Cargando…

1-Deoxysphingolipids, Early Predictors of Type 2 Diabetes, Compromise the Functionality of Skeletal Myoblasts

Metabolic dysfunction, dysregulated differentiation, and atrophy of skeletal muscle occur as part of a cluster of abnormalities associated with the development of Type 2 diabetes mellitus (T2DM). Recent interest has turned to the attention of the role of 1-deoxysphingolipids (1-DSL), atypical class...

Descripción completa

Detalles Bibliográficos
Autores principales: Tran, Duyen, Myers, Stephen, McGowan, Courtney, Henstridge, Darren, Eri, Rajaraman, Sonda, Sabrina, Caruso, Vanni
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8739520/
https://www.ncbi.nlm.nih.gov/pubmed/35002962
http://dx.doi.org/10.3389/fendo.2021.772925
_version_ 1784629117765812224
author Tran, Duyen
Myers, Stephen
McGowan, Courtney
Henstridge, Darren
Eri, Rajaraman
Sonda, Sabrina
Caruso, Vanni
author_facet Tran, Duyen
Myers, Stephen
McGowan, Courtney
Henstridge, Darren
Eri, Rajaraman
Sonda, Sabrina
Caruso, Vanni
author_sort Tran, Duyen
collection PubMed
description Metabolic dysfunction, dysregulated differentiation, and atrophy of skeletal muscle occur as part of a cluster of abnormalities associated with the development of Type 2 diabetes mellitus (T2DM). Recent interest has turned to the attention of the role of 1-deoxysphingolipids (1-DSL), atypical class of sphingolipids which are found significantly elevated in patients diagnosed with T2DM but also in the asymptomatic population who later develop T2DM. In vitro studies demonstrated that 1-DSL have cytotoxic properties and compromise the secretion of insulin from pancreatic beta cells. However, the role of 1-DSL on the functionality of skeletal muscle cells in the pathophysiology of T2DM still remains unclear. This study aimed to investigate whether 1-DSL are cytotoxic and disrupt the cellular processes of skeletal muscle precursors (myoblasts) and differentiated cells (myotubes) by performing a battery of in vitro assays including cell viability adenosine triphosphate assay, migration assay, myoblast fusion assay, glucose uptake assay, and immunocytochemistry. Our results demonstrated that 1-DSL significantly reduced the viability of myoblasts in a concentration and time-dependent manner, and induced apoptosis as well as cellular necrosis. Importantly, myoblasts were more sensitive to the cytotoxic effects induced by 1-DSL rather than by saturated fatty acids, such as palmitate, which are critical mediators of skeletal muscle dysfunction in T2DM. Additionally, 1-DSL significantly reduced the migration ability of myoblasts and the differentiation process of myoblasts into myotubes. 1-DSL also triggered autophagy in myoblasts and significantly reduced insulin-stimulated glucose uptake in myotubes. These findings demonstrate that 1-DSL directly compromise the functionality of skeletal muscle cells and suggest that increased levels of 1-DSL observed during the development of T2DM are likely to contribute to the pathophysiology of muscle dysfunction detected in this disease.
format Online
Article
Text
id pubmed-8739520
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-87395202022-01-08 1-Deoxysphingolipids, Early Predictors of Type 2 Diabetes, Compromise the Functionality of Skeletal Myoblasts Tran, Duyen Myers, Stephen McGowan, Courtney Henstridge, Darren Eri, Rajaraman Sonda, Sabrina Caruso, Vanni Front Endocrinol (Lausanne) Endocrinology Metabolic dysfunction, dysregulated differentiation, and atrophy of skeletal muscle occur as part of a cluster of abnormalities associated with the development of Type 2 diabetes mellitus (T2DM). Recent interest has turned to the attention of the role of 1-deoxysphingolipids (1-DSL), atypical class of sphingolipids which are found significantly elevated in patients diagnosed with T2DM but also in the asymptomatic population who later develop T2DM. In vitro studies demonstrated that 1-DSL have cytotoxic properties and compromise the secretion of insulin from pancreatic beta cells. However, the role of 1-DSL on the functionality of skeletal muscle cells in the pathophysiology of T2DM still remains unclear. This study aimed to investigate whether 1-DSL are cytotoxic and disrupt the cellular processes of skeletal muscle precursors (myoblasts) and differentiated cells (myotubes) by performing a battery of in vitro assays including cell viability adenosine triphosphate assay, migration assay, myoblast fusion assay, glucose uptake assay, and immunocytochemistry. Our results demonstrated that 1-DSL significantly reduced the viability of myoblasts in a concentration and time-dependent manner, and induced apoptosis as well as cellular necrosis. Importantly, myoblasts were more sensitive to the cytotoxic effects induced by 1-DSL rather than by saturated fatty acids, such as palmitate, which are critical mediators of skeletal muscle dysfunction in T2DM. Additionally, 1-DSL significantly reduced the migration ability of myoblasts and the differentiation process of myoblasts into myotubes. 1-DSL also triggered autophagy in myoblasts and significantly reduced insulin-stimulated glucose uptake in myotubes. These findings demonstrate that 1-DSL directly compromise the functionality of skeletal muscle cells and suggest that increased levels of 1-DSL observed during the development of T2DM are likely to contribute to the pathophysiology of muscle dysfunction detected in this disease. Frontiers Media S.A. 2021-12-24 /pmc/articles/PMC8739520/ /pubmed/35002962 http://dx.doi.org/10.3389/fendo.2021.772925 Text en Copyright © 2021 Tran, Myers, McGowan, Henstridge, Eri, Sonda and Caruso https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Endocrinology
Tran, Duyen
Myers, Stephen
McGowan, Courtney
Henstridge, Darren
Eri, Rajaraman
Sonda, Sabrina
Caruso, Vanni
1-Deoxysphingolipids, Early Predictors of Type 2 Diabetes, Compromise the Functionality of Skeletal Myoblasts
title 1-Deoxysphingolipids, Early Predictors of Type 2 Diabetes, Compromise the Functionality of Skeletal Myoblasts
title_full 1-Deoxysphingolipids, Early Predictors of Type 2 Diabetes, Compromise the Functionality of Skeletal Myoblasts
title_fullStr 1-Deoxysphingolipids, Early Predictors of Type 2 Diabetes, Compromise the Functionality of Skeletal Myoblasts
title_full_unstemmed 1-Deoxysphingolipids, Early Predictors of Type 2 Diabetes, Compromise the Functionality of Skeletal Myoblasts
title_short 1-Deoxysphingolipids, Early Predictors of Type 2 Diabetes, Compromise the Functionality of Skeletal Myoblasts
title_sort 1-deoxysphingolipids, early predictors of type 2 diabetes, compromise the functionality of skeletal myoblasts
topic Endocrinology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8739520/
https://www.ncbi.nlm.nih.gov/pubmed/35002962
http://dx.doi.org/10.3389/fendo.2021.772925
work_keys_str_mv AT tranduyen 1deoxysphingolipidsearlypredictorsoftype2diabetescompromisethefunctionalityofskeletalmyoblasts
AT myersstephen 1deoxysphingolipidsearlypredictorsoftype2diabetescompromisethefunctionalityofskeletalmyoblasts
AT mcgowancourtney 1deoxysphingolipidsearlypredictorsoftype2diabetescompromisethefunctionalityofskeletalmyoblasts
AT henstridgedarren 1deoxysphingolipidsearlypredictorsoftype2diabetescompromisethefunctionalityofskeletalmyoblasts
AT erirajaraman 1deoxysphingolipidsearlypredictorsoftype2diabetescompromisethefunctionalityofskeletalmyoblasts
AT sondasabrina 1deoxysphingolipidsearlypredictorsoftype2diabetescompromisethefunctionalityofskeletalmyoblasts
AT carusovanni 1deoxysphingolipidsearlypredictorsoftype2diabetescompromisethefunctionalityofskeletalmyoblasts