Cargando…

HMGB1 Promotes In Vitro and In Vivo Skeletal Muscle Atrophy through an IL-18-Dependent Mechanism

Skeletal muscle atrophy occurs due to muscle wasting or reductions in protein associated with aging, injury, and inflammatory processes. High-mobility group box-1 (HMGB1) protein is passively released from necrotic cells and actively secreted by inflammatory cells, and is implicated in the pathogene...

Descripción completa

Detalles Bibliográficos
Autores principales: Ho, Trung-Loc, Tang, Chih-Hsin, Chang, Sunny Li-Yun, Tsai, Chun-Hao, Chen, Hsien-Te, Su, Chen-Ming
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9740799/
https://www.ncbi.nlm.nih.gov/pubmed/36497194
http://dx.doi.org/10.3390/cells11233936
_version_ 1784848155742830592
author Ho, Trung-Loc
Tang, Chih-Hsin
Chang, Sunny Li-Yun
Tsai, Chun-Hao
Chen, Hsien-Te
Su, Chen-Ming
author_facet Ho, Trung-Loc
Tang, Chih-Hsin
Chang, Sunny Li-Yun
Tsai, Chun-Hao
Chen, Hsien-Te
Su, Chen-Ming
author_sort Ho, Trung-Loc
collection PubMed
description Skeletal muscle atrophy occurs due to muscle wasting or reductions in protein associated with aging, injury, and inflammatory processes. High-mobility group box-1 (HMGB1) protein is passively released from necrotic cells and actively secreted by inflammatory cells, and is implicated in the pathogenesis of various inflammatory and immune diseases. HMGB1 is upregulated in muscle inflammation, and circulating levels of the proinflammatory cytokine interleukin-18 (IL-18) are upregulated in patients with sarcopenia, a muscle-wasting disease. We examined whether an association exists between HMGB1 and IL-18 signaling in skeletal muscle atrophy. HMGB1-induced increases of IL-18 levels enhanced the expression of muscle atrophy markers and inhibited myogenic marker expression in C2C12 and G7 myoblast cell lines. HMGB1-induced increases of IL-18 production in C2C12 cells involved the RAGE/p85/Akt/mTOR/c-Jun signaling pathway. HMGB1 short hairpin RNA (shRNA) treatment rescued the expression of muscle-specific differentiation markers in murine C2C12 myotubes and in mice with glycerol-induced muscle atrophy. HMGB1 and IL-18 signaling was suppressed in the mice after HMGB1 shRNA treatment. These findings suggest that the HMGB1/IL-18 axis is worth targeting for the treatment of skeletal muscle atrophy.
format Online
Article
Text
id pubmed-9740799
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-97407992022-12-11 HMGB1 Promotes In Vitro and In Vivo Skeletal Muscle Atrophy through an IL-18-Dependent Mechanism Ho, Trung-Loc Tang, Chih-Hsin Chang, Sunny Li-Yun Tsai, Chun-Hao Chen, Hsien-Te Su, Chen-Ming Cells Article Skeletal muscle atrophy occurs due to muscle wasting or reductions in protein associated with aging, injury, and inflammatory processes. High-mobility group box-1 (HMGB1) protein is passively released from necrotic cells and actively secreted by inflammatory cells, and is implicated in the pathogenesis of various inflammatory and immune diseases. HMGB1 is upregulated in muscle inflammation, and circulating levels of the proinflammatory cytokine interleukin-18 (IL-18) are upregulated in patients with sarcopenia, a muscle-wasting disease. We examined whether an association exists between HMGB1 and IL-18 signaling in skeletal muscle atrophy. HMGB1-induced increases of IL-18 levels enhanced the expression of muscle atrophy markers and inhibited myogenic marker expression in C2C12 and G7 myoblast cell lines. HMGB1-induced increases of IL-18 production in C2C12 cells involved the RAGE/p85/Akt/mTOR/c-Jun signaling pathway. HMGB1 short hairpin RNA (shRNA) treatment rescued the expression of muscle-specific differentiation markers in murine C2C12 myotubes and in mice with glycerol-induced muscle atrophy. HMGB1 and IL-18 signaling was suppressed in the mice after HMGB1 shRNA treatment. These findings suggest that the HMGB1/IL-18 axis is worth targeting for the treatment of skeletal muscle atrophy. MDPI 2022-12-06 /pmc/articles/PMC9740799/ /pubmed/36497194 http://dx.doi.org/10.3390/cells11233936 Text en © 2022 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
Ho, Trung-Loc
Tang, Chih-Hsin
Chang, Sunny Li-Yun
Tsai, Chun-Hao
Chen, Hsien-Te
Su, Chen-Ming
HMGB1 Promotes In Vitro and In Vivo Skeletal Muscle Atrophy through an IL-18-Dependent Mechanism
title HMGB1 Promotes In Vitro and In Vivo Skeletal Muscle Atrophy through an IL-18-Dependent Mechanism
title_full HMGB1 Promotes In Vitro and In Vivo Skeletal Muscle Atrophy through an IL-18-Dependent Mechanism
title_fullStr HMGB1 Promotes In Vitro and In Vivo Skeletal Muscle Atrophy through an IL-18-Dependent Mechanism
title_full_unstemmed HMGB1 Promotes In Vitro and In Vivo Skeletal Muscle Atrophy through an IL-18-Dependent Mechanism
title_short HMGB1 Promotes In Vitro and In Vivo Skeletal Muscle Atrophy through an IL-18-Dependent Mechanism
title_sort hmgb1 promotes in vitro and in vivo skeletal muscle atrophy through an il-18-dependent mechanism
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9740799/
https://www.ncbi.nlm.nih.gov/pubmed/36497194
http://dx.doi.org/10.3390/cells11233936
work_keys_str_mv AT hotrungloc hmgb1promotesinvitroandinvivoskeletalmuscleatrophythroughanil18dependentmechanism
AT tangchihhsin hmgb1promotesinvitroandinvivoskeletalmuscleatrophythroughanil18dependentmechanism
AT changsunnyliyun hmgb1promotesinvitroandinvivoskeletalmuscleatrophythroughanil18dependentmechanism
AT tsaichunhao hmgb1promotesinvitroandinvivoskeletalmuscleatrophythroughanil18dependentmechanism
AT chenhsiente hmgb1promotesinvitroandinvivoskeletalmuscleatrophythroughanil18dependentmechanism
AT suchenming hmgb1promotesinvitroandinvivoskeletalmuscleatrophythroughanil18dependentmechanism