Cargando…

Alterations to mTORC1 signaling in the skeletal muscle differentially affect whole-body metabolism

BACKGROUND: The mammalian target of rapamycin complex 1 (mTORC1) is a central node in a network of signaling pathways controlling cell growth and survival. This multiprotein complex integrates external signals and affects different nutrient pathways in various organs. However, it is not clear how al...

Descripción completa

Detalles Bibliográficos
Autores principales: Guridi, Maitea, Kupr, Barbara, Romanino, Klaas, Lin, Shuo, Falcetta, Denis, Tintignac, Lionel, Rüegg, Markus A.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4800774/
https://www.ncbi.nlm.nih.gov/pubmed/27004103
http://dx.doi.org/10.1186/s13395-016-0084-8
_version_ 1782422503689289728
author Guridi, Maitea
Kupr, Barbara
Romanino, Klaas
Lin, Shuo
Falcetta, Denis
Tintignac, Lionel
Rüegg, Markus A.
author_facet Guridi, Maitea
Kupr, Barbara
Romanino, Klaas
Lin, Shuo
Falcetta, Denis
Tintignac, Lionel
Rüegg, Markus A.
author_sort Guridi, Maitea
collection PubMed
description BACKGROUND: The mammalian target of rapamycin complex 1 (mTORC1) is a central node in a network of signaling pathways controlling cell growth and survival. This multiprotein complex integrates external signals and affects different nutrient pathways in various organs. However, it is not clear how alterations of mTORC1 signaling in skeletal muscle affect whole-body metabolism. RESULTS: We characterized the metabolic phenotype of young and old raptor muscle knock-out (RAmKO) and TSC1 muscle knock-out (TSCmKO) mice, where mTORC1 activity in skeletal muscle is inhibited or constitutively activated, respectively. Ten-week-old RAmKO mice are lean and insulin resistant with increased energy expenditure, and they are resistant to a high-fat diet (HFD). This correlates with an increased expression of histone deacetylases (HDACs) and a downregulation of genes involved in glucose and fatty acid metabolism. Ten-week-old TSCmKO mice are also lean, glucose intolerant with a decreased activation of protein kinase B (Akt/PKB) targets that regulate glucose transporters in the muscle. The mice are resistant to a HFD and show reduced accumulation of glycogen and lipids in the liver. Both mouse models suffer from a myopathy with age, with reduced fat and lean mass, and both RAmKO and TSCmKO mice develop insulin resistance and increased intramyocellular lipid content. CONCLUSIONS: Our study shows that alterations of mTORC1 signaling in the skeletal muscle differentially affect whole-body metabolism. While both inhibition and constitutive activation of mTORC1 induce leanness and resistance to obesity, changes in the metabolism of muscle and peripheral organs are distinct. These results indicate that a balanced mTORC1 signaling in the muscle is required for proper metabolic homeostasis. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s13395-016-0084-8) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-4800774
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-48007742016-03-22 Alterations to mTORC1 signaling in the skeletal muscle differentially affect whole-body metabolism Guridi, Maitea Kupr, Barbara Romanino, Klaas Lin, Shuo Falcetta, Denis Tintignac, Lionel Rüegg, Markus A. Skelet Muscle Research BACKGROUND: The mammalian target of rapamycin complex 1 (mTORC1) is a central node in a network of signaling pathways controlling cell growth and survival. This multiprotein complex integrates external signals and affects different nutrient pathways in various organs. However, it is not clear how alterations of mTORC1 signaling in skeletal muscle affect whole-body metabolism. RESULTS: We characterized the metabolic phenotype of young and old raptor muscle knock-out (RAmKO) and TSC1 muscle knock-out (TSCmKO) mice, where mTORC1 activity in skeletal muscle is inhibited or constitutively activated, respectively. Ten-week-old RAmKO mice are lean and insulin resistant with increased energy expenditure, and they are resistant to a high-fat diet (HFD). This correlates with an increased expression of histone deacetylases (HDACs) and a downregulation of genes involved in glucose and fatty acid metabolism. Ten-week-old TSCmKO mice are also lean, glucose intolerant with a decreased activation of protein kinase B (Akt/PKB) targets that regulate glucose transporters in the muscle. The mice are resistant to a HFD and show reduced accumulation of glycogen and lipids in the liver. Both mouse models suffer from a myopathy with age, with reduced fat and lean mass, and both RAmKO and TSCmKO mice develop insulin resistance and increased intramyocellular lipid content. CONCLUSIONS: Our study shows that alterations of mTORC1 signaling in the skeletal muscle differentially affect whole-body metabolism. While both inhibition and constitutive activation of mTORC1 induce leanness and resistance to obesity, changes in the metabolism of muscle and peripheral organs are distinct. These results indicate that a balanced mTORC1 signaling in the muscle is required for proper metabolic homeostasis. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s13395-016-0084-8) contains supplementary material, which is available to authorized users. BioMed Central 2016-03-21 /pmc/articles/PMC4800774/ /pubmed/27004103 http://dx.doi.org/10.1186/s13395-016-0084-8 Text en © Guridi et al. 2016 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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
Guridi, Maitea
Kupr, Barbara
Romanino, Klaas
Lin, Shuo
Falcetta, Denis
Tintignac, Lionel
Rüegg, Markus A.
Alterations to mTORC1 signaling in the skeletal muscle differentially affect whole-body metabolism
title Alterations to mTORC1 signaling in the skeletal muscle differentially affect whole-body metabolism
title_full Alterations to mTORC1 signaling in the skeletal muscle differentially affect whole-body metabolism
title_fullStr Alterations to mTORC1 signaling in the skeletal muscle differentially affect whole-body metabolism
title_full_unstemmed Alterations to mTORC1 signaling in the skeletal muscle differentially affect whole-body metabolism
title_short Alterations to mTORC1 signaling in the skeletal muscle differentially affect whole-body metabolism
title_sort alterations to mtorc1 signaling in the skeletal muscle differentially affect whole-body metabolism
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4800774/
https://www.ncbi.nlm.nih.gov/pubmed/27004103
http://dx.doi.org/10.1186/s13395-016-0084-8
work_keys_str_mv AT guridimaitea alterationstomtorc1signalingintheskeletalmuscledifferentiallyaffectwholebodymetabolism
AT kuprbarbara alterationstomtorc1signalingintheskeletalmuscledifferentiallyaffectwholebodymetabolism
AT romaninoklaas alterationstomtorc1signalingintheskeletalmuscledifferentiallyaffectwholebodymetabolism
AT linshuo alterationstomtorc1signalingintheskeletalmuscledifferentiallyaffectwholebodymetabolism
AT falcettadenis alterationstomtorc1signalingintheskeletalmuscledifferentiallyaffectwholebodymetabolism
AT tintignaclionel alterationstomtorc1signalingintheskeletalmuscledifferentiallyaffectwholebodymetabolism
AT rueggmarkusa alterationstomtorc1signalingintheskeletalmuscledifferentiallyaffectwholebodymetabolism