2015
DOI: 10.1530/joe-14-0490
|View full text |Cite
|
Sign up to set email alerts
|

Dexamethasone downregulates caveolin-1 causing muscle atrophy via inhibited insulin signaling

Abstract: Glucocorticoids play a major role in the development of muscle atrophy in various medical conditions, such as cancer, burn injury, and sepsis, by inhibiting insulin signaling. In this study, we report a new pathway in which glucocorticoids reduce the levels of upstream insulin signaling components by downregulating the transcription of the gene encoding caveolin-1 (CAV1), a scaffolding protein present in the caveolar membrane. Treatment with the glucocorticoid dexamethasone (DEX) decreased CAV1 protein and Cav… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
2
1

Citation Types

1
18
1

Year Published

2015
2015
2022
2022

Publication Types

Select...
8

Relationship

0
8

Authors

Journals

citations
Cited by 26 publications
(20 citation statements)
references
References 37 publications
(46 reference statements)
1
18
1
Order By: Relevance
“…As a generally applied model of skeletal muscle atrophy in vitro , the C2C12 murine myoblasts and primary HSMPCs were induced to differentiate into myotubes. The myotube atrophy and protein degradation were induced by Dexa . As shown in Figure A and B, ALN (0.1–1 μM) dose‐dependently promoted the enlargement in diameters of myotubes, which were over 20 and 40 μm of myotubes differentiated from C2C12 myoblast and HSMPCs, respectively.…”
Section: Resultsmentioning
confidence: 84%
See 1 more Smart Citation
“…As a generally applied model of skeletal muscle atrophy in vitro , the C2C12 murine myoblasts and primary HSMPCs were induced to differentiate into myotubes. The myotube atrophy and protein degradation were induced by Dexa . As shown in Figure A and B, ALN (0.1–1 μM) dose‐dependently promoted the enlargement in diameters of myotubes, which were over 20 and 40 μm of myotubes differentiated from C2C12 myoblast and HSMPCs, respectively.…”
Section: Resultsmentioning
confidence: 84%
“…The myotube atrophy and protein degradation were induced by Dexa. [29][30][31] As shown in Figure 1A Alendronate prevents Dexa-induced atrophy in C2C12-derived and human skeletal muscle-derived progenitor cell-derived myotubes via SIRT3 mediation Muscle atrophy in vitro is ordinarily determined by both the reduction of myotube diameters and the induction of ubiquitin ligase muscle atrophy F-box or Atrogin-1 expressions, which drive skeletal muscle specific protein degradation. 32 As shown in Figure 3A, myotubes were treated with 1 μM Dexa in the presence or absence of 0.1-1 μM ALN for 24 h. Dexamethasone significantly elevated the Atrogin-1 protein expressions in both C2C12-derived [ Figure 3A(a)] and HSMPC-derived [ Figure 3A(b)] myotubes, which could be significantly reversed by the treatment of ALN in a dosedependent manner.…”
Section: Resultsmentioning
confidence: 99%
“…A general principle for myoblast differentiation is contact inhibition and serum deprivation 18, followed by division, elongation, and fusion. Many studies have treated differentiated myotubes with DEX and showed atrophic effects 9, 11, 19. The molecular pathways involved in the atrophic process include the ubiquitin-proteasome system, the lysosomal system, and the calcium-dependent system 20.…”
Section: Discussionmentioning
confidence: 99%
“…GC response elements have been identified in the promoter of the CAV1 gene, and dexamethasone treatment reduced CAV1 protein and mRNA expression with a concomitant reduction in insulin receptor alpha (IRa) and IRS1 levels in C2C12 myotubes. In addition, CAV1 knockdown decreased the protein levels of IRa and IRS1, and overexpression of CAV1 prevented the dexamethasone-induced decrease in IRa and IRS1 proteins (217). GCs promote proteolytic degradation of myofibrillar and extracellular matrix proteins by inducing the expression of components of the ubiquitin and proteasome system; activating the lysosomal system, as suggested by the presence of markers of autophagy in the muscle of GC-treated animals, and stimulating the calcium-mediated proteolytic system (218).…”
Section: Muscle In Csmentioning
confidence: 92%