1
|
Yu C, Yu S, Liu Z, Xu L, Zhang Z, Wan J, Ji P, Zhang P, Fu Y, Le Y, Hou R. Morroniside promotes skin wound re-epithelialization by facilitating epidermal stem cell proliferation through GLP-1R-mediated upregulation of β-catenin expression. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1072-1084. [PMID: 38779766 PMCID: PMC11322873 DOI: 10.3724/abbs.2024070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/06/2024] [Indexed: 05/25/2024] Open
Abstract
Epidermal stem cells (EpSCs) play a vital role in skin wound healing through re-epithelialization. Identifying chemicals that can promote EpSC proliferation is helpful for treating skin wounds. This study investigates the effect of morroniside on cutaneous wound healing in mice and explores the underlying mechanisms. Application of 10‒50 μg/mL of morroniside to the skin wound promotes wound healing in mice. In vitro studies demonstrate that morroniside stimulates the proliferation of mouse and human EpSCs in a time- and dose-dependent manner. Mechanistic studies reveal that morroniside promotes the proliferation of EpSCs by facilitating the cell cycle transition from the G1 to S phase. Morroniside increases the expression of β-catenin via the glucagon-like peptide-1 receptor (GLP-1R)-mediated PKA, PKA/PI3K/AKT and PKA/ERK signaling pathways, resulting in an increase in cyclin D1 and cyclin E1 expression, either directly or by upregulating c-Myc expression. This process ultimately leads to EpSC proliferation. Administration of morroniside to mouse skin wounds increases the phosphorylation of AKT and ERK, the expressions of β-catenin, c-Myc, cyclin D1, and cyclin E1, as well as the proliferation of EpSCs, in periwound skin tissue, and accelerates wound re-epithelialization. These effects of morroniside are mediated by the GLP-1R. Overall, these results indicate that morroniside promotes skin wound healing by stimulating the proliferation of EpSCs via increasing β-catenin expression and subsequently upregulating c-Myc, cyclin D1, and cyclin E1 expressions through GLP-1R signaling pathways. Morroniside has clinical potential for treating skin wounds.
Collapse
Affiliation(s)
- Chenghao Yu
- Suzhou Ruihua Orthopedic HospitalSuzhou Medical College of Soochow UniversitySuzhou215104China
- Department of Hand SurgerySuzhou Ruihua Orthopedic HospitalSuzhou215104China
| | - Siyuan Yu
- Department of Hand SurgerySuzhou Ruihua Orthopedic HospitalSuzhou215104China
- Yangzhou University Medical CollegeYangzhou225009China
| | - Zuohua Liu
- Suzhou Ruihua Orthopedic HospitalSuzhou Medical College of Soochow UniversitySuzhou215104China
- Department of Hand SurgerySuzhou Ruihua Orthopedic HospitalSuzhou215104China
| | - Lei Xu
- Suzhou Ruihua Orthopedic HospitalSuzhou Medical College of Soochow UniversitySuzhou215104China
- Department of Hand SurgerySuzhou Ruihua Orthopedic HospitalSuzhou215104China
| | - Zhiqiang Zhang
- Suzhou Ruihua Orthopedic HospitalSuzhou Medical College of Soochow UniversitySuzhou215104China
- Department of Hand SurgerySuzhou Ruihua Orthopedic HospitalSuzhou215104China
| | - Jiaming Wan
- Department of Hand SurgerySuzhou Ruihua Orthopedic HospitalSuzhou215104China
- Yangzhou University Medical CollegeYangzhou225009China
| | - Pengxiang Ji
- Department of Hand SurgerySuzhou Ruihua Orthopedic HospitalSuzhou215104China
| | - Ping Zhang
- Department of Hand SurgerySuzhou Ruihua Orthopedic HospitalSuzhou215104China
| | - Yi Fu
- Department of Human AnatomyHistology and EmbryologySchool of Biology and Basic Medical SciencesSuzhou Medical College of Soochow UniversitySuzhou215123China
| | - Yingying Le
- Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Ruixing Hou
- Suzhou Ruihua Orthopedic HospitalSuzhou Medical College of Soochow UniversitySuzhou215104China
- Department of Hand SurgerySuzhou Ruihua Orthopedic HospitalSuzhou215104China
- Yangzhou University Medical CollegeYangzhou225009China
| |
Collapse
|
2
|
Hamida OB, Kim MK, Kwack MH. The role of dexamethasone in mediating the contradictory effects of Wnt antagonists SFRP2 and SFRP3 on human hair follicle growth. Sci Rep 2023; 13:16504. [PMID: 37783752 PMCID: PMC10545675 DOI: 10.1038/s41598-023-43688-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/27/2023] [Indexed: 10/04/2023] Open
Abstract
Stress can be one of the leading causes of hair loss. Stress related hormones, glucocorticoids (GCs), secretion by hair follicle have been mentioned in literature and proven to exert an inhibitory effect on hair follicle cells growth by modulating the expression of target genes related to cell proliferation and cycling. The gene modulating effect of the synthetic GC, dexamethasone (DEX), in human dermal papilla (DP) cells has been outlined in this study by mediating a contradictory effect on the expression of secreted frizzled related protein 2 (SFRP2) and SFRP3. The SFRP2 and SFRP3 possess a regulating effect on wnt signaling pathway. Their structural similarities to the cysteine-rich-domain of the frizzled receptors (FZD) allow their binding to the wnt ligands causing the blocking of the wnt ligands-receptors complex. The SFRP family members have been known as inhibitors of the wnt signaling modulating the proliferation and development of various cells. In hair follicle cells, SFRP2 activity has been reported positively on the proliferation of keratinocytes. However, the SFRP3 effect hasn't been well addressed. Under stress, the investigation of the mRNA and protein expressions of SFRP members in human DP cells revealed opposite expressions where SFRP2 decreased while SFRP3 increased by DEX. The proliferation rate of hair keratinocytes outer root sheath was detected via immunofluorescence highlighting the stimulatory effect of SFRP2 and the inhibitory effect of SFRP3. Here, we sought to determine the effect of GC agonist on SFRPs expression and their effect on hair follicle growth.
Collapse
Affiliation(s)
- Ons Ben Hamida
- Department of Immunology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-Ro, Jung-Gu, Daegu, 41944, Republic of Korea
| | - Moon Kyu Kim
- Department of Immunology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-Ro, Jung-Gu, Daegu, 41944, Republic of Korea.
- Hair Transplantation Center, Kyungpook National University Hospital, Daegu, Republic of Korea.
| | - Mi Hee Kwack
- Department of Immunology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-Ro, Jung-Gu, Daegu, 41944, Republic of Korea.
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
3
|
Wu S, Wang W, Li Q, Li J, Dong M, Zhou X, Wang L, Song L. CgWnt-1 regulates haemocyte proliferation during immune response of oyster Crassostrea gigas. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 146:104744. [PMID: 37230373 DOI: 10.1016/j.dci.2023.104744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 05/27/2023]
Abstract
Recent findings regarding the immunomodulatory role of Wnt signaling suggest that it is significant in regulating the differentiation and proliferation of immune cells. In the present study, a Wnt-1 homolog (designated as CgWnt-1) with a conserved WNT1 domain was identified from oyster Crassostrea gigas. The transcripts of CgWnt-1 were barely expressed in egg to gastrula stage during early embryogenesis, and up-regulated significantly in the trochophore to juvenile stage. The mRNA transcripts of CgWnt-1 were detected in different tissues of adult oyster, with an extremely high expression level in the mantle, which was 77.38-fold (p < 0.05) of that in labial palp. After Vibrio splendidus stimulation, the mRNA expression levels of CgWnt-1 and Cgβ-catenin in haemocytes up-regulated significantly at 3, 12, 24, and 48 h (p < 0.05). After injection of recombinant protein (rCgWnt-1) into oyster in vivo, the expressions of Cgβ-catenin, cell proliferation related genes CgRunx-1 and CgCDK-2 in haemocytes significantly up-regulated, which were 4.86-fold (p < 0.05), 9.33-fold (p < 0.05), 6.09-fold (p < 0.05) of those in rTrx group, respectively. The percentage of EDU+ cells in haemocytes also significantly increased (2.88-fold of that in control group, p < 0.05) at 12 h after rCgWnt-1 treatment. When the Wnt signal inhibitor C59 was injected simultaneously with rCgWnt-1, the expressions of Cgβ-catenin, CgRunx-1, and CgCDK-2 were significantly reduced, which were 0.32-fold (p < 0.05), 0.16-fold (p < 0.05), and 0.25-fold (p < 0.05) of that in rCgWnt-1 group, respectively, and the percentage of EDU+ cells in haemocytes was also significantly inhibited (0.15-fold compared with that in rCgWnt-1 group, p < 0.05). These results suggested that the conserved CgWnt-1 could modulate haemocytes proliferation via regulating cell cycle related genes and involved in the immune response of oysters.
Collapse
Affiliation(s)
- Shasha Wu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Weilin Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China.
| | - Qing Li
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Jialuo Li
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Miren Dong
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Xiaoxu Zhou
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| |
Collapse
|
4
|
Sukarawan W, Rattanawarawipa P, Yaemkleebbua K, Nowwarote N, Pavasant P, Limjeerajarus CN, Osathanon T. Wnt3a promotes odonto/osteogenic differentiation in vitro and tertiary dentin formation in a rat model. Int Endod J 2023; 56:514-529. [PMID: 36633501 DOI: 10.1111/iej.13888] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/13/2023]
Abstract
AIM To investigate the effect of Wnt3a on odonto/osteogenic differentiation of stem cells isolated from human exfoliated deciduous teeth (SHEDs) and reparative dentine formation in a rat model. METHODOLOGY Stem cells isolated from human exfoliated deciduous teeth were cultured in media with Wnt3a (50-200 ng/ml). Wnt activation was confirmed by β-catenin immunocytochemistry. Colony-forming unit assay (normalized percentage area), osteogenic gene expression analysis by real-time polymerase chain reaction and mineralization assays measured by the absorption at 540 nm were performed. Tertiary dentine formation in vivo was evaluated using 8-week-old, male Wistar rats. Cavities with pinpoint pulp exposure by a sharp instrument were prepared at the mesial surface of the first molars. Teeth were divided into (n = 6): (1) distilled water (negative control), (2) phosphate-buffered saline (PBS), (3) lithium chloride in DI (20 μM), and (4) Wnt3a in PBS (200 ng/ml). Collagen sponge was used as a scaffold. The cavity was sealed with glass ionomer restoration. Four weeks later, animals were euthanized by sodium pentobarbital (120 mg/kg body weight). Hard tissue formation was evaluated using micro-computerized tomography. Sixty consecutive slides from the initial plane were analysed and calculated as bone/dentine volume per total tissue volume. Paraffin sections (2 μm) were stained with haematoxylin and eosin and Masson's trichrome for morphological evaluation. Data are presented as the mean ± standard error. Mann-Whitney U test was used for two-group comparison. Kruskal Wallis followed by pairwise comparison was employed for three or more group comparisons. Statistical analysis was performed using GraphPad Prism 7. Differences were considered significant at p < .05. RESULTS Wnt3a decreased SHEDs colony formation and increased OSX, BMP2, and DMP1 expression, corresponding to an increase in mineralization. Additionally, a significant increase in dentine/bone volume per total tissue volume was observed in Wnt3a treated defects. Dentine bridge formation at the exposure sites treated with Wnt3a demonstrated, while fibrous tissues were observed in the control. CONCLUSIONS Wnt3a suppressed proliferation, increased osteogenic differentiation of SHEDs and promotes tertiary dentine formation. Wnt3a could be utilized as biological molecule for vital pulp therapy.
Collapse
Affiliation(s)
- Waleerat Sukarawan
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Panarat Rattanawarawipa
- Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Kamonwan Yaemkleebbua
- Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Nunthawan Nowwarote
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Centre de Recherche des Cordeliers, Université de Paris, Sorbonne Université, INSERM UMRS 1138, Molecular Oral Pathophysiology, Paris, France.,Dental Faculty Garancière, Oral Biology Department, Université de Paris, Paris, France
| | - Prasit Pavasant
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Chalida Nakalekha Limjeerajarus
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Thanaphum Osathanon
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
5
|
Cao J, Zhong L, Feng Y, Qian K, Xiao Y, Wang G, Tu W, Yue L, Zhang Q, Yang H, Jiao Y, Zhu W, Cao J. Activated Beta-Catenin Signaling Ameliorates Radiation-Induced Skin Injury by Suppressing Marvel D3 Expression. Radiat Res 2021; 195:173-190. [PMID: 33045079 DOI: 10.1667/rade-20-00050.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 09/04/2020] [Indexed: 11/03/2022]
Abstract
Radiation-induced skin injury remains a serious concern for cancer radiotherapy, radiation accidents and occupational exposure, and the damage mainly occurs due to apoptosis and reactive oxygen species (ROS) generation. There is currently no effective treatment for this disorder. The β-catenin signaling pathway is involved in the repair and regeneration of injured tissues. However, the role of the β-catenin signaling pathway in radiation-induced skin injury has not been reported. In this study, we demonstrated that the β-catenin signaling pathway was activated in response to radiation and that its activation by Wnt3a, a ligand-protein involved in the β-catenin signaling pathway, inhibited apoptosis and the production of ROS in irradiated human keratinocyte HaCaT cells and skin fibroblast WS1 cells. Additionally, Wnt3a promoted cell migration after irradiation. In a mouse model of full-thickness skin wounds combined with total-body irradiation, Wnt3a was shown to facilitate skin wound healing. The results from RNA-Seq revealed that 24 genes were upregulated and 154 were downregulated in Wnt3a-treated irradiated skin cells, and these dysregulated genes were mainly enriched in the tight junction pathway. Among them, Marvel D3 showed the most obvious difference. We further found that the activated β-catenin signaling pathway stimulated the phosphorylation of JNK by silencing Marvel D3. Treatment of irradiated cells with SP600125, a JNK inhibitor, augmented ROS production and impeded cell migration. Furthermore, treatment with Wnt3a or transfection with Marvel D3-specific siRNAs could reverse the above effects. Taken together, these findings illustrate that activated β-catenin signaling stimulates the activation of JNK by negatively regulating Marvel D3 to ameliorate radiation-induced skin injury.
Collapse
Affiliation(s)
- Jinming Cao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123 China.,State Key Laboratory of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123 China
| | - Li Zhong
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123 China.,State Key Laboratory of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123 China
| | - Yang Feng
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123 China.,State Key Laboratory of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123 China
| | - Kun Qian
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123 China.,State Key Laboratory of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123 China
| | - Yuji Xiao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123 China.,State Key Laboratory of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123 China
| | - Gaoren Wang
- Nantong Tumor Hospital, Nantong University, Nantong 226000 China
| | - Wenling Tu
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051 China
| | - Ling Yue
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123 China.,State Key Laboratory of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123 China
| | - Qi Zhang
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123 China.,State Key Laboratory of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123 China
| | - Hongying Yang
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123 China.,State Key Laboratory of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123 China
| | - Yang Jiao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123 China.,State Key Laboratory of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123 China
| | - Wei Zhu
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123 China.,State Key Laboratory of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123 China
| | - Jianping Cao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123 China.,State Key Laboratory of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123 China
| |
Collapse
|
6
|
Wang Z, Zhang G, Le Y, Ju J, Zhang P, Wan D, Zhao Q, Jin G, Su H, Liu J, Feng J, Fu Y, Hou R. Quercetin promotes human epidermal stem cell proliferation through the estrogen receptor/β-catenin/c-Myc/cyclin A2 signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1102-1110. [PMID: 32840291 DOI: 10.1093/abbs/gmaa091] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Indexed: 01/08/2023] Open
Abstract
Skin epidermal stem cells (EpSCs) play an important role in wound healing. Quercetin is a phytoestrogen reported to accelerate skin wound healing, but its effect on EpSCs is unknown. In this study, we investigated the effect of quercetin on human EpSC proliferation and explored the underlying mechanisms. We found that quercetin at 0.1~1 μM significantly promoted EpSC proliferation and increased the number of cells in S phase. The pro-proliferative effect of quercetin on EpSCs was confirmed in cultured human skin tissue. Mechanistic studies showed that quercetin significantly upregulated the expressions of β-catenin, c-Myc, and cyclins A2 and E1. Inhibitor for β-catenin or c-Myc significantly inhibited quercetin-induced EpSC proliferation. The β-catenin inhibitor XAV-939 suppressed quercetin-induced expressions of β-catenin, c-Myc, and cyclins A2 and E1. The c-Myc inhibitor 10058-F4 inhibited the upregulation of c-Myc and cyclin A2 by quercetin. Pretreatment of EpSCs with estrogen receptor (ER) antagonist ICI182780, but not the G protein-coupled ER1 antagonist G15, reversed quercetin-induced cell proliferation and upregulation of β-catenin, c-Myc, and cyclin A2. Collectively, these results indicate that quercetin promotes EpSC proliferation through ER-mediated activation of β-catenin/c-Myc/cyclinA2 signaling pathway and ER-independent upregulation of cyclin E1 and that quercetin may accelerate skin wound healing through promoting EpSC proliferation. As EpSCs are used not only in clinic to treat skin wounds but also as seed cells in skin tissue engineering, quercetin is a useful reagent to expand EpSCs for basic research, skin wound treatment, and skin tissue engineering.
Collapse
Affiliation(s)
- Zhaodong Wang
- Institute of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou 215104, China
| | - Guangliang Zhang
- Department of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou 215104, China
| | - Yingying Le
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jihui Ju
- Department of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou 215104, China
| | - Ping Zhang
- Institute of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou 215104, China
| | - Dapeng Wan
- Institute of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou 215104, China
| | - Qiang Zhao
- Department of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou 215104, China
| | - Guangzhe Jin
- Department of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou 215104, China
| | - Hao Su
- Institute of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou 215104, China
| | - Jinwei Liu
- Institute of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou 215104, China
| | - Jiaxuan Feng
- Institute of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou 215104, China
| | - Yi Fu
- Department of Human Anatomy, Histology and Embryology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Ruixing Hou
- Institute of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou 215104, China
- Department of Hand Surgery, Ruihua Affiliated Hospital of Soochow University, Suzhou 215104, China
| |
Collapse
|
7
|
Wang H, Humbatova A, Liu Y, Qin W, Lee M, Cesarato N, Kortüm F, Kumar S, Romano MT, Dai S, Mo R, Sivalingam S, Motameny S, Wu Y, Wang X, Niu X, Geng S, Bornholdt D, Kroisel PM, Tadini G, Walter SD, Hauck F, Girisha KM, Calza AM, Bottani A, Altmüller J, Buness A, Yang S, Sun X, Ma L, Kutsche K, Grzeschik KH, Betz RC, Lin Z. Mutations in SREBF1, Encoding Sterol Regulatory Element Binding Transcription Factor 1, Cause Autosomal-Dominant IFAP Syndrome. Am J Hum Genet 2020; 107:34-45. [PMID: 32497488 DOI: 10.1016/j.ajhg.2020.05.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022] Open
Abstract
IFAP syndrome is a rare genetic disorder characterized by ichthyosis follicularis, atrichia, and photophobia. Previous research found that mutations in MBTPS2, encoding site-2-protease (S2P), underlie X-linked IFAP syndrome. The present report describes the identification via whole-exome sequencing of three heterozygous mutations in SREBF1 in 11 unrelated, ethnically diverse individuals with autosomal-dominant IFAP syndrome. SREBF1 encodes sterol regulatory element-binding protein 1 (SREBP1), which promotes the transcription of lipogenes involved in the biosynthesis of fatty acids and cholesterols. This process requires cleavage of SREBP1 by site-1-protease (S1P) and S2P and subsequent translocation into the nucleus where it binds to sterol regulatory elements (SRE). The three detected SREBF1 mutations caused substitution or deletion of residues 527, 528, and 530, which are crucial for S1P cleavage. In vitro investigation of SREBP1 variants demonstrated impaired S1P cleavage, which prohibited nuclear translocation of the transcriptionally active form of SREBP1. As a result, SREBP1 variants exhibited significantly lower transcriptional activity compared to the wild-type, as demonstrated via luciferase reporter assay. RNA sequencing of the scalp skin from IFAP-affected individuals revealed a dramatic reduction in transcript levels of low-density lipoprotein receptor (LDLR) and of keratin genes known to be expressed in the outer root sheath of hair follicles. An increased rate of in situ keratinocyte apoptosis, which might contribute to skin hyperkeratosis and hypotrichosis, was also detected in scalp samples from affected individuals. Together with previous research, the present findings suggest that SREBP signaling plays an essential role in epidermal differentiation, skin barrier formation, hair growth, and eye function.
Collapse
Affiliation(s)
- Huijun Wang
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Aytaj Humbatova
- Institute of Human Genetics, University of Bonn, Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany
| | - Yuanxiang Liu
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Wen Qin
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Mingyang Lee
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Nicole Cesarato
- Institute of Human Genetics, University of Bonn, Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany
| | - Fanny Kortüm
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Sheetal Kumar
- Institute of Human Genetics, University of Bonn, Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany
| | - Maria Teresa Romano
- Institute of Human Genetics, University of Bonn, Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany
| | - Shangzhi Dai
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Ran Mo
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Sugirthan Sivalingam
- Institute for Medical Biometry, Informatics and Epidemiology, University of Bonn, Medical Faculty, 53127 Bonn, Germany; Institute for Genomic Statistics and Bioinformatics, University of Bonn, Medical Faculty, 53127 Bonn, Germany
| | - Susanne Motameny
- Cologne Center for Genomics, University of Cologne, 50931 Cologne, Germany
| | - Yuan Wu
- Department of Ophthalmology, Peking University First Hospital, Beijing 100034, China
| | - Xiaopeng Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xinwu Niu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Songmei Geng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Dorothea Bornholdt
- Centre for Human Genetics, University of Marburg, 35033 Marburg, Germany
| | - Peter M Kroisel
- Institute of Human Genetics, Medical University of Graz, 8010 Graz, Austria
| | - Gianluca Tadini
- Pediatric Dermatology Unit, Department of Pathophysiology and Transplantation, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Scott D Walter
- Retina Consultants, P.C., 43 Woodland Street, Suite 100, Hartford, CT 06105, USA
| | - Fabian Hauck
- Department of Pediatrics, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katta M Girisha
- Department of Medical Genetics, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Anne-Marie Calza
- Department of Dermatology and Venereology, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Armand Bottani
- Service of Genetic Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Janine Altmüller
- Cologne Center for Genomics, University of Cologne, 50931 Cologne, Germany
| | - Andreas Buness
- Institute for Medical Biometry, Informatics and Epidemiology, University of Bonn, Medical Faculty, 53127 Bonn, Germany; Institute for Genomic Statistics and Bioinformatics, University of Bonn, Medical Faculty, 53127 Bonn, Germany
| | - Shuxia Yang
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China
| | - Xiujuan Sun
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Lin Ma
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Kerstin Kutsche
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | - Regina C Betz
- Institute of Human Genetics, University of Bonn, Medical Faculty & University Hospital Bonn, 53127 Bonn, Germany.
| | - Zhimiao Lin
- Department of Dermatology, Peking University First Hospital, Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, National Clinical Research Center for Skin and Immune Diseases, Beijing 100034, China.
| |
Collapse
|
8
|
Luteolin-7-glucoside Promotes Human Epidermal Stem Cell Proliferation by Upregulating β-Catenin, c-Myc, and Cyclin Expression. Stem Cells Int 2019; 2019:1575480. [PMID: 31281367 PMCID: PMC6589269 DOI: 10.1155/2019/1575480] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 03/31/2019] [Accepted: 04/21/2019] [Indexed: 02/02/2023] Open
Abstract
Skin epidermal stem cells (EpSCs) play critical roles in skin homeostasis and the repair of skin injury. Luteolin-7-glucoside (L7G) has been reported to accelerate skin wound healing through its anti-inflammatory and antioxidative activity. But its effect on EpSCs is not clear. In the present study, we examined the effect of L7G on the proliferation of human EpSCs and explored the mechanisms involved. MTT assay showed that L7G promoted EpSC proliferation in a dose- and time-dependent manner. BrdU incorporation assay and Ki67 immunofluorescence staining confirmed the proproliferative effect of L7G on EpSCs. Cell cycle analysis showed that treatment of EpSCs with L7G decreased the cell number in the G1 phase and increased the cell number in the S phase. In addition, L7G significantly enhanced EpSC migration. Mechanistic studies showed that L7G significantly induced the expression of β-catenin and c-Myc, as well as cyclins D1, A2, and E1 which are critical for G1/S phase transition. L7G stimulated EpSC proliferation through β-catenin and c-Myc. We further examined the effect of L7G on EpSC proliferation in skin tissues by treatment of human skin explants with L7G and examined the number of EpSCs by immunohistochemical stain of EpSC markers α 6 integrin and β 1 integrin. We found that treatment of human skin tissue explants with L7G significantly increased the thickness of the epidermis and increased the numbers of α 6 integrin-positive and β 1 integrin-positive cells at the basal layer of the epidermis. Taken together, these results indicate that L7G promotes EpSC proliferation through upregulating β-catenin, c-Myc, and cyclin expression. L7G can be used to expand EpSCs for generating epidermal autografts and engineered skin equivalents.
Collapse
|
9
|
Deng C, Sun Y, Liu H, Wang W, Wang J, Zhang F. Selective adipogenic differentiation of human periodontal ligament stem cells stimulated with high doses of glucose. PLoS One 2018; 13:e0199603. [PMID: 29979705 PMCID: PMC6034828 DOI: 10.1371/journal.pone.0199603] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/11/2018] [Indexed: 02/07/2023] Open
Abstract
Periodontal tissue damage, accompanied by the degradation and destruction of periodontal tissue collagen, is one of the most clinically common complications and difficulty self-repair in patients with diabetes. Human periodontal ligament stem cells (PDLSC) are the undifferentiated mesenchymal cells that persist in the periodontal ligament after development of periodontal tissue and the ability of PDLSC osteogenic differentiation is responsible for repairing periodontal tissue defects. However, the reasons of high glucose environment in diabetic patients inhibiting PDLSC to repair periodontal tissues are unclear. To address these issues, we propose exposing PDLSC to high-sugar mimics the diabetic environment and investigating the activity of osteogenic differentiation and adipogenic differentiation of PDLSC. At the cellular level, high glucose can promote the adipogenic differentiation and inhibit osteogenic differentiation to decrease the self-repair ability of PDLSC in periodontal tissues. Mechanistically at the molecular level, these effects are elicited via regulating the mRNA and protein expression of C/EBPβ, PPAR-γ.
Collapse
Affiliation(s)
- Chao Deng
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University,Nanjing, China
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Yi Sun
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Hai Liu
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Wei Wang
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Jingmen Wang
- School of Stomatology, Wannan Medical College, Wuhu, China
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University,Nanjing, China
- * E-mail:
| |
Collapse
|
10
|
Ye X, Cheng S, Dong Y, Ren J, Su L, Liu J, Zhou J, Liu Q, Zhu N. Exendin-4 promotes proliferation of adipose-derived stem cells through PI3K/Akt-Wnt signaling pathways. Neurosci Lett 2018; 685:196-202. [PMID: 29920298 DOI: 10.1016/j.neulet.2018.06.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 06/15/2018] [Accepted: 06/15/2018] [Indexed: 01/27/2023]
Abstract
Adipose-derived stem cell (ADSC) transplantation has emerged as a potential tool for the treatment of cardiovascular disease and skin wounds. However, with a limited renewal capacity and the need for mass cells during the engraftment, strategies are needed to enhance ADSC proliferative capacity. In this study, we explored the effects of Exendin-4, a glucagon-like peptide-1 analog, on the growth of ADSCs, focusing in particular on phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt) and Wnt signaling pathways. Firstly, ADSCs were isolated and cultured in vitro. Then, flow cytometry demonstrated that ADSCs were positive for CD44, CD90 and CD29 but negative for CD31, CD34, and CD45. Exendin-4 (0-200 nM) treatment increased ADSC proliferation. In order to examine specific signaling pathways, a western blotting assay was performed. Our results demonstrate that after treated with 50 nM Exendin-4 for 48 h, the phosphorylation of PI3K, Akt, and GSK3β were increased and phosphorylation of β-catenin was decreased. From these results, we concluded that PI3K-Akt and Wnt-β-catenin signaling pathways mediate Exendin-4 induced ADSC proliferation, the function of which might contribute to the regulation of ADSC proliferation. Our findings provided new insights into the function of the mechanisms underlying Exendin-4 of ADSCs.
Collapse
Affiliation(s)
- Xiaolu Ye
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Shimeng Cheng
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Yabing Dong
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Jie Ren
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Lina Su
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Jianlan Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Jing Zhou
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Qingmei Liu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Ningwen Zhu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China.
| |
Collapse
|
11
|
Weiner L, Fu W, Chirico WJ, Brissette JL. Skin as a living coloring book: how epithelial cells create patterns of pigmentation. Pigment Cell Melanoma Res 2014; 27:1014-31. [PMID: 25104547 DOI: 10.1111/pcmr.12301] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 08/05/2014] [Indexed: 12/23/2022]
Abstract
The pigmentation of mammalian skin and hair develops through the interaction of two basic cell types - pigment donors and recipients. The pigment donors are melanocytes, which produce and distribute melanin through specialized structures. The pigment recipients are epithelial cells, which acquire melanin and put it to use, collectively yielding the pigmentation visible to the eye. This review will focus on the pigment recipients, the historically less understood cell type. These end-users of pigment are now known to exert a specialized control over the patterning of pigmentation, as they identify themselves as melanocyte targets, recruit pigment donors, and stimulate the transfer of melanin. As such, this review will discuss the evidence that the skin is like a coloring book: the pigment recipients create a 'picture,' a blueprint for pigmentation, which is colorless initially but outlines where pigment should be placed. Melanocytes then melanize the recipients and 'color in' the picture.
Collapse
Affiliation(s)
- Lorin Weiner
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | | | | | | |
Collapse
|
12
|
Xu X, Wang HY, Zhang Y, Liu Y, Li YQ, Tao K, Wu CT, Jin JD, Liu XY. Adipose-derived stem cells cooperate with fractional carbon dioxide laser in antagonizing photoaging: a potential role of Wnt and β-catenin signaling. Cell Biosci 2014; 4:24. [PMID: 24917925 PMCID: PMC4050444 DOI: 10.1186/2045-3701-4-24] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 04/10/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND It is well established that adipose-derived stem cells (ADSCs) produce and secrete cytokines/growth factors that antagonize UV-induced photoaging of skin. However, the exact molecular basis underlying the anti-photoaging effects exerted by ADSCs is not well understood, and whether ADSCs cooperate with fractional carbon dioxide (CO2) laser to facilitate photoaging skin healing process has not been explored. Here, we investigated the impacts of ADSCs on photoaging in a photoaging animal model, its associated mechanisms, and its functional cooperation with fractional CO2 laser in treatment of photoaging skin. RESULTS We showed that ADSCs improved dermal thickness and activated the proliferation of dermal fibroblast. We further demonstrated that the combined treatment of ADSCs and fractional CO2 laser, the latter which is often used to resurface skin and treat wrinkles, had more beneficial effects on the photoaging skin compared with each individual treatment. In our prepared HDF photoaging model, flow cytometry showed that, after adipose derived stem cells conditioned medium (ADSC-CM) co-cultured HDF photoaging model, the cell proliferation rate is higher than UVB irradiation induced HDF modeling (p < 0.05). Additionally, the expressions of β-catenin and Wnt3a, which were up-regulated after the transplantation of ADSCs alone or in combination with fractional CO2 laser treatment. And the expression of wnt3a and β-catenin has the positive correlation with photoaging related protein TGF-β2 and COLI. We also verified these protein expressions in tissue level. In addition, after injected SFRP2 into ADSC-CM co-cultured HDF photoaging model, wnt3a inhibitor, compared with un-intervened group, wnt3a, β-catenin protein level significantly decreased. CONCLUSION Both ADSCs and fractional CO2 laser improved photoaging skin at least partially via targeting dermal fibroblast activity which was increased in photoaging skin. The combinatorial use of ADSCs and fractional CO2 laser synergistically improved the healing process of photoaging skin. Thus, we provide a strong rationale for a combined use of ADSCs and fractional CO2 laser in treatment of photoaging skin in clinic in the future. Moreover, we provided evidence that the Wnt/β-catenin signaling pathway may contribute to the activation of dermal fibroblast by the transplantation of ADSCs in both vitro and vivo experiment.
Collapse
Affiliation(s)
- Xiao Xu
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, P.R. China
| | - Hong-Yi Wang
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, P.R. China
| | - Yu Zhang
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, P.R. China
| | - Yang Liu
- Department of Experimental Hematology, Institute of Radiation Medicine, Beijing, P.R.China
| | - Yan-Qi Li
- Department of Experimental Hematology, Institute of Radiation Medicine, Beijing, P.R.China
| | - Kai Tao
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, P.R. China
| | - Chu-Tse Wu
- Department of Experimental Hematology, Institute of Radiation Medicine, Beijing, P.R.China
| | - Ji-de Jin
- Department of Experimental Hematology, Institute of Radiation Medicine, Beijing, P.R.China
| | - Xiao-Yan Liu
- Reconstructive and Plastic Surgery, The General Hospital of Shenyang Military Region, Shenyang, P.R. China
| |
Collapse
|
13
|
Yang Q, Du X, Fang Z, Xiong W, Li G, Liao H, Xiao J, Wang G, Li F. Effect of calcitonin gene-related peptide on the neurogenesis of rat adipose-derived stem cells in vitro. PLoS One 2014; 9:e86334. [PMID: 24466033 PMCID: PMC3897681 DOI: 10.1371/journal.pone.0086334] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 12/09/2013] [Indexed: 01/15/2023] Open
Abstract
Calcitonin gene-related peptide (CGRP) promotes neuron recruitment and neurogenic activity. However, no evidence suggests that CGRP affects the ability of stem cells to differentiate toward neurogenesis. In this study, we genetically modified rat adipose-derived stem cells (ADSCs) with the CGRP gene (CGRP-ADSCs) and subsequently cultured in complete neural-induced medium. The formation of neurospheres, cellular morphology, and proliferative capacity of ADSCs were observed. In addition, the expression of the anti-apoptotic protein Bcl-2 and special markers of neural cells, such as Nestin, MAP2, RIP and GFAP, were evaluated using Western blot and immunocytochemistry analysis. The CGRP-ADSCs displayed a greater proliferation than un-transduced (ADSCs) and Vector-transduced (Vector-ADSCs) ADSCs (p<0.05), and lower rates of apoptosis, associated with the incremental expression of Bcl-2, were also observed for CGRP-ADSCs. Moreover, upon neural induction, CGRP-ADSCs formed markedly more and larger neurospheres and showed round cell bodies with more branching extensions contacted with neighboring cells widely. Furthermore, the expression levels of Nestin, MAP2, and RIP in CGRP-ADSCs were markedly increased, resulting in higher levels than the other groups (p<0.05); however, GFAP was distinctly undetectable until day 7, when slight GFAP expression was detected among all groups. Wnt signals, primarily Wnt 3a, Wnt 5a and β-catenin, regulate the neural differentiation of ADSCs, and CGRP gene expression apparently depends on canonical Wnt signals to promote the neurogenesis of ADSCs. Consequently, ADSCs genetically modified with CGRP exhibit stronger potential for differentiation and neurogenesis in vitro, potentially reflecting the usefulness of ADSCs as seed cells in therapeutic strategies for spinal cord injury.
Collapse
Affiliation(s)
- Qin Yang
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P R China
| | - Xingli Du
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P R China
| | - Zhong Fang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P R China
- * E-mail:
| | - Wei Xiong
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P R China
| | - Guanghui Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P R China
| | - Hui Liao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P R China
| | - Jun Xiao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P R China
| | - Guoping Wang
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P R China
| | - Feng Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P R China
| |
Collapse
|
14
|
Dhamdhere GR, Fang MY, Jiang J, Lee K, Cheng D, Olveda RC, Liu B, Mulligan KA, Carlson JC, Ransom RC, Weis WI, Helms JA. Drugging a stem cell compartment using Wnt3a protein as a therapeutic. PLoS One 2014; 9:e83650. [PMID: 24400074 PMCID: PMC3882211 DOI: 10.1371/journal.pone.0083650] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 11/06/2013] [Indexed: 01/08/2023] Open
Abstract
The therapeutic potential of Wnt proteins has long been recognized but challenges associated with in vivo stability and delivery have hindered their development as drug candidates. By exploiting the hydrophobic nature of the protein we provide evidence that exogenous Wnt3a can be delivered in vivo if it is associated with a lipid vesicle. Recombinant Wnt3a associates with the external surface of the lipid membrane; this association stabilizes the protein and leads to prolonged activation of the Wnt pathway in primary cells. We demonstrate the consequences of Wnt pathway activation in vivo using a bone marrow engraftment assay. These data provide validation for the development of WNT3A as a therapeutic protein.
Collapse
Affiliation(s)
- Girija R. Dhamdhere
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, California, United States of America
| | - Mark Y. Fang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, California, United States of America
| | - Jie Jiang
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, California, United States of America
| | - Katherine Lee
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, California, United States of America
| | - Du Cheng
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, California, United States of America
| | - Rebecca C. Olveda
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, California, United States of America
| | - Bo Liu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, California, United States of America
| | - Kimberley A. Mulligan
- Department of Developmental Biology, Howard Hughes Medical Institute (HHMI), Institute for Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford University, Stanford, California, United States of America
| | - Jeffery C. Carlson
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, California, United States of America
| | - Ryan C. Ransom
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, California, United States of America
| | - William I. Weis
- Departments of Structural Biology and Molecular and Cellular Physiology, Stanford School of Medicine, Stanford, California, United States of America
| | - Jill A. Helms
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
15
|
Epidermal development in mammals: key regulators, signals from beneath, and stem cells. Int J Mol Sci 2013; 14:10869-95. [PMID: 23708093 PMCID: PMC3709707 DOI: 10.3390/ijms140610869] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 04/22/2013] [Accepted: 04/23/2013] [Indexed: 12/23/2022] Open
Abstract
Epidermis is one of the best-studied tissues in mammals that contain types of stem cells. Outstanding works in recent years have shed great light on behaviors of different epidermal stem cell populations in the homeostasis and regeneration of the epidermis as well as hair follicles. Also, the molecular mechanisms governing these stem cells are being elucidated, from genetic to epigenetic levels. Compared with the explicit knowledge about adult skin, embryonic development of the epidermis, especially the early period, still needs exploration. Furthermore, stem cells in the embryonic epidermis are largely unstudied or ambiguously depicted. In this review, we will summarize and discuss the process of embryonic epidermal development, with focuses on some key molecular regulators and the role of the sub-epidermal mesenchyme. We will also try to trace adult epidermal stem cell populations back to embryonic development. In addition, we will comment on in vitro derivation of epidermal lineages from ES cells and iPS cells.
Collapse
|
16
|
Guo H, Yang K, Deng F, Xing Y, Li Y, Lian X, Yang T. Wnt3a inhibits proliferation but promotes melanogenesis of melan-a cells. Int J Mol Med 2012; 30:636-42. [PMID: 22710324 DOI: 10.3892/ijmm.2012.1028] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 05/14/2012] [Indexed: 11/05/2022] Open
Abstract
Melanocytes are pigment-producing cells responsible for coloration of skin and hair. Although the importance of Wnt3a in melanocyte development has been well recognized, the role of Wnt3a in mature melanocytes has not been elucidated. This study was conducted to further explore the effects of Wnt3a on melanocyte proliferation and melanogenesis, and to elucidate the possible mechanisms involved. We infected melan-a cells with AdWnt3a to serve as the production source of the Wnt3a protein. MTT assay, 5-bromodeoxyuridine incorporation assay and flow cytometric analysis showed that Wnt3a inhibited the proliferation of melan-a cells and this was associated with decrease of cells in the S phase and increase of cells in the G(1) phase. Melanin content and tyrosinase activity assay revealed that Wnt3a significantly promoted melanogenesis of melan-a cells. Furthermore, western blot analysis showed that Wnt3a upregulated the expression of microphthalmia-associated transcription factor and its downstream target genes, tyrosinase and tyrosinase-related protein 1 in melan-a cells. Collectively, our results suggest that Wnt3a plays an important role in melanocyte homeostasis.
Collapse
Affiliation(s)
- Haiying Guo
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, P.R. China
| | | | | | | | | | | | | |
Collapse
|
17
|
Guo H, Yang K, Deng F, Ye J, Xing Y, Li Y, Lian X, Yang T. Wnt3a promotes melanin synthesis of mouse hair follicle melanocytes. Biochem Biophys Res Commun 2012; 420:799-804. [DOI: 10.1016/j.bbrc.2012.03.077] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2012] [Accepted: 03/14/2012] [Indexed: 10/28/2022]
|
18
|
NASA-approved rotary bioreactor enhances proliferation of human epidermal stem cells and supports formation of 3D epidermis-like structure. PLoS One 2011. [PMID: 22096490 DOI: 10.1371/journal.pone.0026603.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The skin is susceptible to different injuries and diseases. One major obstacle in skin tissue engineering is how to develop functional three-dimensional (3D) substitute for damaged skin. Previous studies have proved a 3D dynamic simulated microgravity (SMG) culture system as a "stimulatory" environment for the proliferation and differentiation of stem cells. Here, we employed the NASA-approved rotary bioreactor to investigate the proliferation and differentiation of human epidermal stem cells (hEpSCs). hEpSCs were isolated from children foreskins and enriched by collecting epidermal stem cell colonies. Cytodex-3 micro-carriers and hEpSCs were co-cultured in the rotary bioreactor and 6-well dish for 15 days. The result showed that hEpSCs cultured in rotary bioreactor exhibited enhanced proliferation and viability surpassing those cultured in static conditions. Additionally, immunostaining analysis confirmed higher percentage of ki67 positive cells in rotary bioreactor compared with the static culture. In contrast, comparing with static culture, cells in the rotary bioreactor displayed a low expression of involucrin at day 10. Histological analysis revealed that cells cultured in rotary bioreactor aggregated on the micro-carriers and formed multilayer 3D epidermis structures. In conclusion, our research suggests that NASA-approved rotary bioreactor can support the proliferation of hEpSCs and provide a strategy to form multilayer epidermis structure.
Collapse
|
19
|
Lei XH, Ning LN, Cao YJ, Liu S, Zhang SB, Qiu ZF, Hu HM, Zhang HS, Liu S, Duan EK. NASA-approved rotary bioreactor enhances proliferation of human epidermal stem cells and supports formation of 3D epidermis-like structure. PLoS One 2011; 6:e26603. [PMID: 22096490 PMCID: PMC3212516 DOI: 10.1371/journal.pone.0026603] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 09/29/2011] [Indexed: 12/12/2022] Open
Abstract
The skin is susceptible to different injuries and diseases. One major obstacle in skin tissue engineering is how to develop functional three-dimensional (3D) substitute for damaged skin. Previous studies have proved a 3D dynamic simulated microgravity (SMG) culture system as a “stimulatory” environment for the proliferation and differentiation of stem cells. Here, we employed the NASA-approved rotary bioreactor to investigate the proliferation and differentiation of human epidermal stem cells (hEpSCs). hEpSCs were isolated from children foreskins and enriched by collecting epidermal stem cell colonies. Cytodex-3 micro-carriers and hEpSCs were co-cultured in the rotary bioreactor and 6-well dish for 15 days. The result showed that hEpSCs cultured in rotary bioreactor exhibited enhanced proliferation and viability surpassing those cultured in static conditions. Additionally, immunostaining analysis confirmed higher percentage of ki67 positive cells in rotary bioreactor compared with the static culture. In contrast, comparing with static culture, cells in the rotary bioreactor displayed a low expression of involucrin at day 10. Histological analysis revealed that cells cultured in rotary bioreactor aggregated on the micro-carriers and formed multilayer 3D epidermis structures. In conclusion, our research suggests that NASA-approved rotary bioreactor can support the proliferation of hEpSCs and provide a strategy to form multilayer epidermis structure.
Collapse
Affiliation(s)
- Xiao-hua Lei
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Li-na Ning
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yu-jing Cao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Shuang Liu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Graduate School, Chinese Academy of Sciences, Beijing, China
| | - Shou-bing Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Graduate School, Chinese Academy of Sciences, Beijing, China
| | - Zhi-fang Qiu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hui-min Hu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Graduate School, Chinese Academy of Sciences, Beijing, China
| | - Hui-shan Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Graduate School, Chinese Academy of Sciences, Beijing, China
| | - Shu Liu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Graduate School, Chinese Academy of Sciences, Beijing, China
| | - En-kui Duan
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- * E-mail:
| |
Collapse
|
20
|
Zabierowski SE, Fukunaga-Kalabis M, Li L, Herlyn M. Dermis-derived stem cells: a source of epidermal melanocytes and melanoma? Pigment Cell Melanoma Res 2011; 24:422-9. [PMID: 21410654 DOI: 10.1111/j.1755-148x.2011.00847.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Human multipotent dermal stem cells (DSCs) have been isolated and propagated from the dermal region of neonatal foreskin. DSCs can self-renew, express the neural crest stem cell markers NGFRp75 and nestin, and are capable of differentiating into a wide variety of cell types including mesenchymal and neuronal lineages and melanocytes, indicative of their neural crest origin. When placed in the context of reconstructed skin, DSCs migrate to the basement membrane zone and differentiate into melanocytes. These findings, combined with the identification of NGFRp75-positive cells in the dermis of human foreskin, which are devoid of hair, suggest that DSCs may be a self-renewing source of extrafollicular epidermal melanocytes. In this review, we discuss the properties of DSCs, the pathways required for melanocyte differentiation, and the value of 3D reconstructed skin to assess the behavior and contribution of DSCs in the naturalized environment of human skin. Potentially, DSCs provide a link to malignant melanoma by being a target of UVA-induced transformation.
Collapse
Affiliation(s)
- Susan E Zabierowski
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
21
|
The role of R-spondin2 in keratinocyte proliferation and epidermal thickening in keloid scarring. J Invest Dermatol 2010; 131:644-54. [PMID: 21160497 DOI: 10.1038/jid.2010.371] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Keloids are found only in humans and the underlying biochemical mechanisms of their pathogenesis remain unknown. R-spondins (Rspos) are a relatively new group of secreted proteins known to be Wnt/β-catenin signaling agonists, but their role in keloids has yet to be elucidated. We investigated the expression levels of R-spondin2 (Rspo2) in cell lysates and conditioned media of monocultures and co-cultures of fibroblasts and keratinocytes derived from keloids and normal skin. In this study we found increased protein expression and secretion of Rspo2 in respective monocultures of keloid fibroblasts and keratinocytes when compared with their normal counterparts. Double-chamber co-culture experiments implicated the role of keloid keratinocytes (KKs) in the induction of Rspo2 secretion from fibroblasts because of epithelial-mesenchymal interactions. Addition of recombinant human Rspo2 in culture increased the proliferation of keratinocytes and it acted synergistically with Wnt3a through the canonical Wnt/β-catenin pathway. Overexpression of Rspo2 in normal fibroblasts brought about thicker epidermis when compared with control fibroblasts in a skin organotypic culture model. This observation coincides with the hyperproliferative phenotype of thickened epidermis seen in keloids. Taken together, the results suggest the possible double paracrine action of KKs in inducing higher expression of Rspo2 in fibroblasts that promotes keratinocyte proliferation and epidermal thickening.
Collapse
|
22
|
Dong J, He Y, Zhang X, Wang L, Sun T, Zhang M, Liang Y, Qi M. Calcitonin gene-related peptide regulates the growth of epidermal stem cells in vitro. Peptides 2010; 31:1860-5. [PMID: 20654671 DOI: 10.1016/j.peptides.2010.07.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Revised: 07/07/2010] [Accepted: 07/07/2010] [Indexed: 11/21/2022]
Abstract
Epidermal stem cells are characterized as slow-cycling, multi-potent, self-renewing cells that not only maintain somatic homeostasis, but also participate in tissue regeneration and repair. Various factors can influence the growth of epidermal stem cells. Recently, dysregulation of epidermal stem cells has been reported to be involved in epidermal hyperproliferative diseases and skin tumors. To determine the effect of calcitonin gene-related protein (CGRP), a cutaneous nerve neuropeptide, on the growth of human epidermal stem cells, epidermal stem cells were isolated from human skin and cultured in vitro. Epidermal stem cells grow well and maintain a high proliferative ability in Epilife medium, and express high levels of β1-integrin. CGRP (10(-8) M) can promote epidermal stem cells to enter the S phase and increase the number of bromodeoxyuridine (BrdU)-labeled cells; the expression of β-catenin and c-myc genes are deregulated during this process, which can be compromised by CGRP8-37 peptide, an antagonist of CGRP receptor. Experimental evidence suggests that epidermal stem cells can be cultured in vitro for a period of time with preservation of stem cell characteristics. CGRP can stimulate epidermal stem cells to detach from their niche, break quiescence, and undergo division; β-catenin and c-myc may functionally be involved in the process.
Collapse
Affiliation(s)
- Jie Dong
- Beijing Chaoyang Hospital, Affiliated to Capital Medical University, Beijing, PR China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Song D, Liu X, Liu R, Yang L, Zuo J, Liu W. Connexin 43 hemichannel regulates H9c2 cell proliferation by modulating intracellular ATP and [Ca2+]. Acta Biochim Biophys Sin (Shanghai) 2010; 42:472-82. [PMID: 20705586 DOI: 10.1093/abbs/gmq047] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Connexin 43 (Cx43), known to be the main protein building blocks of gap junctions and hemichannels in mammalian heart, plays an important role in cardiocytes proliferation. Gap junctional intercellular communication has been suggested to be necessary for cellular proliferation and differentiation. However, the effect of Cx43 hemichannel on cardiocytes proliferation and the mechanism remain unclear. In this study, rat heart cell line H9c2 was used. The Cx43 location, the proliferation rate and hemichannel activity of H9c2 cells and Wnt-3a(+)-H9c2 cells were investigated and the changes of intracellular ATP and [Ca(2+)] were determined. Results showed that the inhibited hemichannel induced by 18beta-glycyrrhetinic acid (GA) evoked intracellular ATP and [Ca(2+)] increase and enhanced H9c2 cell proliferation. Wnt-3a(+)-H9c2 cells displayed enhanced hemichannel activity and proliferation rate. Inhibited hemichannel of Wnt-3a(+)-H9c2 cells induced by 18beta-GA decreased intracellular ATP, increased [Ca(2+)], and enhanced the proliferation of H9c2 cells. This study validated the role of hemichannel in H9c2 cell proliferation regulation, and showed a mechanism involved in the regulation of H9c2 cell proliferation. The proliferation could be enhanced by Cx43 hemichannel-mediated ATP release accompanying intracellular [Ca(2+)] change. However, different changes of ATP were observed in Wnt-3a(+)-H9c2 cells. These findings provided new insights into the molecular mechanisms of proliferation regulation in H9c2 cells and the effect of Wnt-3a on intracellular ATP.
Collapse
Affiliation(s)
- Dongli Song
- Department of Cellular and Genetic Medicine, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
24
|
Desmosomes in developing human epidermis. Dermatol Res Pract 2010; 2010:698761. [PMID: 20592759 PMCID: PMC2879547 DOI: 10.1155/2010/698761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 04/02/2010] [Indexed: 11/17/2022] Open
Abstract
Desmosomes play important roles in the cell differentiation and morphogenesis of tissues. Studies on animal models have greatly increased our knowledge on epidermal development while reports on human developing skin are rare due to the difficult accessibility to the samples. Although the morphology of periderm cells and the process how the epidermis develops very much resemble each other, the timetable and the final outcome of a mature human epidermis markedly differ from those of murine skin. Even the genetic basis of the junctional components may have profound differences between the species, which might affect the implementation of the data from animal models in human studies. The aim of this review is to focus on the development of human skin with special emphasis on desmosomes. Desmosomal development is mirrored in perspective with other simultaneous events, such as maturation of adherens, tight and gap junctions, and the basement membrane zone.
Collapse
|
25
|
Li L, Fukunaga-Kalabis M, Yu H, Xu X, Kong J, Lee JT, Herlyn M. Human dermal stem cells differentiate into functional epidermal melanocytes. J Cell Sci 2010; 123:853-60. [PMID: 20159965 DOI: 10.1242/jcs.061598] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Melanocytes sustain a lifelong proliferative potential, but a stem cell reservoir in glabrous skin has not yet been found. Here, we show that multipotent dermal stem cells isolated from human foreskins lacking hair follicles are able to home to the epidermis to differentiate into melanocytes. These dermal stem cells, grown as three-dimensional spheres, displayed a capacity for self-renewal and expressed NGFRp75, nestin and OCT4, but not melanocyte markers. In addition, cells derived from single-cell clones were able to differentiate into multiple lineages including melanocytes. In a three-dimensional skin equivalent model, sphere-forming cells differentiated into HMB45-positive melanocytes, which migrated from the dermis to the epidermis and aligned singly among the basal layer keratinocytes in a similar fashion to pigmented melanocytes isolated from the epidermis. The dermal stem cells were negative for E-cadherin and N-cadherin, whereas they acquired E-cadherin expression and lost NGFRp75 expression upon contact with epidermal keratinocytes. These results demonstrate that stem cells in the dermis of human skin with neural-crest-like characteristics can become mature epidermal melanocytes. This finding could significantly change our understanding of the etiological factors in melanocyte transformation and pigmentation disorders; specifically, that early epigenetic or genetic alterations leading to transformation may take place in the dermis rather than in the epidermis.
Collapse
Affiliation(s)
- Ling Li
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Zhao L, Rooker SM, Morrell N, Leucht P, Simanovskii D, Helms JA. Controlling the in vivo activity of Wnt liposomes. Methods Enzymol 2009; 465:331-47. [PMID: 19913175 DOI: 10.1016/s0076-6879(09)65017-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Liposomes offer a method of delivering small molecules, nucleic acids, and proteins to sites within the body. Typically, bioactive materials are encapsulated within the liposomal aqueous core and liposomal phase transition is elicited by pH or temperature changes. We developed a new class of liposomes for the in vivo delivery of lipid-modified proteins. First, we show that the inclusion of a chromophore into the liposomal or vesosomal membrane renders these lipid vesicles extremely sensitive to very small (muJ) changes in energy. Next, we demonstrate that the lipid-modified Wnt protein is not encapsulated within a liposome but rather is tethered to the exoliposomal surface in an active configuration. When applied to intact skin, chromophore-modified liposomes do not penetrate past the corneal layer of the epidermis, but remain localized to the site of application. Injury to the epidermis allows rapid penetration of liposomes into the dermis, which suggests that mild forms of dermabrasion will greatly enhance transdermal delivery of liposome-packaged molecules. Finally, we demonstrate that topical application of Wnt3a liposomes rapidly stimulates proliferation of cells in the corneal layer, resulting in a thicker, more fibrillous epidermis.
Collapse
Affiliation(s)
- L Zhao
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | | | | | | | | | | |
Collapse
|
27
|
Karow M, Popp T, Egea V, Ries C, Jochum M, Neth P. Wnt signalling in mouse mesenchymal stem cells: impact on proliferation, invasion and MMP expression. J Cell Mol Med 2008; 13:2506-2520. [PMID: 19413884 DOI: 10.1111/j.1582-4934.2008.00619.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Based on the capacity of mesenchymal stem cells (MSC) to differentiate into multiple cell types in vitro and in vivo, MCS may be a suitable source for cell therapy and regeneration strategies. A prerequisite for effective clinical applications of human MSC (hMSC) is a profound knowledge of signal transduction cascades that mediate processes like proliferation, targeted migration and differentiation. Recently, we identified the canonical Wnt signal transduction pathway as a key player in hMSC proliferation and invasion. To evaluate whether those findings are transferable to the equivalent counterparts in mice, we studied important steps in the wingless/int-1 (Wnt) signal transduction pathway in mouse MSC (mMSC) and mMSC carrying a T cell specific transcription factor (TCF)/lymphoid enhancer binding factor (LEF)-reporter transgene. We found that the induction of the canonical Wnt pathway resulted in the up-regulation of the known Wnt target gene cyclin D1, closely associated with an enhanced proliferation capacity of mMSC. Interestingly, the expression of the Wnt target gene membrane type 1-matrix metalloproteinase (MT1-MMP) was diminished in mMSC upon Wnt3a stimulation, which came along with an impaired invasion. In line with these findings, MMP-2 and MMP-9 expression levels in mMSC were also decreased after Wnt3a treatment. In contrast, inhibition of Wnt signalling by the knockdown of the transcriptional activator beta-catenin resulted in an up-regulation of MT1-MMP and mMSC invasion. By comparing these findings with the settings in hMSC, major differences in Wnt-regulated MMP expression were observed in mMSC. Thus, our data advice caution when mouse model systems represent the pre-clinical validation of MSC-mediated therapeutical approaches.
Collapse
Affiliation(s)
- Marisa Karow
- Division of Clinical Chemistry and Clinical Biochemistry, Department of Surgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Tanja Popp
- Division of Clinical Chemistry and Clinical Biochemistry, Department of Surgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Virginia Egea
- Division of Clinical Chemistry and Clinical Biochemistry, Department of Surgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Christian Ries
- Division of Clinical Chemistry and Clinical Biochemistry, Department of Surgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Marianne Jochum
- Division of Clinical Chemistry and Clinical Biochemistry, Department of Surgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Peter Neth
- Division of Clinical Chemistry and Clinical Biochemistry, Department of Surgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
28
|
Effect of Wnt signaling pathway on wound healing. Biochem Biophys Res Commun 2008; 378:149-51. [PMID: 19013436 DOI: 10.1016/j.bbrc.2008.11.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2008] [Accepted: 11/06/2008] [Indexed: 12/15/2022]
Abstract
Wnt signaling pathway has been divided into two subclasses: the canonical pathway (Wnt/beta-catenin pathway) and the non-canonical pathway. It has been proven that Wnt/beta-catenin pathway can enhance wound healing, and some glycoprotein of Wnt family may directly or indirectly improve wound healing.
Collapse
|
29
|
Leucht P, Minear S, Ten Berge D, Nusse R, Helms JA. Translating insights from development into regenerative medicine: the function of Wnts in bone biology. Semin Cell Dev Biol 2008; 19:434-43. [PMID: 18824114 DOI: 10.1016/j.semcdb.2008.09.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2008] [Revised: 08/20/2008] [Accepted: 09/02/2008] [Indexed: 11/25/2022]
Abstract
The Wnt pathway constitutes one of the most attractive candidates for modulating skeletal tissue regeneration based on its functions during skeletal development and homeostasis. Wnts participate in every stage of skeletogenesis, from the self-renewal and proliferation of skeletal stem cells to the specification of osteochondroprogenitor cells and the maturation of chondrocytes and osteoblasts. We propose that the function of Wnts depend upon a skeletogenic cell's state of differentiation. In this review we summarize recent data with a focus on the roles of Wnt signaling in mesenchymal stem cell fate, osteoprogenitor cell differentiation, chondrocyte maturation, bone remodeling, and bone regeneration.
Collapse
Affiliation(s)
- P Leucht
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|