Cargando…

Activation of Akt–mTORC1 signalling reverts cancer‐dependent muscle wasting

BACKGROUND: Cancer‐related muscle wasting occurs in most cancer patients. An important regulator of adult muscle mass and function is the Akt–mTORC1 pathway. While Akt–mTORC1 signalling is important for adult muscle homeostasis, it is also a major target of numerous cancer treatments. Which role Akt...

Descripción completa

Detalles Bibliográficos
Autores principales: Geremia, Alessia, Sartori, Roberta, Baraldo, Martina, Nogara, Leonardo, Balmaceda, Valeria, Dumitras, Georgia Ana, Ciciliot, Stefano, Scalabrin, Marco, Nolte, Hendrik, Blaauw, Bert
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8818597/
https://www.ncbi.nlm.nih.gov/pubmed/34741441
http://dx.doi.org/10.1002/jcsm.12854
_version_ 1784645857872707584
author Geremia, Alessia
Sartori, Roberta
Baraldo, Martina
Nogara, Leonardo
Balmaceda, Valeria
Dumitras, Georgia Ana
Ciciliot, Stefano
Scalabrin, Marco
Nolte, Hendrik
Blaauw, Bert
author_facet Geremia, Alessia
Sartori, Roberta
Baraldo, Martina
Nogara, Leonardo
Balmaceda, Valeria
Dumitras, Georgia Ana
Ciciliot, Stefano
Scalabrin, Marco
Nolte, Hendrik
Blaauw, Bert
author_sort Geremia, Alessia
collection PubMed
description BACKGROUND: Cancer‐related muscle wasting occurs in most cancer patients. An important regulator of adult muscle mass and function is the Akt–mTORC1 pathway. While Akt–mTORC1 signalling is important for adult muscle homeostasis, it is also a major target of numerous cancer treatments. Which role Akt–mTORC1 signalling plays during cancer cachexia in muscle is currently not known. Here, we aimed to determine how activation or inactivation of the pathway affects skeletal muscle during cancer cachexia. METHODS: We used inducible, muscle‐specific Raptor ko (mTORC1) mice to determine the effect of reduced mTOR signalling during cancer cachexia. On the contrary, in order to understand if skeletal muscles maintain their anabolic capacity and if activation of Akt–mTORC1 signalling can reverse cancer cachexia, we generated mice in which we can inducibly activate Akt specifically in skeletal muscles. RESULTS: We found that mTORC1 signalling is impaired during cancer cachexia, using the Lewis lung carcinoma and C26 colon cancer model, and is accompanied by a reduction in protein synthesis rates of 57% (P < 0.01). Further reduction of mTOR signalling, as seen in Raptor ko animals, leads to a 1.5‐fold increase in autophagic flux (P > 0.001), but does not further increase muscle wasting. On the other hand, activation of Akt–mTORC1 signalling in already cachectic animals completely reverses the 15–20% loss in muscle mass and force (P < 0.001). Interestingly, Akt activation only in skeletal muscle completely normalizes the transcriptional deregulation observed in cachectic muscle, despite having no effect on tumour size or spleen mass. In addition to stimulating muscle growth, it is also sufficient to prevent the increase in protein degradation normally observed in muscles from tumour‐bearing animals. CONCLUSIONS: Here, we show that activation of Akt–mTORC1 signalling is sufficient to completely revert cancer‐dependent muscle wasting. Intriguingly, these results show that skeletal muscle maintains its anabolic capacities also during cancer cachexia, possibly giving a rationale behind some of the beneficial effects observed in exercise in cancer patients.
format Online
Article
Text
id pubmed-8818597
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-88185972022-02-09 Activation of Akt–mTORC1 signalling reverts cancer‐dependent muscle wasting Geremia, Alessia Sartori, Roberta Baraldo, Martina Nogara, Leonardo Balmaceda, Valeria Dumitras, Georgia Ana Ciciliot, Stefano Scalabrin, Marco Nolte, Hendrik Blaauw, Bert J Cachexia Sarcopenia Muscle Original Articles: Basic Science BACKGROUND: Cancer‐related muscle wasting occurs in most cancer patients. An important regulator of adult muscle mass and function is the Akt–mTORC1 pathway. While Akt–mTORC1 signalling is important for adult muscle homeostasis, it is also a major target of numerous cancer treatments. Which role Akt–mTORC1 signalling plays during cancer cachexia in muscle is currently not known. Here, we aimed to determine how activation or inactivation of the pathway affects skeletal muscle during cancer cachexia. METHODS: We used inducible, muscle‐specific Raptor ko (mTORC1) mice to determine the effect of reduced mTOR signalling during cancer cachexia. On the contrary, in order to understand if skeletal muscles maintain their anabolic capacity and if activation of Akt–mTORC1 signalling can reverse cancer cachexia, we generated mice in which we can inducibly activate Akt specifically in skeletal muscles. RESULTS: We found that mTORC1 signalling is impaired during cancer cachexia, using the Lewis lung carcinoma and C26 colon cancer model, and is accompanied by a reduction in protein synthesis rates of 57% (P < 0.01). Further reduction of mTOR signalling, as seen in Raptor ko animals, leads to a 1.5‐fold increase in autophagic flux (P > 0.001), but does not further increase muscle wasting. On the other hand, activation of Akt–mTORC1 signalling in already cachectic animals completely reverses the 15–20% loss in muscle mass and force (P < 0.001). Interestingly, Akt activation only in skeletal muscle completely normalizes the transcriptional deregulation observed in cachectic muscle, despite having no effect on tumour size or spleen mass. In addition to stimulating muscle growth, it is also sufficient to prevent the increase in protein degradation normally observed in muscles from tumour‐bearing animals. CONCLUSIONS: Here, we show that activation of Akt–mTORC1 signalling is sufficient to completely revert cancer‐dependent muscle wasting. Intriguingly, these results show that skeletal muscle maintains its anabolic capacities also during cancer cachexia, possibly giving a rationale behind some of the beneficial effects observed in exercise in cancer patients. John Wiley and Sons Inc. 2021-11-06 2022-02 /pmc/articles/PMC8818597/ /pubmed/34741441 http://dx.doi.org/10.1002/jcsm.12854 Text en © 2021 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders. https://creativecommons.org/licenses/by/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Articles: Basic Science
Geremia, Alessia
Sartori, Roberta
Baraldo, Martina
Nogara, Leonardo
Balmaceda, Valeria
Dumitras, Georgia Ana
Ciciliot, Stefano
Scalabrin, Marco
Nolte, Hendrik
Blaauw, Bert
Activation of Akt–mTORC1 signalling reverts cancer‐dependent muscle wasting
title Activation of Akt–mTORC1 signalling reverts cancer‐dependent muscle wasting
title_full Activation of Akt–mTORC1 signalling reverts cancer‐dependent muscle wasting
title_fullStr Activation of Akt–mTORC1 signalling reverts cancer‐dependent muscle wasting
title_full_unstemmed Activation of Akt–mTORC1 signalling reverts cancer‐dependent muscle wasting
title_short Activation of Akt–mTORC1 signalling reverts cancer‐dependent muscle wasting
title_sort activation of akt–mtorc1 signalling reverts cancer‐dependent muscle wasting
topic Original Articles: Basic Science
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8818597/
https://www.ncbi.nlm.nih.gov/pubmed/34741441
http://dx.doi.org/10.1002/jcsm.12854
work_keys_str_mv AT geremiaalessia activationofaktmtorc1signallingrevertscancerdependentmusclewasting
AT sartoriroberta activationofaktmtorc1signallingrevertscancerdependentmusclewasting
AT baraldomartina activationofaktmtorc1signallingrevertscancerdependentmusclewasting
AT nogaraleonardo activationofaktmtorc1signallingrevertscancerdependentmusclewasting
AT balmacedavaleria activationofaktmtorc1signallingrevertscancerdependentmusclewasting
AT dumitrasgeorgiaana activationofaktmtorc1signallingrevertscancerdependentmusclewasting
AT ciciliotstefano activationofaktmtorc1signallingrevertscancerdependentmusclewasting
AT scalabrinmarco activationofaktmtorc1signallingrevertscancerdependentmusclewasting
AT noltehendrik activationofaktmtorc1signallingrevertscancerdependentmusclewasting
AT blaauwbert activationofaktmtorc1signallingrevertscancerdependentmusclewasting