Cargando…

Mechanisms involved in follistatin‐induced hypertrophy and increased insulin action in skeletal muscle

BACKGROUND: Skeletal muscle wasting is often associated with insulin resistance. A major regulator of muscle mass is the transforming growth factor β (TGF‐β) superfamily, including activin A, which causes atrophy. TGF‐β superfamily ligands also negatively regulate insulin‐sensitive proteins, but whe...

Descripción completa

Detalles Bibliográficos
Autores principales: Han, Xiuqing, Møller, Lisbeth Liliendal Valbjørn, De Groote, Estelle, Bojsen‐Møller, Kirstine Nyvold, Davey, Jonathan, Henríquez‐Olguin, Carlos, Li, Zhencheng, Knudsen, Jonas Roland, Jensen, Thomas Elbenhardt, Madsbad, Sten, Gregorevic, Paul, Richter, Erik Arne, Sylow, Lykke
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7663972/
https://www.ncbi.nlm.nih.gov/pubmed/31402604
http://dx.doi.org/10.1002/jcsm.12474
_version_ 1783609750341025792
author Han, Xiuqing
Møller, Lisbeth Liliendal Valbjørn
De Groote, Estelle
Bojsen‐Møller, Kirstine Nyvold
Davey, Jonathan
Henríquez‐Olguin, Carlos
Li, Zhencheng
Knudsen, Jonas Roland
Jensen, Thomas Elbenhardt
Madsbad, Sten
Gregorevic, Paul
Richter, Erik Arne
Sylow, Lykke
author_facet Han, Xiuqing
Møller, Lisbeth Liliendal Valbjørn
De Groote, Estelle
Bojsen‐Møller, Kirstine Nyvold
Davey, Jonathan
Henríquez‐Olguin, Carlos
Li, Zhencheng
Knudsen, Jonas Roland
Jensen, Thomas Elbenhardt
Madsbad, Sten
Gregorevic, Paul
Richter, Erik Arne
Sylow, Lykke
author_sort Han, Xiuqing
collection PubMed
description BACKGROUND: Skeletal muscle wasting is often associated with insulin resistance. A major regulator of muscle mass is the transforming growth factor β (TGF‐β) superfamily, including activin A, which causes atrophy. TGF‐β superfamily ligands also negatively regulate insulin‐sensitive proteins, but whether this pathway contributes to insulin action remains to be determined. METHODS: To elucidate if TGF‐β superfamily ligands regulate insulin action, we used an adeno‐associated virus gene editing approach to overexpress an activin A inhibitor, follistatin (Fst288), in mouse muscle of lean and diet‐induced obese mice. We determined basal and insulin‐stimulated 2‐deoxy‐glucose uptake using isotopic tracers in vivo. Furthermore, to evaluate whether circulating Fst and activin A concentrations are associated with obesity, insulin resistance, and weight loss in humans, we analysed serum from morbidly obese subjects before, 1 week, and 1 year after Roux‐en‐Y gastric bypass (RYGB). RESULTS: Fst288 muscle overexpression markedly increased in vivo insulin‐stimulated (but not basal) glucose uptake (+75%, P < 0.05) and increased protein expression and intracellular insulin signalling of AKT, TBC1D4, PAK1, pyruvate dehydrogenase‐E1α, and p70S6K, while decreasing TBC1D1 signaling (P < 0.05). Fst288 increased both basal and insulin‐stimulated protein synthesis, but no correlation was observed between the Fst288‐driven hypertrophy and the increase in insulin‐stimulated glucose uptake. Importantly, Fst288 completely normalized muscle glucose uptake in insulin‐resistant diet‐induced obese mice. RYGB surgery doubled circulating Fst and reduced activin A (−24%, P < 0.05) concentration 1 week after surgery before any significant weight loss in morbidly obese normoglycemic patients, while major weight loss after 1 year did not further change the concentrations. CONCLUSIONS: We here present evidence that Fst is a potent regulator of insulin action in muscle, and in addition to AKT and p70S6K, we identify TBC1D1, TBC1D4, pyruvate dehydrogenase‐E1α, and PAK1 as Fst targets. Circulating Fst more than doubled post‐RYGB surgery, a treatment that markedly improved insulin sensitivity, suggesting a role for Fst in regulating glycaemic control. These findings demonstrate the therapeutic potential of inhibiting TGF‐β superfamily ligands to improve insulin action and Fst's relevance to muscle wasting‐associated insulin‐resistant conditions in mice and humans.
format Online
Article
Text
id pubmed-7663972
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-76639722020-11-17 Mechanisms involved in follistatin‐induced hypertrophy and increased insulin action in skeletal muscle Han, Xiuqing Møller, Lisbeth Liliendal Valbjørn De Groote, Estelle Bojsen‐Møller, Kirstine Nyvold Davey, Jonathan Henríquez‐Olguin, Carlos Li, Zhencheng Knudsen, Jonas Roland Jensen, Thomas Elbenhardt Madsbad, Sten Gregorevic, Paul Richter, Erik Arne Sylow, Lykke J Cachexia Sarcopenia Muscle Original Articles BACKGROUND: Skeletal muscle wasting is often associated with insulin resistance. A major regulator of muscle mass is the transforming growth factor β (TGF‐β) superfamily, including activin A, which causes atrophy. TGF‐β superfamily ligands also negatively regulate insulin‐sensitive proteins, but whether this pathway contributes to insulin action remains to be determined. METHODS: To elucidate if TGF‐β superfamily ligands regulate insulin action, we used an adeno‐associated virus gene editing approach to overexpress an activin A inhibitor, follistatin (Fst288), in mouse muscle of lean and diet‐induced obese mice. We determined basal and insulin‐stimulated 2‐deoxy‐glucose uptake using isotopic tracers in vivo. Furthermore, to evaluate whether circulating Fst and activin A concentrations are associated with obesity, insulin resistance, and weight loss in humans, we analysed serum from morbidly obese subjects before, 1 week, and 1 year after Roux‐en‐Y gastric bypass (RYGB). RESULTS: Fst288 muscle overexpression markedly increased in vivo insulin‐stimulated (but not basal) glucose uptake (+75%, P < 0.05) and increased protein expression and intracellular insulin signalling of AKT, TBC1D4, PAK1, pyruvate dehydrogenase‐E1α, and p70S6K, while decreasing TBC1D1 signaling (P < 0.05). Fst288 increased both basal and insulin‐stimulated protein synthesis, but no correlation was observed between the Fst288‐driven hypertrophy and the increase in insulin‐stimulated glucose uptake. Importantly, Fst288 completely normalized muscle glucose uptake in insulin‐resistant diet‐induced obese mice. RYGB surgery doubled circulating Fst and reduced activin A (−24%, P < 0.05) concentration 1 week after surgery before any significant weight loss in morbidly obese normoglycemic patients, while major weight loss after 1 year did not further change the concentrations. CONCLUSIONS: We here present evidence that Fst is a potent regulator of insulin action in muscle, and in addition to AKT and p70S6K, we identify TBC1D1, TBC1D4, pyruvate dehydrogenase‐E1α, and PAK1 as Fst targets. Circulating Fst more than doubled post‐RYGB surgery, a treatment that markedly improved insulin sensitivity, suggesting a role for Fst in regulating glycaemic control. These findings demonstrate the therapeutic potential of inhibiting TGF‐β superfamily ligands to improve insulin action and Fst's relevance to muscle wasting‐associated insulin‐resistant conditions in mice and humans. John Wiley and Sons Inc. 2019-08-11 2019-12 /pmc/articles/PMC7663972/ /pubmed/31402604 http://dx.doi.org/10.1002/jcsm.12474 Text en © 2019 The Authors Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc-nd/4.0/ License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.
spellingShingle Original Articles
Han, Xiuqing
Møller, Lisbeth Liliendal Valbjørn
De Groote, Estelle
Bojsen‐Møller, Kirstine Nyvold
Davey, Jonathan
Henríquez‐Olguin, Carlos
Li, Zhencheng
Knudsen, Jonas Roland
Jensen, Thomas Elbenhardt
Madsbad, Sten
Gregorevic, Paul
Richter, Erik Arne
Sylow, Lykke
Mechanisms involved in follistatin‐induced hypertrophy and increased insulin action in skeletal muscle
title Mechanisms involved in follistatin‐induced hypertrophy and increased insulin action in skeletal muscle
title_full Mechanisms involved in follistatin‐induced hypertrophy and increased insulin action in skeletal muscle
title_fullStr Mechanisms involved in follistatin‐induced hypertrophy and increased insulin action in skeletal muscle
title_full_unstemmed Mechanisms involved in follistatin‐induced hypertrophy and increased insulin action in skeletal muscle
title_short Mechanisms involved in follistatin‐induced hypertrophy and increased insulin action in skeletal muscle
title_sort mechanisms involved in follistatin‐induced hypertrophy and increased insulin action in skeletal muscle
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7663972/
https://www.ncbi.nlm.nih.gov/pubmed/31402604
http://dx.doi.org/10.1002/jcsm.12474
work_keys_str_mv AT hanxiuqing mechanismsinvolvedinfollistatininducedhypertrophyandincreasedinsulinactioninskeletalmuscle
AT møllerlisbethliliendalvalbjørn mechanismsinvolvedinfollistatininducedhypertrophyandincreasedinsulinactioninskeletalmuscle
AT degrooteestelle mechanismsinvolvedinfollistatininducedhypertrophyandincreasedinsulinactioninskeletalmuscle
AT bojsenmøllerkirstinenyvold mechanismsinvolvedinfollistatininducedhypertrophyandincreasedinsulinactioninskeletalmuscle
AT daveyjonathan mechanismsinvolvedinfollistatininducedhypertrophyandincreasedinsulinactioninskeletalmuscle
AT henriquezolguincarlos mechanismsinvolvedinfollistatininducedhypertrophyandincreasedinsulinactioninskeletalmuscle
AT lizhencheng mechanismsinvolvedinfollistatininducedhypertrophyandincreasedinsulinactioninskeletalmuscle
AT knudsenjonasroland mechanismsinvolvedinfollistatininducedhypertrophyandincreasedinsulinactioninskeletalmuscle
AT jensenthomaselbenhardt mechanismsinvolvedinfollistatininducedhypertrophyandincreasedinsulinactioninskeletalmuscle
AT madsbadsten mechanismsinvolvedinfollistatininducedhypertrophyandincreasedinsulinactioninskeletalmuscle
AT gregorevicpaul mechanismsinvolvedinfollistatininducedhypertrophyandincreasedinsulinactioninskeletalmuscle
AT richtererikarne mechanismsinvolvedinfollistatininducedhypertrophyandincreasedinsulinactioninskeletalmuscle
AT sylowlykke mechanismsinvolvedinfollistatininducedhypertrophyandincreasedinsulinactioninskeletalmuscle