A Novel Hepatic Anti-Fibrotic Strategy Utilizing the Secretome Released from Etanercept-Synthesizing Adipose-Derived Stem Cells

Tumor necrosis factor-α (TNF-α)-driven inflammatory reaction plays a crucial role in the initiation of liver fibrosis. We herein attempted to design genetically engineered adipose-derived stem cells (ASCs) producing etanercept (a potent TNF-α inhibitor), and to determine the anti-fibrotic potential...

Full description

Saved in:
Bibliographic Details
Published inInternational journal of molecular sciences Vol. 20; no. 24; p. 6302
Main Authors Han, Jae Hyun, Kim, Ok-Hee, Lee, Sang Chul, Kim, Kee-Hwan, Park, Jung Hyun, Lee, Jae Im, Lee, Kyung Hee, Hong, Ha-Eun, Seo, Haeyeon, Choi, Ho Joong, Ju, Ji Hyeon, Kim, Say-June
Format Journal Article
LanguageEnglish
Published Switzerland MDPI AG 13.12.2019
MDPI
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Tumor necrosis factor-α (TNF-α)-driven inflammatory reaction plays a crucial role in the initiation of liver fibrosis. We herein attempted to design genetically engineered adipose-derived stem cells (ASCs) producing etanercept (a potent TNF-α inhibitor), and to determine the anti-fibrotic potential of the secretome released from the etanercept-synthesizing ASCs (etanercept-secretome). First, we generated the etanercept-synthesizing ASCs by transfecting the ASCs with mini-circle plasmids containing the gene insert encoding for etanercept. We subsequently collected the secretory material released from the etanercept-synthesizing ASCs and determined its anti-fibrotic effects both in vitro (in thioacetamide [TAA]-treated AML12 and LX2 cells) and in vivo (in TAA-treated mice) models of liver fibrosis. We observed that while etanercept-secretome increased the viability of the TAA-treated AML12 hepatocytes ( = 0.021), it significantly decreased the viability of the TAA-treated LX2 HSCs ( = 0.021). In the liver of mice with liver fibrosis, intravenous administration of the etanercept-secretome induced significant reduction in the expression of both fibrosis-related and inflammation-related markers compared to the control group (all s < 0.05). The etanercept-secretome group also showed significantly lower serum levels of liver enzymes as well as pro-inflammatory cytokines, such as TNF-α ( = 0.020) and IL-6 ( = 0.021). Histological examination of the liver showed the highest reduction in the degree of fibrosis in the entanercept-secretome group ( = 0.006). Our results suggest that the administration of etanercept-secretome improves liver fibrosis by inhibiting TNF-α-driven inflammation in the mice with liver fibrosis. Thus, blocking TNF-α-driven inflammation at the appropriate stage of liver fibrosis could be an efficient strategy to prevent fibrosis.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:1422-0067
1661-6596
1422-0067
DOI:10.3390/ijms20246302