Cargando…

Lipopolysaccharide preconditioning of adipose-derived stem cells improves liver-regenerating activity of the secretome

INTRODUCTION: Growing recognition of paracrine mechanisms in stem cell plasticity has resulted in considerable interest in stem cell-derived secretome. The aim of this study was to investigate the effects of lipopolysaccharide (LPS) preconditioning on the composition and hepatic regenerative activit...

Descripción completa

Detalles Bibliográficos
Autores principales: Lee, Sang Chul, Jeong, Hye Jin, Lee, Sang Kuon, Kim, Say-June
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4416308/
https://www.ncbi.nlm.nih.gov/pubmed/25890074
http://dx.doi.org/10.1186/s13287-015-0072-7
_version_ 1782369214336598016
author Lee, Sang Chul
Jeong, Hye Jin
Lee, Sang Kuon
Kim, Say-June
author_facet Lee, Sang Chul
Jeong, Hye Jin
Lee, Sang Kuon
Kim, Say-June
author_sort Lee, Sang Chul
collection PubMed
description INTRODUCTION: Growing recognition of paracrine mechanisms in stem cell plasticity has resulted in considerable interest in stem cell-derived secretome. The aim of this study was to investigate the effects of lipopolysaccharide (LPS) preconditioning on the composition and hepatic regenerative activity of adipose-derived stem cell (ASC) secretome. METHODS: Conditioned medium (CM) and LPS-CM were obtained after culturing human ASCs without or with low-dose LPS (0.5 ng/mL) for 24 hours. Untreated and thioacetamide-treated mouse AML12 hepatocytes were incubated for 24 hours with the control medium, LPS (0.5 ng/mL), CM, and LPS-CM and then cell viabilities were compared. CM and LPS-CM were also intravenously administered to partially hepatectomized mice, and their effects on liver regeneration were assessed by using liver weight measurements, immunohistochemistry, and Western blotting. RESULTS: In the in vitro experiments, LPS preconditioning of ASCs enhanced the mRNA expression levels of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), hepatocyte growth factor, and vascular endothelial growth factor, which evoke inflammatory response or liver regeneration. LPS-CM significantly promoted thioacetamide-damaged AML12 cell viability compared with CM-incubated cells and the control cells (77%, 69%, and 65% P <0.05). In the in vivo experiment, LPS-CM infusion into the partially hepatectomized mice significantly reduced serum IL-6 and TNF-α levels compared with the other groups (P <0.05) on days 1 and 2 after partial hepatectomy. Moreover, LPS-CM infusion enhanced liver regeneration (based on the liver weight changes at day 7 after partial hepatectomy, 3.73% versus 3.22% in the CM group; P <0.05) and significantly reduced the elevated serum levels of aspartate transaminase and alanine transaminase (at day 1, P <0.05). CONCLUSIONS: Our results suggest that LPS preconditioning effectively stimulates ASCs to produce the secretome beneficial to hepatic regeneration. Thus, optimizing ASC secretome profile by LPS preconditioning could be a promising approach to treat liver diseases by using stem cells.
format Online
Article
Text
id pubmed-4416308
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-44163082015-05-02 Lipopolysaccharide preconditioning of adipose-derived stem cells improves liver-regenerating activity of the secretome Lee, Sang Chul Jeong, Hye Jin Lee, Sang Kuon Kim, Say-June Stem Cell Res Ther Research INTRODUCTION: Growing recognition of paracrine mechanisms in stem cell plasticity has resulted in considerable interest in stem cell-derived secretome. The aim of this study was to investigate the effects of lipopolysaccharide (LPS) preconditioning on the composition and hepatic regenerative activity of adipose-derived stem cell (ASC) secretome. METHODS: Conditioned medium (CM) and LPS-CM were obtained after culturing human ASCs without or with low-dose LPS (0.5 ng/mL) for 24 hours. Untreated and thioacetamide-treated mouse AML12 hepatocytes were incubated for 24 hours with the control medium, LPS (0.5 ng/mL), CM, and LPS-CM and then cell viabilities were compared. CM and LPS-CM were also intravenously administered to partially hepatectomized mice, and their effects on liver regeneration were assessed by using liver weight measurements, immunohistochemistry, and Western blotting. RESULTS: In the in vitro experiments, LPS preconditioning of ASCs enhanced the mRNA expression levels of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), hepatocyte growth factor, and vascular endothelial growth factor, which evoke inflammatory response or liver regeneration. LPS-CM significantly promoted thioacetamide-damaged AML12 cell viability compared with CM-incubated cells and the control cells (77%, 69%, and 65% P <0.05). In the in vivo experiment, LPS-CM infusion into the partially hepatectomized mice significantly reduced serum IL-6 and TNF-α levels compared with the other groups (P <0.05) on days 1 and 2 after partial hepatectomy. Moreover, LPS-CM infusion enhanced liver regeneration (based on the liver weight changes at day 7 after partial hepatectomy, 3.73% versus 3.22% in the CM group; P <0.05) and significantly reduced the elevated serum levels of aspartate transaminase and alanine transaminase (at day 1, P <0.05). CONCLUSIONS: Our results suggest that LPS preconditioning effectively stimulates ASCs to produce the secretome beneficial to hepatic regeneration. Thus, optimizing ASC secretome profile by LPS preconditioning could be a promising approach to treat liver diseases by using stem cells. BioMed Central 2015-04-14 /pmc/articles/PMC4416308/ /pubmed/25890074 http://dx.doi.org/10.1186/s13287-015-0072-7 Text en © Lee et al.; licensee BioMed Central. 2015 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Lee, Sang Chul
Jeong, Hye Jin
Lee, Sang Kuon
Kim, Say-June
Lipopolysaccharide preconditioning of adipose-derived stem cells improves liver-regenerating activity of the secretome
title Lipopolysaccharide preconditioning of adipose-derived stem cells improves liver-regenerating activity of the secretome
title_full Lipopolysaccharide preconditioning of adipose-derived stem cells improves liver-regenerating activity of the secretome
title_fullStr Lipopolysaccharide preconditioning of adipose-derived stem cells improves liver-regenerating activity of the secretome
title_full_unstemmed Lipopolysaccharide preconditioning of adipose-derived stem cells improves liver-regenerating activity of the secretome
title_short Lipopolysaccharide preconditioning of adipose-derived stem cells improves liver-regenerating activity of the secretome
title_sort lipopolysaccharide preconditioning of adipose-derived stem cells improves liver-regenerating activity of the secretome
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4416308/
https://www.ncbi.nlm.nih.gov/pubmed/25890074
http://dx.doi.org/10.1186/s13287-015-0072-7
work_keys_str_mv AT leesangchul lipopolysaccharidepreconditioningofadiposederivedstemcellsimprovesliverregeneratingactivityofthesecretome
AT jeonghyejin lipopolysaccharidepreconditioningofadiposederivedstemcellsimprovesliverregeneratingactivityofthesecretome
AT leesangkuon lipopolysaccharidepreconditioningofadiposederivedstemcellsimprovesliverregeneratingactivityofthesecretome
AT kimsayjune lipopolysaccharidepreconditioningofadiposederivedstemcellsimprovesliverregeneratingactivityofthesecretome