Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Pharmacol ; 192: 114750, 2021 10.
Article in English | MEDLINE | ID: mdl-34461118

ABSTRACT

Statins decrease the serum LDL-cholesterol concentration and reduce the risk for cardiovascular diseases but can cause myopathy, which may be related to mTORC inhibition. In the current study, we investigated which mTORC is inhibited by simvastatin and by which mechanisms. In C2C12 myoblasts and myotubes and mouse gastrocnemius, simvastatin was cytotoxic and inhibited S6rp and Akt Ser473 phosphorylation, indicating inhibition of mTORC1 and mTORC2, respectively. In contrast to simvastatin, the mTORC1 inhibitor rapamycin did not inhibit mTORC2 activity and was not cytotoxic. Like simvastatin, knock-down of Rictor, an essential component of mTORC2, impaired Akt Ser473 and S6rp phosphorylation and was cytotoxic for C2C12 myoblasts, suggesting that mTORC2 inhibition is an important myotoxic mechanism. The investigation of the mechanism of mTORC2 inhibition showed that simvastatin impaired Ras farnesylation, which was prevented by farnesol but without restoring mTORC2 activity. In comparison, Rap1 knock-down reduced mTORC2 activity and was cytotoxic for C2C12 myoblasts. Simvastatin impaired Rap1 geranylgeranylation and function, which was prevented by geranylgeraniol. In addition, simvastatin and the complex III inhibitor antimycin A caused mitochondrial superoxide accumulation and impaired the activity of mTORC2, which could partially be prevented by the antioxidant MitoTEMPO. In conclusion, mTORC2 inhibition is an important mechanism of simvastatin-induced myotoxicity. Simvastatin inhibits mTORC2 by impairing geranylgeranylation of Rap1 and by inducing mitochondrial dysfunction.


Subject(s)
Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mitochondria/drug effects , Muscle, Skeletal/drug effects , Prenylation/drug effects , Simvastatin/toxicity , rap1 GTP-Binding Proteins/antagonists & inhibitors , Animals , Cell Line , Drug Delivery Systems/methods , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Male , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Prenylation/physiology , Simvastatin/administration & dosage , rap1 GTP-Binding Proteins/metabolism
2.
Biochem Pharmacol ; 190: 114649, 2021 08.
Article in English | MEDLINE | ID: mdl-34111424

ABSTRACT

Statins reduce cardiovascular complications in patients with high LDL-cholesterol but are associated with myopathy. We compared the toxicity of simvastatin of C2C12 myoblasts and myotubes. Since myoblasts can proliferate and fuse to myotubes, myoblasts can be considered as satellite cells and myotubes as mature muscle fibers. Simvastatin increased plasma membrane permeability and decreased the cellular ATP content in both myoblasts and myotubes, but with a stronger effect on myoblasts. While insulin prevented cytotoxicity up to 8 h after addition of simvastatin to myotubes, prevention in myoblasts required simultaneous addition. Mevalonate and geranylgeraniol prevented simvastatin-associated cytotoxicity in both myoblasts and myotubes. Simvastatin impaired the phosphorylation of the insulin receptor (IR ß), Akt ser473 and S6rp, and increased phosphorylation of AMPK thr172 in both myotubes and myoblasts, which was prevented by insulin and mevalonate. Simvastatin impaired oxygen consumption and increased superoxide production by myoblasts and myotubes and induced apoptosis via cytochrome c release. In addition, simvastatin impaired proliferation and fusion of myoblasts to myotubes by inhibiting the expression of the nuclear transcription factor MyoD and of the metalloprotease ADAM-12. Decreased expression of the proliferation factor Ki-67 and of ADAM-12 were also observed in gastrocnemius of mice treated with simvastatin. In conclusion, myoblasts were more susceptible to the toxic effects of simvastatin and simvastatin impaired myoblast proliferation and myotube formation. Impaired muscle regeneration may represent a new mechanism of statin myotoxicity.


Subject(s)
Cell Proliferation/drug effects , Muscle Fibers, Skeletal/drug effects , Simvastatin/pharmacology , Animals , Anticholesteremic Agents/pharmacology , Cell Survival/drug effects , Diterpenes/pharmacology , Male , Mevalonic Acid/pharmacology , Mice , Mice, Inbred C57BL , Random Allocation
3.
Pharmacol Res ; 154: 104201, 2020 04.
Article in English | MEDLINE | ID: mdl-30877064

ABSTRACT

Statins lower the serum low-density lipoprotein cholesterol and prevent cardiovascular events by inhibiting 3-hydroxy-3-methyl-glutaryl-CoA reductase. Although the safety of statins is documented, many patients ingesting statins may suffer from skeletal muscle-associated symptoms (SAMS). Importantly, SAMS are a common reason for stopping the treatment with statins. Statin-associated muscular symptoms include fatigue, weakness and pain, possibly accompanied by elevated serum creatine kinase activity. The most severe muscular adverse reaction is the potentially fatal rhabdomyolysis. The frequency of SAMS is variable but in up to 30% of the patients ingesting statins, depending on the population treated and the statin used. The mechanisms leading to SAMS are currently not completely clarified. Over the last 15 years, several research articles focused on statin-induced mitochondrial dysfunction as a reason for SAMS. Statins can impair the function of the mitochondrial respiratory chain, thereby reducing ATP and increasing ROS production. This can induce mitochondrial membrane permeability transition, release of cytochrome c into the cytosol and induce apoptosis. In parallel, statins inhibit activation of Akt, mainly due to reduced function of mTORC2, which may be related to mitochondrial dysfunction. Mitochondrial dysfunction by statins is also responsible for activation of AMPK, which is associated with impaired activation of mTORC1. Reduced activation of mTORC1 leads to increased skeletal muscle protein degradation, impaired protein synthesis and stimulation of apoptosis. In this paper, we discuss some of the different hypotheses how statins affect skeletal muscle in more detail, focusing particularly on those related to mitochondrial dysfunction and the impairment of the Akt/mTOR pathway.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscle, Skeletal/drug effects , Muscular Diseases/chemically induced , Animals , Humans , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism
4.
Sci Rep ; 9(1): 7409, 2019 05 15.
Article in English | MEDLINE | ID: mdl-31092879

ABSTRACT

Simvastatin is an inhibitor of the 3-hydroxy-3-methylglutaryl-CoA reductase used for decreasing low density lipoprotein (LDL)-cholesterol in patients. It is well-tolerated but can cause myopathy. Our aims were to enlarge our knowledge regarding mechanisms and effects of insulin on simvastatin-associated myotoxicity in C2C12 myotubes. Simvastatin (10 µM) reduced membrane integrity and ATP content in myotubes treated for 24 hours, which could be prevented and partially reversed concentration- and time-dependently by insulin. Furthermore, simvastatin impaired the phosphorylation of Akt (Protein Kinase B) mainly at Ser473 and less at Thr308, indicating impaired activity of the mammalian Target of Rapamycin Complex 2 (mTORC2). Impaired activation of Akt increased mRNA expression of the muscle atrophy F-Box (MAFbx), decreased activation of the mammalian Target of Rapamycin Complex 1 (mTORC1) and stimulated apoptosis by impairing the Ser9 phosphorylation of glycogen synthase kinase 3ß. Decreased phosphorylation of Akt at both phosphorylation sites and of downstream substrates as well as apoptosis were prevented concentration-dependently by insulin. In addition, simvastatin caused accumulation of the insulin receptor ß-chain in the endoplasmic reticulum (ER) and increased cleavage of procaspase-12, indicating ER stress. Insulin reduced the expression of the insulin receptor ß-chain but increased procaspase-12 activation in the presence of simvastatin. In conclusion, simvastatin impaired activation of Akt Ser473 most likely as a consequence of reduced activity of mTORC2. Insulin could prevent the effects of simvastatin on the insulin signaling pathway and on apoptosis, but not on the endoplasmic reticulum (ER) stress induction.


Subject(s)
Insulin/pharmacology , Muscle Cells/drug effects , Muscle, Skeletal/drug effects , Simvastatin/toxicity , Animals , Cell Line , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Heterocyclic Compounds, 3-Ring/toxicity , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/cytology , Receptor, Insulin/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Simvastatin/antagonists & inhibitors
5.
Int J Mol Sci ; 20(7)2019 Mar 28.
Article in English | MEDLINE | ID: mdl-30925718

ABSTRACT

Synthetic cathinones are popular psychoactive substances that may cause skeletal muscle damage. In addition to indirect sympathomimetic myotoxicity, these substances could be directly myotoxic. Since studies in myocytes are currently lacking, the aim of the present study was to investigate potential toxicological effects by synthetic cathinones on C2C12 myoblasts (mouse skeletal muscle cell line). We exposed C2C12 myoblasts to 3-methylmethcathinone, 4-methylmethcathinone (mephedrone), 3,4-methylenedioxymethcathinone (methylone), 3,4-methylenedioxypyrovalerone (MDPV), alpha-pyrrolidinovalerophenone (α-PVP), and naphthylpyrovalerone (naphyrone) for 1 or 24 h before cell membrane integrity, ATP content, mitochondrial oxygen consumption, and mitochondrial superoxide production was measured. 3,4-Methylenedioxymethamphetamine (MDMA) was included as a reference compound. All investigated synthetic cathinones, as well as MDMA, impaired cell membrane integrity, depleted ATP levels, and increased mitochondrial superoxide concentrations in a concentration-dependent manner in the range of 50⁻2000 µM. The two pyrovalerone derivatives α-PVP and naphyrone, and MDMA, additionally impaired basal and maximal cellular respiration, suggesting mitochondrial dysfunction. Alpha-PVP inhibited complex I, naphyrone complex II, and MDMA complex I and III, whereas complex IV was not affected. We conclude that, in addition to sympathetic nervous system effects and strenuous muscle exercise, direct effects of some cathinones on skeletal muscle mitochondria may contribute to myotoxicity in susceptible synthetic cathinone drugs users.


Subject(s)
Benzodioxoles/toxicity , Methamphetamine/analogs & derivatives , Myoblasts/drug effects , Pentanones/toxicity , Psychotropic Drugs/toxicity , Pyrrolidines/toxicity , Adenosine Triphosphate/metabolism , Animals , Cell Line , Methamphetamine/toxicity , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Myoblasts/metabolism , Myoblasts/pathology , Oxygen Consumption/drug effects , Superoxides/metabolism , Synthetic Cathinone
6.
Biochem Pharmacol ; 164: 23-33, 2019 06.
Article in English | MEDLINE | ID: mdl-30796916

ABSTRACT

Statins inhibit cholesterol biosynthesis and lower serum LDL-cholesterol levels. They are generally well tolerated, but can cause insulin resistance in patients. Therefore, we investigated the mechanisms underlying the statin-induced insulin resistance. We used mice and C2C12 myotubes (murine cell line): mice (n = 10) were treated with oral simvastatin (5 mg/kg/day) or water (control) for 21 days and C2C12 cells were exposed to 10 µM simvastatin for 24 h. After intraperitoneal glucose application (2 g/kg), simvastatin-treated mice had higher glucose but equal insulin plasma concentrations than controls and lower glucose transport into skeletal muscle. Similarly, glucose uptake by C2C12 myotubes exposed to 10 µM simvastatin for 24 h was impaired compared to control cells. In simvastatin-treated C2C12 myotubes, mRNA and protein expression of the insulin receptor (IR) ß-chain was increased, but the phosphorylation (Tyr1361) was impaired. Simvastatin decreased numerically Akt/PKB Thr308 phosphorylation (via insulin signaling pathway) and significantly Akt/PKB Ser473 phosphorylation (via mTORC2), which was explained by impaired phosphorylation of mTOR Ser2448. Reduced phosphorylation of Akt/PKB impaired downstream phosphorylation of GSK3ß, leading to impaired translocation of GLUT4 into plasma membranes of C2C12 myotubes. In contrast, reduced phosphorylation of AS160 could be excluded as a reason for impaired GLUT4 translocation. In conclusion, simvastatin caused insulin resistance in mice and impaired glucose uptake in C2C12 myotubes. The findings in myotubes can be explained by diminished activation of Akt/PKB by mTORC2 and downstream effects on GSK3ß, impairing the translocation of GLUT4 and the uptake of glucose.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Insulin Resistance/physiology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Simvastatin/toxicity , Animals , Cell Line , Glucose/antagonists & inhibitors , Glucose/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , Random Allocation
7.
Arch Toxicol ; 91(5): 2223-2234, 2017 May.
Article in English | MEDLINE | ID: mdl-27734117

ABSTRACT

Statins are generally well tolerated, but treatment with these drugs may be associated with myopathy. The mechanisms of statin-associated myopathy are not completely understood. Statins inhibit AKT phosphorylation by an unclear mechanism, whereas insulin-like growth factor (IGF-1) activates the IGF-1/AKT signaling pathway and promotes muscle growth. The aims of the study were to investigate mechanisms of impaired AKT phosphorylation by simvastatin and to assess effects of IGF-1 on simvastatin-induced myotoxicity in C2C12 myotubes. C2C12 mouse myotubes were exposed to 10 µM simvastatin and/or 10 ng/mL IGF-1 for 18 h. Simvastatin inhibited the IGF-1/AKT signaling pathway, resulting in increased breakdown of myofibrillar proteins, impaired protein synthesis and increased apoptosis. Simvastatin inhibited AKT S473 phosphorylation, indicating reduced activity of mTORC2. In addition, simvastatin impaired stimulation of AKT T308 phosphorylation by IGF-1, indicating reduced activation of the IGF-1R/PI3K pathway by IGF-1. Nevertheless, simvastatin-induced myotoxicity could be at least partially prevented by IGF-1. The protective effects of IGF-1 were mediated by activation of the IGF-1R/AKT signaling cascade. Treatment with IGF-1 also suppressed muscle atrophy markers, restored protein synthesis and inhibited apoptosis. These results were confirmed by normalization of myotube morphology and protein content of C2C12 cells exposed to simvastatin and treated with IGF-1. In conclusion, impaired activity of AKT can be explained by reduced function of mTORC2 and of the IGF-1R/PI3K pathway. IGF-1 can prevent simvastatin-associated cytotoxicity and metabolic effects on C2C12 cells. The study gives insight into mechanisms of simvastatin-associated myotoxicity and provides potential targets for therapeutic intervention.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Muscle Fibers, Skeletal/drug effects , Simvastatin/adverse effects , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Dose-Response Relationship, Drug , Forkhead Box Protein O3/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/administration & dosage , Mice , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism
8.
Biochim Biophys Acta ; 1853(8): 1841-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25913013

ABSTRACT

Statins are drugs that lower blood cholesterol levels and reduce cardiovascular morbidity and mortality. They are generally well-tolerated, but myopathy is a potentially severe adverse reaction of these compounds. The mechanisms by which statins induce myotoxicity are not completely understood, but may be related to inhibition of the AKT signaling pathway. The current studies were performed to explore the down-stream effects of the statin-associated inhibition of AKT within the AKT signaling pathway and on myocyte biology and morphology in C2C12 myotubes and in mice in vivo. We exposed C2C12 myotubes to 10 µM or 50 µM simvastatin, atorvastatin or rosuvastatin for 24 h. Simvastatin and atorvastatin inhibited AKT phosphorylation and were cytotoxic starting at 10 µM, whereas similar effects were observed for rosuvastatin at 50 µM. Inhibition of AKT phosphorylation was associated with impaired phosphorylation of S6 kinase, ribosomal protein S6, 4E-binding protein 1 and FoxO3a, resulting in reduced protein synthesis, accelerated myofibrillar degradation and atrophy of C2C12 myotubes. Furthermore, impaired AKT phosphorylation was associated with activation of caspases and PARP, reflecting induction of apoptosis. Similar findings were detected in skeletal muscle of mice treated orally with 5 mg/kg/day simvastatin for 3 weeks. In conclusion, this study highlights the importance of the AKT/mTOR signaling pathway in statin-induced myotoxicity and reveals potential drug targets for treatment of patients with statin-associated myopathies.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscle, Skeletal/drug effects , Muscular Diseases/chemically induced , Proto-Oncogene Proteins c-akt/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Apoptosis/drug effects , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiology , Muscular Diseases/metabolism , Muscular Diseases/pathology , Signal Transduction/physiology , Simvastatin/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL