PMCID: PMC7427034 DOI: 10.1093/burnst/tkaa028] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/24/2020] [Indexed: 01/19/2023]
Abstract
BACKGROUND Autologous platelet-rich plasma (PRP) has been suggested to be effective for wound healing. However, evidence for its use in patients with acute and chronic wounds remains insufficient. The aims of this study were to comprehensively examine the effectiveness, synergy and possible mechanism of PRP-mediated improvement of acute skin wound repair. METHODS Full-thickness wounds were made on the back of C57/BL6 mice. PRP or saline solution as a control was administered to the wound area. Wound healing rate, local inflammation, angiogenesis, re-epithelialization and collagen deposition were measured at days 3, 5, 7 and 14 after skin injury. The biological character of epidermal stem cells (ESCs), which reflect the potential for re-epithelialization, was further evaluated in vitro and in vivo. RESULTS PRP strongly improved skin wound healing, which was associated with regulation of local inflammation, enhancement of angiogenesis and re-epithelialization. PRP treatment significantly reduced the production of inflammatory cytokines interleukin-17A and interleukin-1β. An increase in the local vessel intensity and enhancement of re-epithelialization were also observed in animals with PRP administration and were associated with enhanced secretion of growth factors such as vascular endothelial growth factor and insulin-like growth factor-1. Moreover, PRP treatment ameliorated the survival and activated the migration and proliferation of primary cultured ESCs, and these effects were accompanied by the differentiation of ESCs into adult cells following the changes of CD49f and keratin 10 and keratin 14. CONCLUSION PRP improved skin wound healing by modulating inflammation and increasing angiogenesis and re-epithelialization. However, the underlying regulatory mechanism needs to be investigated in the future. Our data provide a preliminary theoretical foundation for the clinical administration of PRP in wound healing and skin regeneration.
Collapse
Affiliation(s)
- Pengcheng Xu
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command, PLA, Guangzhou, China
| | - Yaguang Wu
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lina Zhou
- Department of Endocrinology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zengjun Yang
- Department of Dermatology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaorong Zhang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Xiaohong Hu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Jiacai Yang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Mingying Wang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Binjie Wang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Gaoxing Luo
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing 400038, China
| | - Biao Cheng
- Department of Burn and Plastic Surgery, General Hospital of Southern Theater Command, PLA, Guangzhou, China
| |
Collapse
|
21
|
Sukmawati D, Eryani A, Damayanti L. Silver Sulfadiazine's Effect on Keratin-19 Expression as Stem Cell Marker in Burn Wound Healing. Biomedicine (Taipei) 2020; 10:5-11. [PMID: 33854915 PMCID: PMC7608848 DOI: 10.37796/2211-8039.1014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/13/2019] [Indexed: 10/25/2022] Open
Abstract
BACKGROUND Burn wounds are one of the causes of cutaneous injury that involve both epidermal and dermal layers of skin. Silver sulfadiazine (SSD) has been widely used to treat burn wounds, however recent studies have found the treatment to have some drawbacks, such as cellular toxicity effects. Cutaneous wound regeneration is known to start from the basal layer of the epidermal epithelial cells, which are enriched with highly proliferative cells. Keratin-19 (K19) is one of the epidermal stem cell biomarkers found in the skin. This study aims to explore the expression of K19 in burn wound tissue and to investigate the effect of SSD on its expression. METHODS We created a burn wound model in Sprague Dawley rats and randomly divided them into control and SSD groups. Wound closure was evaluated (visitrak) overtime series followed by histological evaluation of K19 expression in the wound tissue (immunohistochemistry staining). RESULTS Our model successfully represents full-thickness damage caused by a burn wound. The SSD group showed a faster reduction of wound surface area (wound closure) compared to the control group with the peak at day 18 post wounding (p < 0.05). K19 expression was found in both groups and was distributed on epidermal layers, hair follicles and dermis of granulation tissue showing similar patterns. CONCLUSION Topical application of SSD on burn wounds showed superiority in wound closure and is likely to have no harmful effect on epidermal stem cells. However, further study is required to investigate the effect of silver species on cell viability and toxicity effects during long term treatment.
Collapse
Affiliation(s)
- Dewi Sukmawati
- Department of Histology, Faculty of Medicine Universitas Indonesia, Jln. Salemba Raya No. 6 Jakarta, 10430, Jakarta, Indonesia
| | - Astheria Eryani
- Department of Histology, Faculty of Medicine Tarumanagara University, Jln. Letjen S. Parman No.1, Tomang, Grogol Petamburan, Jakarta, 11440, Indonesia
| | - Lia Damayanti
- Department of Histology, Faculty of Medicine Universitas Indonesia, Jln. Salemba Raya No. 6 Jakarta, 10430, Jakarta, Indonesia
| |
Collapse
|
22
|
Effect of photobiomodulation on cellular migration and survival in diabetic and hypoxic diabetic wounded fibroblast cells. Lasers Med Sci 2020; 36:365-374. [PMID: 32483750 DOI: 10.1007/s10103-020-03041-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/17/2020] [Indexed: 10/24/2022]
Abstract
A disrupted wound repair process often leads to the development of chronic wounds, and pose a major physical, social and economic inconvenience on patients and the public health sector. Chronic wounds are a common complication seen in diabetes mellitus (DM), and often the severity necessitates amputation of the lower limbs. Recently, there has been increasing evidence that photobiomodulation (PBM) initiates wound healing, including increased protein transcription for cell proliferation, viability, migration and tissue reepithelialisation. Here, the hypothesis that PBM at a wavelength of 660 nm and energy density of 5 J/cm2 regulates wound repair in diabetic wounded and hypoxic diabetic wounded fibroblasts by enhancing cell migration and survival was investigated. PBM increased migration and survival in diabetic wounded and hypoxic diabetic wounded fibroblasts. Our findings suggest that PBM enhances migration and survival in diabetic wounded and hypoxic diabetic wounded fibroblasts, indicating that this therapeutic method may be beneficial against chronic wounds in diabetic patients.
Collapse
|
23
|
Anti-Aging Effects of GDF11 on Skin. Int J Mol Sci 2020; 21:ijms21072598. [PMID: 32283613 PMCID: PMC7177281 DOI: 10.3390/ijms21072598] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/02/2020] [Accepted: 04/07/2020] [Indexed: 12/24/2022] Open
Abstract
Human skin is composed of three layers: the epidermis, the dermis, and the hypodermis. The epidermis has four major cell layers made up of keratinocytes in varying stages of progressive differentiation. Skin aging is a multi-factorial process that affects every phase of its biology and function. The expression profiles of inflammation-related genes analyzed in resident immune cells demonstrated that these cells have a strong ability to regenerate adult skin stem cells and to produce endogenous substances such as growth differentiation factor 11 (GDF11). GDF11 appears to be the key to progenitor proliferation and/or differentiation. The preservation of youthful phenotypes has been tied to the presence of GDF11 in different human tissues, and, in the skin, this factor inhibits inflammatory responses. The protective role of GDF11 depends on a multi-factorial process implicating various types of skin cells such as keratinocytes, fibroblasts and inflammatory cells. GDF11 should be further studied for the purpose of developing novel therapies for the treatment of skin diseases.
Collapse
|
24
|
Yang GN, Strudwick XL, Bonder C, Kopecki Z, Cowin AJ. Effect of Flightless I Expression on Epidermal Stem Cell Niche During Wound Repair. Adv Wound Care (New Rochelle) 2020; 9:161-173. [PMID: 32117580 DOI: 10.1089/wound.2018.0884] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 06/03/2019] [Indexed: 12/31/2022] Open
Abstract
Objective: Activation of epidermal stem cells (EpSCs) from their quiescent niche is an integral component of wound reepithelialization and involves Wnt/β-catenin (β-Cat) signaling and remodeling of the actin cytoskeleton. The aim of this study was to investigate the effect of Flightless I (Flii), a cytoskeletal protein and inhibitor of wound healing, on EpSC activation during wound repair. Approach: Genetically modified Flii mice (Flii knockdown: Flii+/- , wild type: WT, Flii overexpressing: FliiTg/Tg ) received two incisional wounds along the lateral axis of the dorsal skin. Indicators of EpSC activation (epidermal growth factor receptor 1 [EGFR1], leucine-rich repeats and immunoglobulin-like domains-1 [Lrig1], K14), Wnt/β-Cat signaling (Lgr6, Flap2, β-Cat, and axis inhibition protein 2 [Axin2]), and cell proliferation (proliferating cell nuclear antigen [PCNA]) were assessed using immunohistochemistry. β-Cat stabilization was examined using western blotting with cell cycling and differentiation of isolated CD34+ITGA6high EpSCs examined using real time-quantitative polymerase chain reaction after treatment with wound-conditioned media. Results: Flii+/- led to increased numbers of activated EpSCs expressing PCNA, elevated EGFR1, and decreased Lrig1. EpSCs in Flii+/- hair follicle niches adjacent to the wounds also showed expression of Wnt-activation markers including increased β-Cat and Lgr6, and decreased Axin2. EpSCs (CD34+ITGA6high) isolated from Flii+/- unwounded skin showed elevated expression of cell-cycling genes including ΔNp63, filaggrin (Fila), involucrin (Invo), cyclin D1 (Ccnd1), and cell-division cycle protein-20 (Cdc20); and elevated ΔNp63 and Invo after treatment with wound-conditioned media compared with WT and FliiTg/Tg counterparts. Innovation: Flii was identified as an inhibitor of EpSC activation that may explain its negative effects on wound reepithelialization. Conclusion: Flii may inhibit EpSC activation by interrupting Wnt/β-Cat signaling. Strategies that reduce Flii may increase activation of EpSCs and promote reepithelialization of wounds.
Collapse
Affiliation(s)
- Gink N. Yang
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Xanthe L. Strudwick
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Claudine Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Zlatko Kopecki
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - Allison J. Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
25
|
Mazini L, Rochette L, Admou B, Amal S, Malka G. Hopes and Limits of Adipose-Derived Stem Cells (ADSCs) and Mesenchymal Stem Cells (MSCs) in Wound Healing. Int J Mol Sci 2020; 21:E1306. [PMID: 32075181 PMCID: PMC7072889 DOI: 10.3390/ijms21041306] [Citation(s) in RCA: 309] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 12/11/2022] Open
Abstract
Adipose tissue derived stem cells (ADSCs) are mesenchymal stem cells identified within subcutaneous tissue at the base of the hair follicle (dermal papilla cells), in the dermal sheets (dermal sheet cells), in interfollicular dermis, and in the hypodermis tissue. These cells are expected to play a major role in regulating skin regeneration and aging-associated morphologic disgraces and structural deficits. ADSCs are known to proliferate and differentiate into skin cells to repair damaged or dead cells, but also act by an autocrine and paracrine pathway to activate cell regeneration and the healing process. During wound healing, ADSCs have a great ability in migration to be recruited rapidly into wounded sites added to their differentiation towards dermal fibroblasts (DF), endothelial cells, and keratinocytes. Additionally, ADSCs and DFs are the major sources of the extracellular matrix (ECM) proteins involved in maintaining skin structure and function. Their interactions with skin cells are involved in regulating skin homeostasis and during healing. The evidence suggests that their secretomes ensure: (i) The change in macrophages inflammatory phenotype implicated in the inflammatory phase, (ii) the formation of new blood vessels, thus promoting angiogenesis by increasing endothelial cell differentiation and cell migration, and (iii) the formation of granulation tissues, skin cells, and ECM production, whereby proliferation and remodeling phases occur. These characteristics would be beneficial to therapeutic strategies in wound healing and skin aging and have driven more insights in many clinical investigations. Additionally, it was recently presented as the tool key in the new free-cell therapy in regenerative medicine. Nevertheless, ADSCs fulfill the general accepted criteria for cell-based therapies, but still need further investigations into their efficiency, taking into consideration the host-environment and patient-associated factors.
Collapse
Affiliation(s)
- Loubna Mazini
- Laboratoire Cellules Souches et Régénération Cellulaire et Tissulaire, Centre interface Applications Médicales (CIAM), Université Mohammed VI Polytechnique, Ben-Guerir 43 150, Morocco;
| | - Luc Rochette
- Equipe d’Accueil (EA 7460), Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Faculté des Sciences de Santé Université de Bourgogne—Franche Comté, 7 Bd Jeanne d’Arc, 21000 Dijon, France;
| | - Brahim Admou
- Laboratoire d’immunologie, Centre de Recherche Clinique, Faculté de Médecine et Pharmacie, Université Cadi Ayyad, Centre Hospitalier Universitaire, Marrakech 40 000, Morocco;
| | - Said Amal
- Service de dermatologie, Faculté de Médecine et Pharmacie, Université Cadi Ayyad, Centre hospitalier universitaire, Marrakech 40000, Morocco;
| | - Gabriel Malka
- Laboratoire Cellules Souches et Régénération Cellulaire et Tissulaire, Centre interface Applications Médicales (CIAM), Université Mohammed VI Polytechnique, Ben-Guerir 43 150, Morocco;
| |
Collapse
|
26
|
Scieglinska D, Krawczyk Z, Sojka DR, Gogler-Pigłowska A. Heat shock proteins in the physiology and pathophysiology of epidermal keratinocytes. Cell Stress Chaperones 2019; 24:1027-1044. [PMID: 31734893 PMCID: PMC6882751 DOI: 10.1007/s12192-019-01044-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/09/2019] [Accepted: 10/15/2019] [Indexed: 12/21/2022] Open
Abstract
Heat shock proteins (HSPs), a large group of highly evolutionary conserved proteins, are considered to be main elements of the cellular proteoprotection system. HSPs are encoded by genes activated during the exposure of cells to proteotoxic factors, as well as by genes that are expressed constitutively under physiological conditions. HSPs, having properties of molecular chaperones, are involved in controlling/modulation of multiple cellular and physiological processes. In the presented review, we summarize the current knowledge on HSPs in the biology of epidermis, the outer skin layer composed of stratified squamous epithelium. This tissue has a vital barrier function preventing from dehydratation due to passive diffusion of water out of the skin, and protecting from infection and other environmental insults. We focused on HSPB1 (HSP27), HSPA1 (HSP70), HSPA2, and HSPC (HSP90), because only these HSPs have been studied in the context of physiology and pathophysiology of the epidermis. The analysis of literature data shows that HSPB1 plays a role in the regulation of final steps of keratinization; HSPA1 is involved in the cytoprotection, HSPA2 contributes to the early steps of keratinocyte differentiation, while HSPC is essential in the re-epithelialization process. Since HSPs have diverse functions in various types of somatic tissues, in spite of multiple investigations, open questions still remain about detailed roles of a particular HSP isoform in the biology of epidermal keratinocytes.
Collapse
Affiliation(s)
- Dorota Scieglinska
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Institute-Oncology Center Gliwice Branch, ul. Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland.
| | - Zdzisław Krawczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Institute-Oncology Center Gliwice Branch, ul. Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
| | - Damian Robert Sojka
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Institute-Oncology Center Gliwice Branch, ul. Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
| | - Agnieszka Gogler-Pigłowska
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie Institute-Oncology Center Gliwice Branch, ul. Wybrzeże Armii Krajowej 15, 44-101, Gliwice, Poland
| |
Collapse
|
27
|
Aljagthmi AA, Hill NT, Cooke M, Kazanietz MG, Abba MC, Long W, Kadakia MP. ΔNp63α suppresses cells invasion by downregulating PKCγ/Rac1 signaling through miR-320a. Cell Death Dis 2019; 10:680. [PMID: 31515469 PMCID: PMC6742631 DOI: 10.1038/s41419-019-1921-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/20/2019] [Accepted: 08/26/2019] [Indexed: 01/02/2023]
Abstract
ΔNp63α, a member of the p53 family of transcription factors, is overexpressed in a number of cancers and plays a role in proliferation, differentiation, migration, and invasion. ΔNp63α has been shown to regulate several microRNAs that are involved in development and cancer. We identified miRNA miR-320a as a positively regulated target of ΔNp63α. Previous studies have shown that miR-320a is downregulated in colorectal cancer and targets the small GTPase Rac1, leading to a reduction in noncanonical WNT signaling and EMT, thereby inhibiting tumor metastasis and invasion. We showed that miR-320a is a direct target of ΔNp63α. Knockdown of ΔNp63α in HaCaT and A431 cells downregulates miR-320a levels and leads to a corresponding elevation in PKCγ transcript and protein levels. Rac1 phosphorylation at Ser71 was increased in the absence of ΔNp63α, whereas overexpression of ΔNp63α reversed S71 phosphorylation of Rac1. Moreover, increased PKCγ levels, Rac1 phosphorylation and cell invasion observed upon knockdown of ΔNp63α was reversed by either overexpressing miR-320a mimic or Rac1 silencing. Finally, silencing PKCγ or treatment with the PKC inhibitor Gö6976 reversed increased Rac1 phosphorylation and cell invasion observed upon silencing ΔNp63α. Taken together, our data suggest that ΔNp63α positively regulates miR-320a, thereby inhibiting PKCγ expression, Rac1 phosphorylation, and cancer invasion.
Collapse
Affiliation(s)
- Amjad A Aljagthmi
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, Dayton, OH, 45435, USA
| | - Natasha T Hill
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, Dayton, OH, 45435, USA
| | - Mariana Cooke
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Marcelo G Kazanietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Martín C Abba
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Universidad Nacional de La Plata, CP1900, La Plata, Argentina
| | - Weiwen Long
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, Dayton, OH, 45435, USA
| | - Madhavi P Kadakia
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, Dayton, OH, 45435, USA.
| |
Collapse
|
28
|
De Rosa L, Secone Seconetti A, De Santis G, Pellacani G, Hirsch T, Rothoeft T, Teig N, Pellegrini G, Bauer JW, De Luca M. Laminin 332-Dependent YAP Dysregulation Depletes Epidermal Stem Cells in Junctional Epidermolysis Bullosa. Cell Rep 2019; 27:2036-2049.e6. [PMID: 31091444 DOI: 10.1016/j.celrep.2019.04.055] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/12/2019] [Accepted: 04/10/2019] [Indexed: 01/09/2023] Open
Abstract
Laminin 332-deficient junctional epidermolysis bullosa (JEB) is a severe genetic skin disease. JEB is marked by epidermal stem cell depletion, the origin of which is unknown. We show that dysregulation of the YAP and TAZ pathway underpins such stem cell depletion. Laminin 332-mediated YAP activity sustains human epidermal stem cells, detected as holoclones. Ablation of YAP selectively depletes holoclones, while enforced YAP blocks conversion of stem cells into progenitors and indefinitely extends the keratinocyte lifespan. YAP is dramatically decreased in JEB keratinocytes, which contain only phosphorylated, inactive YAP. In normal keratinocytes, laminin 332 and α6β4 ablation abolish YAP activity and recapitulate the JEB phenotype. In JEB keratinocytes, laminin 332-gene therapy rescues YAP activity and epidermal stem cells in vitro and in vivo. In JEB cells, enforced YAP recapitulates laminin 332-gene therapy, thus uncoupling adhesion from proliferation in epidermal stem cells. This work has important clinical implication for ex vivo gene therapy of JEB.
Collapse
Affiliation(s)
- Laura De Rosa
- Centre for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessia Secone Seconetti
- Centre for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giorgio De Santis
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanni Pellacani
- Department of Surgery, Medicine, Dentistry, and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Tobias Hirsch
- Department of Plastic Surgery, Burn Centre, BG University Hospital Bergmannsheil, Ruhr-University Bochum, Germany
| | - Tobias Rothoeft
- Department of Neonatology and Pediatric Intensive Care, University Children's Hospital, Ruhr-University Bochum, Germany
| | - Norbert Teig
- Department of Neonatology and Pediatric Intensive Care, University Children's Hospital, Ruhr-University Bochum, Germany
| | - Graziella Pellegrini
- Centre for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy; Department of Surgery, Medicine, Dentistry, and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Johann W Bauer
- EB House Austria and Department of Dermatology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Michele De Luca
- Centre for Regenerative Medicine "Stefano Ferrari," Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
29
|
Prodinger CM, Reichelt J, Bauer JW, Laimer M. Current and Future Perspectives of Stem Cell Therapy in Dermatology. Ann Dermatol 2017; 29:667-687. [PMID: 29200755 PMCID: PMC5705348 DOI: 10.5021/ad.2017.29.6.667] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 09/05/2017] [Indexed: 12/19/2022] Open
Abstract
Stem cells are undifferentiated cells capable of generating, sustaining, and replacing terminally differentiated cells and tissues. They can be isolated from embryonic as well as almost all adult tissues including skin, but are also generated through genetic reprogramming of differentiated cells. Preclinical and clinical research has recently tremendously improved stem cell therapy, being a promising treatment option for various diseases in which current medical therapies fail to cure, prevent progression or relieve symptoms. With the main goal of regeneration or sustained genetic correction of damaged tissue, advanced tissue-engineering techniques are especially applicable for many dermatological diseases including wound healing, genodermatoses (like the severe blistering disorder epidermolysis bullosa) and chronic (auto-)inflammatory diseases. This review summarizes general aspects as well as current and future perspectives of stem cell therapy in dermatology.
Collapse
Affiliation(s)
- Christine M Prodinger
- Department of Dermatology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Julia Reichelt
- Department of Dermatology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Johann W Bauer
- Department of Dermatology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Martin Laimer
- Department of Dermatology, Paracelsus Medical University of Salzburg, Salzburg, Austria
| |
Collapse
|
30
|
Transit-Amplifying Cells in the Fast Lane from Stem Cells towards Differentiation. Stem Cells Int 2017; 2017:7602951. [PMID: 28835754 PMCID: PMC5556613 DOI: 10.1155/2017/7602951] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/23/2017] [Accepted: 07/11/2017] [Indexed: 12/13/2022] Open
Abstract
Stem cells have a high potential to impact regenerative medicine. However, stem cells in adult tissues often proliferate at very slow rates. During development, stem cells may change first to a pluripotent and highly proliferative state, known as transit-amplifying cells. Recent advances in the identification and isolation of these undifferentiated and fast-dividing cells could bring new alternatives for cell-based transplants. The skin epidermis has been the target of necessary research about transit-amplifying cells; this work has mainly been performed in mammalian cells, but further work is being pursued in other vertebrate models, such as zebrafish. In this review, we present some insights about the molecular repertoire regulating the transition from stem cells to transit-amplifying cells or playing a role in the transitioning to fully differentiated cells, including gene expression profiles, cell cycle regulation, and cellular asymmetrical events. We also discuss the potential use of this knowledge in effective progenitor cell-based transplants in the treatment of skin injuries and chronic disease.
Collapse
|
31
|
Wound Healing from Dermal Grafts Containing CD34+ Cells Is Comparable to Wound Healing with Split-Thickness Skin Micrografts. Plast Reconstr Surg 2017; 140:306-314. [DOI: 10.1097/prs.0000000000003516] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Wang P, Shu B, Xu Y, Zhu J, Liu J, Zhou Z, Chen L, Zhao J, Liu X, Qi S, Xiong K, Xie J. Basic fibroblast growth factor reduces scar by inhibiting the differentiation of epidermal stem cells to myofibroblasts via the Notch1/Jagged1 pathway. Stem Cell Res Ther 2017; 8:114. [PMID: 28511663 PMCID: PMC5434520 DOI: 10.1186/s13287-017-0549-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/28/2017] [Accepted: 03/31/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Basic fibroblast growth factor (bFGF) plays an important role in promoting wound healing and reducing scar, but the possible molecular mechanisms are still unclear. Our previous studies have found that activating the Notch1/Jagged1 pathway can inhibit the differentiation of epidermal stem cells (ESCs) to myofibroblasts (MFB). Herein, we document that bFGF reduces scar by inhibiting the differentiation of ESCs to MFB via activating the Notch1/Jagged1 pathway. METHODS In in-vitro study, ESCs were isolated from 10 neonatal SD rats (1-3 days old), cultured in keratinocyte serum-free medium, and divided into six groups: bFGF group, bFGF + SU5402 group, bFGF + DAPT group, siJagged1 group, bFGF + siJagged1 group, and control group. Jagged1 of the ESCs in the siJagged1 group and bFGF + siJagged1 group was knocked down by small-interfering RNA transfection. Expression of ESC markers (CK15/CK10), MFB markers (α-SMA, Collagen I, Collagen III), and Notch1/Jagged1 components (Jagged1, Notch1, Hes1) was detected by FCM, qRT-PCR, and western blot analysis to study the relationships of bFGF, ESCs, and Notch1/Jagged1 pathway. In in-vivo study, the wound healing time and scar hyperplasia were observed on rabbit ear scar models. The quality of wound healing was estimated by hematoxylin and eosin staining and Masson staining. Expression of ESC markers, MFB markers and Notch1/Jagged1 components was elucidated by immunohistochemistry, immunofluorescence, and western blot analysis. RESULTS The in-vitro study showed that bFGF could significantly upregulate the expression of ESC markers and Notch1/Jagged1 components, while downregulating the expression of MFB markers at the same time. However, these effects could be obviously decreased when we knocked down Jagged1 or added DAPT. Similarly, in in-vivo study, bFGF also exhibited its functions in inhibiting the differentiation of rabbit ESCs to MFB by activating the Notch1/Jagged1 pathway, which improved the wound healing quality and alleviated scar significantly. CONCLUSION These results provide evidence that bFGF can reduce scar by inhibiting the differentiation of ESCs to MFB via the Notch1/Jagged1 pathway, and present a new promising potential direction for the treatment of scar.
Collapse
Affiliation(s)
- Peng Wang
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Bin Shu
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Yingbin Xu
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Jiayuan Zhu
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Jian Liu
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Ziheng Zhou
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Lei Chen
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Jingling Zhao
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Xusheng Liu
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Shaohai Qi
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province 410013 People’s Republic of China
| | - Julin Xie
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Yuexiu District, Guangzhou, Guangdong Province 510080 People’s Republic of China
| |
Collapse
|
33
|
Yao Z, Li H, He W, Yang S, Zhang X, Zhan R, Xu R, Tan J, Zhou J, Wu J, Luo G. P311 Accelerates Skin Wound Reepithelialization by Promoting Epidermal Stem Cell Migration Through RhoA and Rac1 Activation. Stem Cells Dev 2017; 26:451-460. [PMID: 27927130 DOI: 10.1089/scd.2016.0249] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Zhihui Yao
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
- People's Liberation Army Hospital 59, Kaiyuan, China
| | - Haisheng Li
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Sisi Yang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xiaorong Zhang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Rixing Zhan
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Rui Xu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jianglin Tan
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Junyi Zhou
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jun Wu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Gaoxing Luo
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
34
|
Yang GN, Kopecki Z, Cowin AJ. Role of Actin Cytoskeleton in the Regulation of Epithelial Cutaneous Stem Cells. Stem Cells Dev 2016; 25:749-59. [PMID: 27021878 DOI: 10.1089/scd.2016.0051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cutaneous stem cells (CSCs) orchestrate the homeostasis and regeneration of mammalian skin. Epithelial CSCs have been isolated and characterized from the skin and hold great potential for tissue engineering and clinical applications. The actin cytoskeleton is known to regulate cell adhesion and motility through its intricate participation in signal transduction and structural modifications. The dynamics of actin cytoskeleton can directly influence CSCs behaviors including tissue morphogenesis, homeostasis, niche maintenance, activation, and wound repair. Various regulators of the actin cytoskeleton including kinases, actin-remodeling proteins, paracrine signals, and micro-RNAs collaborate and contribute to epithelial CSC proliferation, adhesion, and differentiation. This review brings together the latest mechanistic insights into how the actin cytoskeleton participates in the regulation of epithelial CSCs during development, homeostasis, and wound repair.
Collapse
Affiliation(s)
- Gink N Yang
- Future Industries Institute, University of South Australia , Adelaide, South Australia, Australia
| | - Zlatko Kopecki
- Future Industries Institute, University of South Australia , Adelaide, South Australia, Australia
| | - Allison J Cowin
- Future Industries Institute, University of South Australia , Adelaide, South Australia, Australia
| |
Collapse
|
35
|
Epigenetic Regulation of Epidermal Stem Cell Biomarkers and Their Role in Wound Healing. Int J Mol Sci 2015; 17:ijms17010016. [PMID: 26712738 PMCID: PMC4730263 DOI: 10.3390/ijms17010016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 12/11/2015] [Accepted: 12/16/2015] [Indexed: 12/11/2022] Open
Abstract
As an actively renewable tissue, changes in skin architecture are subjected to the regulation of stem cells that maintain the population of cells responsible for the formation of epidermal layers. Stems cells retain their self-renewal property and express biomarkers that are unique to this population. However, differential regulation of the biomarkers can initiate the pathway of terminal cell differentiation. Although, pockets of non-clarity in stem cell maintenance and differentiation in skin still exist, the influence of epigenetics in epidermal stem cell functions and differentiation in skin homeostasis and wound healing is clearly evident. The focus of this review is to discuss the epigenetic regulation of confirmed and probable epidermal stem cell biomarkers in epidermal stratification of normal skin and in diseased states. The role of epigenetics in wound healing, especially in diseased states of diabetes and cancer, will also be conveyed.
Collapse
|
36
|
Gozali MV, Zhou B. Effective treatments of atrophic acne scars. THE JOURNAL OF CLINICAL AND AESTHETIC DERMATOLOGY 2015; 8:33-40. [PMID: 26029333 PMCID: PMC4445894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Atrophic scarring is often an unfortunate and permanent complication of acne vulgaris. It has high prevalence, significant impact on quality of life, and therapeutic challenge for dermatologists. The treatment of atrophic acne scars varies depending on the types of acne scars and the limitations of the treatment modalities in their ability to improve scars. Therefore, many options are available for the treatment of acne scarring, including chemical peeling, dermabrasion, laser treatment, punch techniques, fat transplantation, other tissue augmenting agents, needling, subcision, and combined therapy. Various modalities have been used to treat scars, but limited efficacy and problematic side effects have restricted their application. In order to optimally treat a patient's scar, we need to consider which treatment offers the most satisfactory result. There are also promising procedures in the future, such as stem cell therapy. In this article, the authors review the different treatment options of atrophic acne scars. This may be useful for selecting the best therapeutic strategy, whether it be single or combined therapy, in the treatment of atrophic acne scars while reducing or avoiding the side effects and complications.
Collapse
Affiliation(s)
- Maya Valeska Gozali
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bingrong Zhou
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
37
|
Ido Y, Duranton A, Lan F, Weikel KA, Breton L, Ruderman NB. Resveratrol prevents oxidative stress-induced senescence and proliferative dysfunction by activating the AMPK-FOXO3 cascade in cultured primary human keratinocytes. PLoS One 2015; 10:e0115341. [PMID: 25647160 PMCID: PMC4315597 DOI: 10.1371/journal.pone.0115341] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/21/2014] [Indexed: 01/27/2023] Open
Abstract
The aging process is perceived as resulting from a combination of intrinsic factors such as changes in intracellular signaling and extrinsic factors, most notably environmental stressors. In skin, the relationship between intrinsic changes and keratinocyte function is not clearly understood. Previously, we found that increasing the activity of AMP-activated protein kinase (AMPK) suppressed senescence in hydrogen peroxide (H2O2)-treated human primary keratinocytes, a model of oxidative stress-induced cellular aging. Using this model in the present study, we observed that resveratrol, an agent that increases the activities of both AMPK and sirtuins, ameliorated two age-associated phenotypes: cellular senescence and proliferative dysfunction. In addition, we found that treatment of keratinocytes with Ex527, a specific inhibitor of sirtuin 1 (SIRT1), attenuated the ability of resveratrol to suppress senescence. In keeping with the latter observation, we noted that compared to non-senescent keratinocytes, senescent cells lacked SIRT1. In addition to these effects on H2O2-induced senescence, resveratrol also prevented the H2O2-induced decrease in proliferation (as indicated by 3H-thymidine incorporation) in the presence of insulin. This effect was abrogated by inhibition of AMPK but not SIRT1. Compared to endothelium, we found that human keratinocytes expressed relatively high levels of Forkhead box O3 (FOXO3), a downstream target of both AMPK and SIRT1. Treatment of keratinocytes with resveratrol transactivated FOXO3 and increased the expression of its target genes including catalase. Resveratrol’s effects on both senescence and proliferation disappeared when FOXO3 was knocked down. Finally, we performed an exploratory study which showed that skin from humans over 50 years old had lower AMPK activity than skin from individuals under age 20. Collectively, these findings suggest that the effects of resveratrol on keratinocyte senescence and proliferation are regulated by the AMPK-FOXO3 pathway and in some situations, but not all, by SIRT1.
Collapse
Affiliation(s)
- Yasuo Ido
- Diabetes and Metabolism Unit, Boston University Medical Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| | | | - Fan Lan
- Endocrinology, Second Affiliated Hospital Chongqing Medical University, Chongqing, China
| | - Karen A. Weikel
- Diabetes and Metabolism Unit, Boston University Medical Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Lionel Breton
- L’OREAL Research and Innovation, Aulnay sous bois, France
| | - Neil B. Ruderman
- Diabetes and Metabolism Unit, Boston University Medical Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
38
|
Suzuki D, Sahu R, Leu NA, Senoo M. The carboxy-terminus of p63 links cell cycle control and the proliferative potential of epidermal progenitor cells. Development 2014; 142:282-90. [PMID: 25503409 DOI: 10.1242/dev.118307] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The transcription factor p63 (Trp63) plays a key role in homeostasis and regeneration of the skin. The p63 gene is transcribed from dual promoters, generating TAp63 isoforms with growth suppressive functions and dominant-negative ΔNp63 isoforms with opposing properties. p63 also encodes multiple carboxy (C)-terminal variants. Although mutations of C-terminal variants have been linked to the pathogenesis of p63-associated ectodermal disorders, the physiological role of the p63 C-terminus is poorly understood. We report here that deletion of the p63 C-terminus in mice leads to ectodermal malformation and hypoplasia, accompanied by a reduced proliferative capacity of epidermal progenitor cells. Notably, unlike the p63-null condition, we find that p63 C-terminus deficiency promotes expression of the cyclin-dependent kinase inhibitor p21(Waf1/Cip1) (Cdkn1a), a factor associated with reduced proliferative capacity of both hematopoietic and neuronal stem cells. These data suggest that the p63 C-terminus plays a key role in the cell cycle progression required to maintain the proliferative potential of stem cells of many different lineages. Mechanistically, we show that loss of Cα, the predominant C-terminal p63 variant in epithelia, promotes the transcriptional activity of TAp63 and also impairs the dominant-negative activity of ΔNp63, thereby controlling p21(Waf1/Cip1) expression. We propose that the p63 C-terminus links cell cycle control and the proliferative potential of epidermal progenitor cells via mechanisms that equilibrate TAp63 and ΔNp63 isoform function.
Collapse
Affiliation(s)
- Daisuke Suzuki
- Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Raju Sahu
- Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - N Adrian Leu
- Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Makoto Senoo
- Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
39
|
Gemini-Piperni S, Takamori ER, Sartoretto SC, Paiva KBS, Granjeiro JM, de Oliveira RC, Zambuzzi WF. Cellular behavior as a dynamic field for exploring bone bioengineering: a closer look at cell-biomaterial interface. Arch Biochem Biophys 2014; 561:88-98. [PMID: 24976174 DOI: 10.1016/j.abb.2014.06.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 06/14/2014] [Accepted: 06/18/2014] [Indexed: 01/09/2023]
Abstract
Bone is a highly dynamic and specialized tissue, capable of regenerating itself spontaneously when afflicted by minor injuries. Nevertheless, when major lesions occur, it becomes necessary to use biomaterials, which are not only able to endure the cellular proliferation and migration, but also to substitute the original tissue or integrate itself to it. With the life expectancy growth, regenerative medicine has been gaining constant attention in the reconstructive field of dentistry and orthopedy. Focusing on broadening the therapeutic possibilities for the regeneration of injured organs, the development of biomaterials allied with the applicability of gene therapy and bone bioengineering has been receiving vast attention over the recent years. The progress of cellular and molecular biology techniques gave way to new-guided therapy possibilities. Supported by multidisciplinary activities, tissue engineering combines the interaction of physicists, chemists, biologists, engineers, biotechnologist, dentists and physicians with common goals: the search for materials that could promote and lead cell activity. A well-oriented combining of scaffolds, promoting factors, cells, together with gene therapy advances may open new avenues to bone healing in the near future. In this review, our target was to write a report bringing overall concepts on tissue bioengineering, with a special attention to decisive biological parameters for the development of biomaterials, as well as to discuss known intracellular signal transduction as a new manner to be explored within this field, aiming to predict in vitro the quality of the host cell/material and thus contributing with the development of regenerative medicine.
Collapse
Affiliation(s)
- Sara Gemini-Piperni
- Laboratório de Bioensaios e Dinâmica Celular, Depto. Química e Bioquímica, Instituto de Biociência, Universidade Estadual Paulista, UNESP, Campus Botucatu, Botucatu, SP, Brazil; Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | | | | | - Katiúcia B S Paiva
- Extracellular Matrix Biology and Cellular Interaction Group, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - José Mauro Granjeiro
- Instituto Nacional de Metrologia, Normalização e Qualidade Industrial (INMETRO), Diretoria de Programas (DIPRO)/Bioengenharia, Xerém, RJ, Brazil
| | - Rodrigo Cardoso de Oliveira
- Department of Biological Sciences, Bauru Dental School, University of São Paulo (USP), Alameda Dr. Octávio Pinheiro Brisolla 9-75, Bauru, São Paulo, SP 17012-901, Brazil
| | - Willian Fernando Zambuzzi
- Laboratório de Bioensaios e Dinâmica Celular, Depto. Química e Bioquímica, Instituto de Biociência, Universidade Estadual Paulista, UNESP, Campus Botucatu, Botucatu, SP, Brazil.
| |
Collapse
|