Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cosmet Investig Dermatol ; 17: 1049-1058, 2024.
Article in English | MEDLINE | ID: mdl-38737946

ABSTRACT

Introduction: Angiotensin (1-7) (Ang-(1-7)) is an emerging component of the renin-angiotensin system (RAS) with effective anti-fibrosis properties and has been shown to interfere with epithelial-mesenchymal transition (EMT) by numerous studies. In recent years, EMT has been proposed as a new therapeutic target for skin fibrotic diseases such as keloids. However, the effect of Ang-(1-7) on EMT in skin is still unclear. Hence, the purpose of this study was to explore the effect of Ang-(1-7) on Transforming growth factor-ß1(TGF-ß1)-induced EMT of human immortalized keratinocytes HaCaT in vitro. Methods: The study involved the use of the human immortalized keratinocyte cell line (HaCaT). The cells were cultured in high-glucose DMEM medium with 10% fetal bovine serum and 1% penicillin-streptomycin. Four groups were created for experimentation: control group (Group C), TGF-ß1-treated group (Group T), Ang-(1-7)-treated group (Group A), and a group treated with both TGF-ß1 and Ang-(1-7) (Group A + T). Various assays were conducted, including a cell proliferation assay using CCK-8 solution, a scratch wound healing assay to evaluate cell migration, and Western blotting to detect protein expressions related to cell characteristics. Additionally, quantitative real-time polymerase chain reaction (PCR) was performed to analyze epithelial-mesenchymal transition (EMT) related gene expression levels. The study aimed to investigate the effects of TGF-ß1 and Ang-(1-7) on HaCaT cells. Results: We found that Ang-(1-7) not only reduced the migration of HaCaT cells induced by TGF-ß1 in vitro but also reduced the expression of α-SMA and vimentin, and restored the protein expression of E-cadherin and claudin-1. Mechanistically, Ang-(1-7) inhibits the phosphorylation levels of Smad2 and Smad3 in the TGF-ß1 canonical pathway, and suppresses the expression of EMT-related transcription factors (EMT-TFs) such as SNAI2, TWIST1, and ZEB1. Discussion: Taken together, our findings suggest that Ang-(1-7) inhibits TGF-ß1-induced EMT in HaCaT cells in vitro by disrupting the TGF-ß1-Smad canonical signaling pathway. These results may be helpful in the treatment of EMT in skin fibrotic diseases such as keloids.

3.
Acta Histochem ; 125(4): 152042, 2023 May.
Article in English | MEDLINE | ID: mdl-37137202

ABSTRACT

This study aimed to investigate the effect of hypoxia on the anti-inflammatory effect of adipose-derived mesenchymal stem cells (AMSCs) in vitro and its possible mechanism. AMSCs were cultured in vitro in a hypoxic environment with 3% O2, and a normoxic (21% O2) environment was used as the control. The cells were identified by in vitro adipogenic and osteogenic differentiation and cell surface antigen detection, and the cell viability were detected. The effect of hypoxic AMSCs on macrophage inflammation was analyzed by co-culture. The results showed that under hypoxia, AMSCs had better viability, significantly downregulated the expression of inflammatory factors, alleviated macrophage inflammation, and activated the PI3K/AKT/HIF-1α pathway.


Subject(s)
Mesenchymal Stem Cells , Proto-Oncogene Proteins c-akt , Humans , Cell Hypoxia/physiology , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/metabolism , Obesity/metabolism , Osteogenesis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
4.
Exp Cell Res ; 415(1): 113109, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35318048

ABSTRACT

Tissue engineering technology will be the main approach to tissue regeneration in the future and are promising for the treatment of large-area burns and refractory wounds. Dedifferentiated fat cells (DFAT) derived from mature adipocytes (MAs) as a seed cell have great potential in cell therapy and tissue engineering for the treatment of a variety of clinical diseases because of their wider availability, stronger proliferation ability, multidirectional redifferentiation potential, higher cell purity, lower heterogeneity, and greater biosafety profile. However, the triggering mechanism for MAs reprogramming in vitro is unclear. In this study, MAs were successfully induced to dedifferentiate into DFAT in a short time in vitro using an "improved ceiling culture method". Flow cytometry, adipogenic, and osteogenic differentiation experiments verified that DFAT cells present the biological characteristics of stem cells. In addition, changing the stiffness of the extracellular matrix can inhibit the dedifferentiation of MAs to DFAT, and increase the expression of Yes-associated protein/transcriptional co-activator with the PDZ-binding motif (YAP/TAZ), nuclear translocation, and the expression of reprogramming transcription factors. In conclusion, extracellular matrix stiffness can induce MAs to dedifferentiate into DFAT in vitro, and can directly transmit mechanical force signals to the nucleus via YAP/TAZ binding to trigger the expression of stem cell-related reprogramming factors.


Subject(s)
Cell Dedifferentiation , Osteogenesis , Adipocytes/metabolism , Adipogenesis , Cell Differentiation , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...