Cargando…

Thymosin Beta 4 Inhibits LPS and ATP-Induced Hepatic Stellate Cells via the Regulation of Multiple Signaling Pathways

Risk signals are characteristic of many common inflammatory diseases and can function to activate nucleotide-binding oligomerization (NLR) family pyrin domain-containing 3 (NLRP3), the innate immune signal receptor in cytoplasm. The NLRP3 inflammasome plays an important role in the development of li...

Descripción completa

Detalles Bibliográficos
Autores principales: Choi, Jihye, Cho, Yunsang, Choi, Hwal, Lee, Sangmin, Han, Hyeju, Lee, Jeonghyeon, Kwon, Jungkee
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9959661/
https://www.ncbi.nlm.nih.gov/pubmed/36834849
http://dx.doi.org/10.3390/ijms24043439
_version_ 1784895332563288064
author Choi, Jihye
Cho, Yunsang
Choi, Hwal
Lee, Sangmin
Han, Hyeju
Lee, Jeonghyeon
Kwon, Jungkee
author_facet Choi, Jihye
Cho, Yunsang
Choi, Hwal
Lee, Sangmin
Han, Hyeju
Lee, Jeonghyeon
Kwon, Jungkee
author_sort Choi, Jihye
collection PubMed
description Risk signals are characteristic of many common inflammatory diseases and can function to activate nucleotide-binding oligomerization (NLR) family pyrin domain-containing 3 (NLRP3), the innate immune signal receptor in cytoplasm. The NLRP3 inflammasome plays an important role in the development of liver fibrosis. Activated NLRP3 nucleates the assembly of inflammasomes, leading to the secretion of interleukin (IL)-1β and IL-18, the activation of caspase-1, and the initiation of the inflammatory process. Therefore, it is essential to inhibit the activation of the NLRP3 inflammasome, which plays a vital role in the immune response and in initiating inflammation. RAW 264.7 and LX-2 cells were primed with lipopolysaccharide (LPS) for 4 h and subsequently stimulated for 30 min with 5 mM of adenosine 5′-triphosphate (ATP) to activate the NLRP3 inflammasome. Thymosin beta 4 (Tβ4) was supplemented to RAW264.7 and LX-2 cells 30 min before ATP was added. As a result, we investigated the effects of Tβ4 on the NLRP3 inflammasome. Tβ4 prevented LPS-induced NLRP3 priming by inhibiting NF-kB and JNK/p38 MAPK expression and the LPS and ATP-induced production of reactive oxygen species. Moreover, Tβ4 induced autophagy by controlling autophagy markers (LC3A/B and p62) through the inhibition of the PI3K/AKT/mTOR pathway. LPS combined with ATP significantly increased thee protein expression of inflammatory mediators and NLRP3 inflammasome markers. These events were remarkably suppressed by Tβ4. In conclusion, Tβ4 attenuated NLRP3 inflammasomes by inhibiting NLRP3 inflammasome-related proteins (NLRP3, ASC, IL-1β, and caspase-1). Our results indicate that Tβ4 attenuated the NLRP3 inflammasome through multiple signaling pathway regulations in macrophage and hepatic stellate cells. Therefore, based on the above findings, it is hypothesized that Tβ4 could be a potential inflammatory therapeutic agent targeting the NLRP3 inflammasome in hepatic fibrosis regulation.
format Online
Article
Text
id pubmed-9959661
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-99596612023-02-26 Thymosin Beta 4 Inhibits LPS and ATP-Induced Hepatic Stellate Cells via the Regulation of Multiple Signaling Pathways Choi, Jihye Cho, Yunsang Choi, Hwal Lee, Sangmin Han, Hyeju Lee, Jeonghyeon Kwon, Jungkee Int J Mol Sci Article Risk signals are characteristic of many common inflammatory diseases and can function to activate nucleotide-binding oligomerization (NLR) family pyrin domain-containing 3 (NLRP3), the innate immune signal receptor in cytoplasm. The NLRP3 inflammasome plays an important role in the development of liver fibrosis. Activated NLRP3 nucleates the assembly of inflammasomes, leading to the secretion of interleukin (IL)-1β and IL-18, the activation of caspase-1, and the initiation of the inflammatory process. Therefore, it is essential to inhibit the activation of the NLRP3 inflammasome, which plays a vital role in the immune response and in initiating inflammation. RAW 264.7 and LX-2 cells were primed with lipopolysaccharide (LPS) for 4 h and subsequently stimulated for 30 min with 5 mM of adenosine 5′-triphosphate (ATP) to activate the NLRP3 inflammasome. Thymosin beta 4 (Tβ4) was supplemented to RAW264.7 and LX-2 cells 30 min before ATP was added. As a result, we investigated the effects of Tβ4 on the NLRP3 inflammasome. Tβ4 prevented LPS-induced NLRP3 priming by inhibiting NF-kB and JNK/p38 MAPK expression and the LPS and ATP-induced production of reactive oxygen species. Moreover, Tβ4 induced autophagy by controlling autophagy markers (LC3A/B and p62) through the inhibition of the PI3K/AKT/mTOR pathway. LPS combined with ATP significantly increased thee protein expression of inflammatory mediators and NLRP3 inflammasome markers. These events were remarkably suppressed by Tβ4. In conclusion, Tβ4 attenuated NLRP3 inflammasomes by inhibiting NLRP3 inflammasome-related proteins (NLRP3, ASC, IL-1β, and caspase-1). Our results indicate that Tβ4 attenuated the NLRP3 inflammasome through multiple signaling pathway regulations in macrophage and hepatic stellate cells. Therefore, based on the above findings, it is hypothesized that Tβ4 could be a potential inflammatory therapeutic agent targeting the NLRP3 inflammasome in hepatic fibrosis regulation. MDPI 2023-02-08 /pmc/articles/PMC9959661/ /pubmed/36834849 http://dx.doi.org/10.3390/ijms24043439 Text en © 2023 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Choi, Jihye
Cho, Yunsang
Choi, Hwal
Lee, Sangmin
Han, Hyeju
Lee, Jeonghyeon
Kwon, Jungkee
Thymosin Beta 4 Inhibits LPS and ATP-Induced Hepatic Stellate Cells via the Regulation of Multiple Signaling Pathways
title Thymosin Beta 4 Inhibits LPS and ATP-Induced Hepatic Stellate Cells via the Regulation of Multiple Signaling Pathways
title_full Thymosin Beta 4 Inhibits LPS and ATP-Induced Hepatic Stellate Cells via the Regulation of Multiple Signaling Pathways
title_fullStr Thymosin Beta 4 Inhibits LPS and ATP-Induced Hepatic Stellate Cells via the Regulation of Multiple Signaling Pathways
title_full_unstemmed Thymosin Beta 4 Inhibits LPS and ATP-Induced Hepatic Stellate Cells via the Regulation of Multiple Signaling Pathways
title_short Thymosin Beta 4 Inhibits LPS and ATP-Induced Hepatic Stellate Cells via the Regulation of Multiple Signaling Pathways
title_sort thymosin beta 4 inhibits lps and atp-induced hepatic stellate cells via the regulation of multiple signaling pathways
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9959661/
https://www.ncbi.nlm.nih.gov/pubmed/36834849
http://dx.doi.org/10.3390/ijms24043439
work_keys_str_mv AT choijihye thymosinbeta4inhibitslpsandatpinducedhepaticstellatecellsviatheregulationofmultiplesignalingpathways
AT choyunsang thymosinbeta4inhibitslpsandatpinducedhepaticstellatecellsviatheregulationofmultiplesignalingpathways
AT choihwal thymosinbeta4inhibitslpsandatpinducedhepaticstellatecellsviatheregulationofmultiplesignalingpathways
AT leesangmin thymosinbeta4inhibitslpsandatpinducedhepaticstellatecellsviatheregulationofmultiplesignalingpathways
AT hanhyeju thymosinbeta4inhibitslpsandatpinducedhepaticstellatecellsviatheregulationofmultiplesignalingpathways
AT leejeonghyeon thymosinbeta4inhibitslpsandatpinducedhepaticstellatecellsviatheregulationofmultiplesignalingpathways
AT kwonjungkee thymosinbeta4inhibitslpsandatpinducedhepaticstellatecellsviatheregulationofmultiplesignalingpathways