Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Endocrinol Metab Clin North Am ; 41(2): 297-322, vi, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22682632

ABSTRACT

This review describes the current literature on the interaction between insulin-like growth factors, endocrine hormones, and branched-chain amino acids on muscle physiology in healthy young individuals and during select pathologic conditions. Emphasis is placed on the mechanism by which physical and hormonal signals are transduced at the cellular level to either grow or atrophy skeletal muscle. The key role of the mammalian target of rapamycin and its ability to respond to hypertrophic and atrophic signals informs our understanding how a combination of physical, nutritional, and pharmacologic therapies may be used in tandem to prevent or ameliorate reductions in muscle mass.


Subject(s)
Muscle, Skeletal/metabolism , Somatomedins/metabolism , TOR Serine-Threonine Kinases/metabolism , Amino Acids, Branched-Chain/metabolism , Androgens/metabolism , Animals , Exercise/physiology , Female , Follistatin/metabolism , Glucocorticoids/metabolism , Humans , Male , Mice , Muscle, Skeletal/growth & development , Muscular Atrophy/metabolism , Myostatin/metabolism , Rats
2.
Physiology (Bethesda) ; 26(2): 83-96, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21487027

ABSTRACT

The mammalian target of rapamycin (mTOR) is an evolutionarily conserved protein kinase that exquisitely regulates protein metabolism in skeletal muscle. mTOR integrates input from amino acids, growth factors, and intracellular cues to make or break muscle protein. mTOR accomplishes this task by stimulating the phosphorylation of substrates that control protein translation while simultaneously inhibiting proteasomal and autophagic protein degradation. In a metabolic twist of fate, sepsis induces muscle atrophy in part by the aberrant regulation of mTOR. In this review, we track the steps of normal mTOR signaling in muscle and examine where they go astray in sepsis and inflammation.


Subject(s)
Muscle, Skeletal/metabolism , Myositis/metabolism , Sepsis/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Humans , Inflammation/metabolism , Inflammation/pathology , Muscle, Skeletal/pathology , Myositis/pathology , Sepsis/pathology , Signal Transduction
3.
Shock ; 35(2): 117-25, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20577146

ABSTRACT

Sepsis-induced muscle atrophy is produced in part by decreased protein synthesis mediated by inhibition of mTOR (mammalian target of rapamycin). The present study tests the hypothesis that alteration of specific protein-protein interactions within the mTORC1 (mTOR complex 1) contributes to the decreased mTOR activity observed after cecal ligation and puncture in rats. Sepsis decreased in vivo translational efficiency in gastrocnemius and reduced the phosphorylation of eukaryotic initiation factor (eIF) 4E-binding protein (BP) 1, S6 kinase (S6K) 1, and mTOR, compared with time-matched pair-fed controls. Sepsis decreased T246-phosphorylated PRAS40 (proline-rich Akt substrate 40) and reciprocally increased S792-phosphorylated raptor (regulatory associated protein of mTOR). Despite these phosphorylation changes, sepsis did not alter PRAS40 binding to raptor. The amount of the mTOR-raptor complex did not differ between groups. In contrast, the binding and retention of both 4E-BP1 and S6K1 to raptor were increased, and, conversely, the binding of raptor with eIF3 was decreased in sepsis. These changes in mTORC1 in the basal state were associated with enhanced 5'-AMP activated kinase activity. Acute in vivo leucine stimulation increased muscle protein synthesis in control, but not septic rats. This muscle leucine resistance was associated with coordinated changes in raptor-eIF3 binding and 4E-BP1 phosphorylation. Overall, our data suggest the sepsis-induced decrease in muscle protein synthesis may be mediated by the inability of 4E-BP1 and S6K1 to be phosphorylated and released from mTORC1 as well as the decreased recruitment of eIF3 necessary for a functional 48S complex. These data provide additional mechanistic insight into the molecular mechanisms by which sepsis impairs both basal protein synthesis and the anabolic response to the nutrient signal leucine in skeletal muscle.


Subject(s)
Leucine/pharmacology , Multiprotein Complexes/metabolism , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Protein Biosynthesis/drug effects , Sepsis/metabolism , Animals , Male , Phosphorylation , Rats , Rats, Sprague-Dawley
4.
Endocrinology ; 152(1): 151-63, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21106878

ABSTRACT

Bacterial infection decreases skeletal muscle protein synthesis via inhibition of the mammalian target of rapamycin (mTOR), a key regulator of translation initiation. To better define the mechanism by which muscle mTOR activity is decreased, we used an in vitro model of C2C12 myotubes treated with endotoxin [lipopolysaccharide (LPS)]and interferon (IFN)-γ to determine whether stable lipophilic pyruvate derivatives restore mTOR signaling. Myotubes treated with a combination of LPS and IFNγ down-regulated the phosphorylation of the mTOR substrates S6 kinase-1 and 4E binding protein-1. The phosphorylation of ribosomal protein S6 was decreased, whereas phosphorylation of elongation factor-2 was enhanced; all results consistent with defects in both translation initiation and elongation. LPS/IFNγ decreased protein synthesis 60% in myotubes. Treatment with methyl or ethyl pyruvate partially protected against the LPS/IFNγ-induced fall in mTOR signaling. The protective effect of ethyl and methyl pyruvate could not be replicated by an equimolar amount of sodium pyruvate. Although LPS/IFNγ treated myotubes were initially IGF-I responsive, prolonged exposure (≥ 17 h) resulted in IGF-I resistance at the level of mTOR despite normal IGF-I receptor phosphorylation. Ethyl pyruvate treatment restored IGF-I sensitivity as evidenced by the left shift in the IGF-I dose-response curve and maintained IGF-I responsiveness for a prolonged period of time. Ethyl pyruvate also restored IGF-I-stimulated protein synthesis in LPS/IFNγ-treated myotubes. Cotreatment with N-acetyl cysteine or ascorbic acid also preserved IGF-I sensitivity and mTOR activity. The data suggest that the combination of LPS and IFNγ inhibits mTOR activity and that prolonged exposure induces IGF-I resistance in myotubes. Lipophilic pyruvate derivatives and antioxidants show promise at rescuing mTOR activity and muscle protein synthesis by maintaining IGF-I sensitivity in this model.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Muscle Fibers, Skeletal/drug effects , Pyruvates/pharmacology , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , GTP Phosphohydrolases/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Mice , Myoblasts , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Regulatory-Associated Protein of mTOR , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors
5.
Am J Physiol Endocrinol Metab ; 298(6): E1283-94, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20388826

ABSTRACT

Sepsis and lipopolysaccharide (LPS) may decrease skeletal muscle protein synthesis by impairing mTOR (mammalian target of rapamycin) activity. The role of mTOR in regulating muscle protein synthesis was assessed in wild-type (WT) and mTOR heterozygous (+/-) mice under basal conditions and in response to LPS and/or leucine stimulation. No difference in body weight of mTOR(+/-) mice was observed compared with WT mice; whereas whole body lean body mass was reduced. Gastrocnemius weight was decreased in mTOR(+/-) mice, which was attributable in part to a reduced rate of basal protein synthesis. LPS decreased muscle protein synthesis in WT and mTOR(+/-) mice to the same extent. Reduced muscle protein synthesis in mTOR(+/-) mice under basal and LPS-stimulated conditions was associated with lower 4E-BP1 and S6K1 phosphorylation. LPS also decreased PRAS40 phosphorylation and increased phosphorylation of raptor and IRS-1 (Ser(307)) to the same extent in WT and mTOR(+/-) mice. Muscle atrogin-1 and MuRF1 mRNA content was elevated in mTOR(+/-) mice under basal conditions, implying increased ubiquitin-proteasome-mediated proteolysis, but the LPS-induced increase in these atrogenes was comparable between groups. Plasma insulin and IGF-I as well as tissue expression of TNFalpha, IL-6, or NOS2 did not differ between WT and mTOR(+/-) mice. Finally, whereas LPS impaired the ability of leucine to stimulate muscle protein synthesis and 4E-BP1 phosphorylation in WT mice, this inflammatory state rendered mTOR(+/-) mice leucine unresponsive. These data support the idea that the LPS-induced reduction in mTOR activity is relatively more important in regulating skeletal muscle mass in response to nutrient stimulation than under basal conditions.


Subject(s)
Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Leucine/metabolism , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Blotting, Northern , Blotting, Western , Body Weight/physiology , Carrier Proteins/metabolism , Cell Cycle Proteins , Chimera , Eukaryotic Initiation Factors , Insulin Receptor Substrate Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscle Proteins/genetics , Muscle Proteins/metabolism , Organ Size/physiology , Phosphoproteins/metabolism , Phosphorylation , Polymerase Chain Reaction , RNA/chemistry , RNA/genetics , Regulatory-Associated Protein of mTOR , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Specific Pathogen-Free Organisms , TOR Serine-Threonine Kinases , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
6.
J Cell Biochem ; 108(5): 1192-202, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19795384

ABSTRACT

Insulin-like growth factor-I (IGF-I) is a major anabolic hormone for skeletal muscle and a potent stimulus for protein synthesis and translation initiation. Recent studies suggest that translation can be inhibited by over expression of the mammalian target of rapamycin (mTOR) repressor REDD1. The purpose of the present study was to determine whether IGF-I alters the expression of REDD1 and whether this is associated with a concomitant change in protein synthesis in vitro. Subcutaneous injection of IGF-I or intravenous delivery of insulin for 3-4 h increased REDD1 mRNA in skeletal muscle 7-10-fold. A threefold increase in REDD1 was observed when C2C12 myotubes were treated with IGF-I. REDD1 protein continued to be expressed for up to 24 h after addition of IGF-I to cells. Withdrawal of IGF-I from myotubes lead to a rapid loss of REDD1 protein content. IGF-I-induced REDD1 mRNA and protein expression were prevented by inhibitors of transcription and translation. IGF-I had an additive effect with dexamethasone (Dex) on REDD1 protein content in myotubes. The PI3K inhibitor LY294002 blocked IGF-I but not Dex induced REDD1. IGF-I also stimulated REDD1 promoter activity. Although REDD1 protein was elevated 5-6 h after addition of IGF-I to myotubes, protein synthesis measured during this 1 h window was paradoxically greater in myotubes expressing more REDD1. In contrast to the IGF-I induced increase in REDD1 mRNA, REDD2 mRNA was decreased by IGF-I. We conclude that IGF-I stimulates REDD1 expression in skeletal muscle and myotubes but under these conditions the REDD1 response is not sufficient to repress protein synthesis.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Line , Chromones/pharmacology , DNA-Binding Proteins , Dexamethasone/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Insulin/metabolism , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Mice , Morpholines/pharmacology , Muscle Fibers, Skeletal/drug effects , Muscle, Skeletal/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Biosynthesis/drug effects , Proteins/genetics , Proteins/metabolism , Rats , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors , Transcriptional Activation/drug effects , Up-Regulation
7.
Metabolism ; 58(6): 787-97, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19375133

ABSTRACT

The present study tests the hypotheses that local bioavailability of insulin-like growth factor I (IGF-I) is capable of regulating muscle protein balance and that muscle-directed IGF-I can selectively maintain muscle mass during bacterial infection. Initial studies in C57BL/6 mice demonstrated that increasing or decreasing bioavailable IGF-I within muscle by local administration of either Leu(24) Ala(31) IGF-I or IGF binding protein 1, respectively, produced proportional changes in surrogate markers (eg, phosphorylation of 4E-BP1 and S6K1) of protein synthesis. We next examined the ability of a sustained local administration of IGF-I to prevent sepsis-induced muscle atrophy over a 5-day period. At the time of cecal ligation and puncture or sham surgery, mice had a time-release pellet containing IGF-I implanted next to the gastrocnemius and a placebo pellet placed in the contralateral limb. Data indicated that IGF-I released locally only affected the adjacent muscle and was not released into the circulation. Gastrocnemius from septic mice containing the placebo pellet was atrophied and had a reduced IGF-I protein content. In contrast, locally directed IGF-I increased IGF-I protein within adjacent muscle to basal control levels. This change was associated with a proportional increase in muscle weight and protein, as well as increased phosphorylation of 4E-BP1 and the redistribution of eIF4E from the inactive eIF4E4EBP1 complex to the active eIF4EeIF4G complex. Local IGF-I also prevented the sepsis-induced increase in atrogin-1 messenger RNA in the exposed muscle. Finally, local IGF-I prevented the sepsis-induced increase in muscle interleukin-6 messenger RNA. Thus, muscle-directed IGF-I attenuates the sepsis-induced atrophic response apparently by increasing muscle protein synthesis and potentially decreasing proteolysis. Collectively, our data suggest that agents that increase the bioavailability of IGF-I within muscle per se might be effective in ameliorating the sepsis-induced loss of muscle mass without having undesirable effects on metabolic processes in distant organs.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Muscular Atrophy/prevention & control , Sepsis/complications , Animals , Insulin-Like Growth Factor I/administration & dosage , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscular Atrophy/etiology , Protein Biosynthesis/drug effects , RNA, Messenger/drug effects
8.
Shock ; 32(4): 416-26, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19295495

ABSTRACT

The purpose of the present study was to test the hypothesis that endogenous NO negatively affects translation in skeletal muscle cells after exposure to a combination of endotoxin (LPS) and interferon-gamma (IFN-gamma). Individually, LPS and IFN-gamma did not alter protein synthesis, but in combination, they inhibited protein synthesis by 80% in C2C12 myotubes. The combination of LPS and IFN-gamma dramatically downregulated the autophosphorylation of the mammalian target of rapamycin and its substrates S6K1 and 4EBP-1. The phosphorylation of ribosomal protein S6 was decreased, whereas phosphorylation of elongation factor 2 and raptor was enhanced, consistent with defects in both translation initiation and elongation. Reduced S6 phosphorylation occurred 8 to 18 h after LPS/IFN-gamma and coincided with a prolonged upregulation of NOS2 messenger RNA and protein. NOS2 protein expression and the LPS/IFN-gamma-induced fall in phosphorylated S6 were prevented by the proteasome inhibitor MG-132. The general NOS inhibitor, L-NAME, and the specific NOS2 inhibitor, 1400W, also prevented the LPS/IFN-gamma-induced decrease in protein synthesis and restored translational signaling. LPS/IFN-gamma downregulated the phosphorylation of multiple Akt substrates, including the proline-rich Akt substrate 40, while enhancing the phosphorylation of raptor on a 5'-AMP-activated kinase (AMPK)-regulated site. The negative effects of LPS/IFN-gamma were blunted by the AMPK inhibitor compound C. The data suggest that, in combination, LPS and IFN-gamma induce a prolonged expression of NOS2 and excessive production of NO that reciprocally alter Akt and AMPK activity and consequently downregulate translation via reduced mammalian target of rapamycin signaling.


Subject(s)
Endotoxins/pharmacology , Interferon-gamma/pharmacology , Muscle Cells/drug effects , Muscle Cells/enzymology , Muscle, Skeletal/cytology , Nitric Oxide Synthase/metabolism , AMP-Activated Protein Kinases/metabolism , Amidines/pharmacology , Animals , Benzylamines/pharmacology , Blotting, Western , Carrier Proteins/metabolism , Cell Line , Cell Survival/drug effects , Enzyme Activation/drug effects , Mice , Muscle Cells/metabolism , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Oxazines/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases
9.
J Nutr ; 138(10): 1887-94, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18806097

ABSTRACT

Several stress conditions are characterized by activation of 5'-AMP-activated protein kinase (AMPK) and the development of leucine resistance in skeletal muscle. In the present study, we determined whether direct activation of the AMPK by 5-aminoimidazole-4-carboxamide-1-beta-D-ribonucleoside (AICAR) prevents the characteristic leucine-induced increase in protein synthesis by altering mammalian target of rapamycin (mTOR) signal transduction. Rats were injected with AICAR or saline (Sal) and 1 h thereafter received an oral gavage of leucine (or Sal). Efficacy of AICAR was verified by increased AMPK phosphorylation. AICAR decreased basal in vivo muscle (gastrocnemius) protein synthesis and completely prevented the leucine-induced increase, independent of a change in muscle adenine nucleotide concentration. AICAR also prevented the hyperphosphorylation of eukaryotic initiation factor (eIF) 4E binding protein (4E-BP1), ribosomal protein S6 kinase (S6K1), S6, and eIF4G in response to leucine, suggesting a decrease in mTOR activity. Moreover, AICAR prevented the leucine-induced redistribution of eIF4E from the inactive eIF4E.4E-BP1 to the active eIF4E.eIF4G complex. This ability of AICAR to produce muscle leucine resistance could not be attributed to a change in phosphorylation of tuberous sclerosis complex (TSC)2, the formation of a TSC1.TSC2 complex, the binding of raptor with mTOR, or the phosphorylation of eukaryotic elongation factor-2. However, the inhibitory actions of AICAR were associated with reduced phosphorylation of proline-rich Akt substrate-40 and increased phosphorylation of raptor, which represent potential mechanisms by which AICAR might be expected to inhibit leucine-induced increases in mTOR activity and protein synthesis under in vivo conditions.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Leucine/pharmacology , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Ribonucleosides/pharmacology , Adenine Nucleotides/metabolism , Amino Acids, Branched-Chain/blood , Aminoimidazole Carboxamide/administration & dosage , Aminoimidazole Carboxamide/pharmacology , Animals , Enzyme Activation , Injections, Subcutaneous , Insulin/blood , Leucine/blood , Male , Muscle Proteins/antagonists & inhibitors , Muscle, Skeletal/drug effects , Rats , Rats, Sprague-Dawley , Ribonucleosides/administration & dosage
10.
Am J Physiol Endocrinol Metab ; 295(4): E964-73, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18713959

ABSTRACT

The mechanism by which human immunodeficiency virus (HIV)-1 infection in humans leads to the erosion of lean body mass is poorly defined. Therefore, the purpose of the present study was to determine whether transgenic (Tg) rats that constitutively overexpress HIV-1 viral proteins exhibit muscle wasting and to elucidate putative mechanisms. Over 7 mo, Tg rats gained less body weight than pair-fed controls exclusively as a result of a proportional reduction in lean, not fat, mass. Fast- and slow-twitch muscle atrophy in Tg rats did not result from a reduction in the in vivo-determined rate of protein synthesis. In contrast, urinary excretion of 3-methylhistidine, as well as the content of atrogin-1 and the 14-kDa actin fragment, was elevated in gastrocnemius of Tg rats, suggesting increased muscle proteolysis. Similarly, Tg rats had reduced cardiac mass, which was independent of a change in protein synthesis. This decreased cardiac mass was associated with a reduction in stroke volume, but cardiac output was maintained by a compensatory increase in heart rate. The HIV-induced muscle atrophy was associated with increased whole body energy expenditure, which was not due to an elevated body temperature or secondary bacterial infection. Furthermore, the atrophic response could not be attributed to the development of insulin resistance, decreased levels of circulating amino acids, or increased tissue cytokines. However, skeletal muscle and, to a lesser extent, circulating insulin-like growth factor I was reduced in Tg rats. Although hepatic injury was implicated by increased plasma levels of aspartate and alanine aminotransferases, hepatic protein synthesis was not different between control and Tg rats. Hence, HIV-1 Tg rats develop atrophy of cardiac and skeletal muscle, the latter of which results primarily from an increased protein degradation and may be related to the marked reduction in muscle insulin-like growth factor I.


Subject(s)
HIV Wasting Syndrome/genetics , HIV Wasting Syndrome/pathology , HIV-1/genetics , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Myocytes, Cardiac/pathology , Amino Acids/blood , Animals , Animals, Genetically Modified , Atrophy/pathology , Blotting, Northern , Body Composition/physiology , Body Temperature/physiology , Body Weight/physiology , Calorimetry, Indirect , Cytokines/metabolism , Energy Metabolism/physiology , Human Immunodeficiency Virus Proteins/biosynthesis , Human Immunodeficiency Virus Proteins/genetics , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Kidney/physiopathology , Male , Muscle Proteins/biosynthesis , Muscular Diseases/pathology , Nuclease Protection Assays , Organ Size/physiology , Rats , Rats, Inbred F344
11.
Am J Physiol Regul Integr Comp Physiol ; 294(6): R1777-89, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18401005

ABSTRACT

Acute alcohol intoxication decreases muscle protein synthesis, but there is a paucity of data on the ability of alcohol to regulate muscle protein degradation. Furthermore, various types of atrophic stimuli appear to regulate ubiquitin-proteasome-dependent proteolysis by increasing the muscle-specific E3 ligases atrogin-1 and MuRF1 (i.e., "atrogenes"). Therefore, the present study was designed to test the hypothesis that acute alcohol intoxication increases atrogene expression leading to an elevated rate of muscle protein breakdown. In male rats, the intraperitoneal injection of alcohol dose- and time-dependently increased atrogin-1 and MuRF1 mRNA in gastrocnemius, the latter of which was most pronounced. A comparable change was absent in the soleus and heart. The ability of in vivo-administered ethanol to increase atrogene expression was independent of the route of alcohol administration (intraperitoneal vs. oral), as well as of nutritional status (fed vs. fasted) and gender (male vs. female). The increase in atrogin-1 and MuRF1 was independent of alcohol metabolism, and the overproduction of endogenous glucocorticoids and could not be prevented by maintaining the circulating concentration of insulin-like growth factor-I. Despite marked changes in atrogene expression, acute alcohol in vivo did not alter the release of either 3-methylhistidine (MH) or tyrosine from the isolated perfused hindlimb, suggesting that the rate of muscle proteolysis remains unchanged. Moreover, alcohol did not increase the directly determined rate of protein degradation in isolated epitrochlearis muscles or cultured myocytes. Finally, no increase in atrogene expression or 3-MH release was detected in muscle from rats fed an alcohol-containing diet. Our results indicate that although acute alcohol intoxication increases atrogin-1 and MuRF1 mRNA preferentially in fast-twitch skeletal muscle, this change was not associated with increased rates of muscle proteolysis. Therefore, the loss of muscle mass/protein in response to chronic alcohol abuse appears to result primarily from a decrement in muscle protein synthesis, not an increase in degradation.


Subject(s)
Alcoholic Intoxication/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , RNA, Messenger/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin-Protein Ligases/metabolism , Alcohol Drinking/metabolism , Animals , Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Glucocorticoids/metabolism , Insulin-Like Growth Factor I/metabolism , Male , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Fast-Twitch/metabolism , Muscle, Skeletal/drug effects , Polyubiquitin/metabolism , Protein Denaturation/drug effects , Rats , Rats, Sprague-Dawley , Tripartite Motif Proteins
12.
Alcohol Clin Exp Res ; 32(5): 796-805, 2008 May.
Article in English | MEDLINE | ID: mdl-18336631

ABSTRACT

BACKGROUND: The mechanism by which acute alcohol (EtOH) intoxication decreases basal muscle protein synthesis via inhibition of the Ser/Thr kinase mammalian target of rapamycin (mTOR) is poorly defined. In this regard, mTOR activity is impaired after over expression of the regulatory protein REDD1. Hence, the present study assessed the ability of REDD1 as a potential mediator of the EtOH-induced decrease in muscle protein synthesis. METHODS: The effect of acute EtOH intoxication on REDD1 mRNA and protein was determined in striated muscle of rats and mouse myocytes using an RNase protection assay and Western blotting, respectively. Other components of the mTOR signaling pathway were also assessed by immunoblotting. For comparison, REDD1 mRNA/protein was also determined in the muscle of rats chronically fed an alcohol-containing diet for 14 weeks. RESULTS: Intraperitoneal (IP) injection of EtOH increased gastrocnemius REDD1 mRNA in a dose- and time-dependent manner, and these changes were associated with reciprocal decreases in the phosphorylation of 4E-BP1, which is a surrogate marker for mTOR activity and protein synthesis. No change in REDD1 mRNA was detected in the slow-twitch soleus muscle or heart. Acute EtOH produced comparable increases in muscle REDD1 protein. The EtOH-induced increase in gastrocnemius REDD1 was independent of the route of EtOH administration (oral vs. IP), the nutritional state (fed vs. fasted), gender, and age of the rat. The nonmetabolizable alcohol tert-butanol increased REDD1 and the EtOH-induced increase in REDD1 was not prevented by pretreatment with the alcohol dehydrogenase inhibitor 4-methylpyrazole. In contrast, REDD1 mRNA and protein were not increased in the isolated hindlimb perfused with EtOH or in C2C12 myocytes incubated with EtOH, under conditions previously reported to decrease protein synthesis. Pretreatment with the glucocorticoid receptor antagonist RU486 failed to prevent the EtOH-induced increase in REDD1. Finally, the EtOH-induced increase in REDD1 was not associated with altered formation of the TSC1*TSC2 complex or the phosphorylation of TSC2 which is down stream in the REDD1 stress response pathway. In contradistinction to the changes observed with acute EtOH intoxication, REDD1 mRNA/protein was not changed in gastrocnemius from chronic alcohol-fed rats despite the reduction in 4E-BP1 phosphorylation. CONCLUSIONS: These data indicate that in fast-twitch skeletal muscle (i) REDD1 mRNA/protein is increased in vivo by acute EtOH intoxication but not in response to chronic alcohol feeding, (ii) elevated REDD1 in response to acute EtOH appears due to the production of an unknown secondary mediator which is not corticosterone, and (iii) the EtOH-induced decrease in protein synthesis can be dissociated from a change in REDD1 suggesting that the induction of this protein is not responsible for the rapid decrease in protein synthesis after acute EtOH administration or for the development of alcoholic myopathy in rats fed an alcohol-containing diet.


Subject(s)
Alcoholic Intoxication/metabolism , DNA-Binding Proteins/metabolism , Muscle Fibers, Fast-Twitch/metabolism , Muscle, Skeletal/metabolism , Protein Kinases/metabolism , Repressor Proteins/metabolism , Alcohol Drinking/metabolism , Animals , Carrier Proteins/metabolism , Cell Line , Female , Intracellular Signaling Peptides and Proteins , Male , Mice , Myoblasts, Skeletal , Phosphoproteins/metabolism , Phosphorylation , RNA, Messenger/analysis , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Signal Transduction/physiology , TOR Serine-Threonine Kinases , Transcription Factors/metabolism
13.
J Trauma ; 64(2): 477-86, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18301218

ABSTRACT

BACKGROUND: Sepsis stimulates the sympathetic nervous system. The resultant elevation in plasma catecholamines, both norepinephrine and epinephrine (Epi), might be expected to alter the expression of inflammatory cytokines, which may directly or indirectly influence muscle protein balance. The purpose of this study was twofold: (1) determine whether Epi per se increases cytokine expression in skeletal muscle, and (2) determine whether beta-adrenergic blockade alters the sepsis-induced expression of inflammatory cytokines and mediators of protein balance in skeletal muscle. METHODS: In the first study, rats were infused with Epi for 2 hour to increase the circulating Epi concentration to levels seen in septic animals. In the second study, sepsis was induced by cecal ligation and puncture and a nonspecific beta-adrenergic blockade produced with a continuous infusion of propranolol (PP). Tissues were obtained 24 after induction of sepsis and analyzed for tumor necrosis factor (TNF)-alpha interleukin (IL)-1beta, IL-6 mRNA and protein content. In addition, the tissue content of insulin-like growth factor (IGF)-I and various regulators of protein synthesis were assessed. RESULTS: Epi acutely increased TNF-alpha IL-6 and IL-1beta mRNA content in muscle (3- to 40-fold). However, only the TNF-alpha and IL-6 protein content was increased in muscle by Epi. In the second study, beta-adrenergic blockade with PP exacerbated the sepsis-induced increase in muscle IL-6 and TNF-alpha mRNA but did not alter the increment in IL-1beta or HMGB1. Propranolol also accentuated the sepsis-induced increase in both IL-6 and TNF-alpha protein in muscle. The exaggerated muscle cytokine response in septic rats treated with PP was associated with a reduction in muscle IGF-I protein that was greater than detected in saline-infused septic rats. Finally, the combination of sepsis + PP also accentuated the sepsis-induced decrease in the phosphorylation of 4E-binding protein-1, ribosomal protein S6, and mTOR, which are key proteins controlling protein synthesis. CONCLUSIONS: These results demonstrate that although Epi is capable of increasing tissue cytokines in naive rats, inhibition of the beta-adrenergic effects of catecholamines exacerbates the sepsis-induced increase of selected inflammatory cytokines. This exaggerated tissue response is associated with alterations in muscle IGF-I protein and translation initiation, which would be expected to impair tissue protein synthesis.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Cytokines/biosynthesis , Epinephrine/pharmacology , Muscle, Skeletal/metabolism , Protein Biosynthesis/drug effects , Sepsis/metabolism , Animals , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Male , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/biosynthesis
14.
Am J Physiol Endocrinol Metab ; 293(2): E453-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17505052

ABSTRACT

Prolonged sepsis and exposure to an inflammatory milieu decreases muscle protein synthesis and reduces muscle mass. As a result of its ability to integrate diverse signals, including hormones and nutrients, the mammalian target of rapamycin (mTOR) is a dominant regulator in the translational control of protein synthesis. Under postabsorptive conditions, sepsis decreases mTOR kinase activity in muscle, as evidenced by reduced phosphorylation of both eukaryotic initiation factor (eIF)4E-binding protein (BP)-1 and ribosomal S6 kinase (S6K)1. These sepsis-induced changes, along with the redistribution of eIF4E from the active eIF4E.eIF4G complex to the inactive eIF4E.4E-BP1 complex, are preventable by neutralization of tumor necrosis factor (TNF)-alpha but not by antagonizing glucocorticoid action. Although the ability of mTOR to respond to insulin-like growth factor (IGF)-I is not disrupted by sepsis, the ability of leucine to increase 4E-BP1 and S6K1 phosphorylation is greatly attenuated. This "leucine resistance" results from a cooperative interaction between both TNF-alpha and glucocorticoids. Finally, although septic animals are not IGF-I resistant, the anabolic actions of IGF-I are nonetheless reduced because of the development of growth hormone resistance, which decreases both circulating and muscle IGF-I. Herein, we highlight recent advances in the mTOR signaling network and emphasize their connection to the atrophic response observed in skeletal muscle during sepsis. Although many unanswered questions remain, understanding the cellular basis of the sepsis-induced decrease in translational activity will contribute to the rational development of therapeutic interventions and thereby minimize the debilitating affects of the atrophic response that impairs patient recovery.


Subject(s)
Gene Expression Regulation , Inflammation/genetics , Muscle Proteins/genetics , Protein Biosynthesis/physiology , Sepsis/genetics , Amino Acids/physiology , Animals , Humans , Inflammation/metabolism , Metabolic Networks and Pathways/physiology , Models, Biological , Muscle Proteins/metabolism , Nutritional Physiological Phenomena , Peptide Initiation Factors/physiology , Protein Kinases/physiology , RNA Cap-Binding Proteins/metabolism , RNA Cap-Binding Proteins/physiology , Ribosomal Protein S6/metabolism , Sepsis/metabolism , TOR Serine-Threonine Kinases
15.
J Appl Physiol (1985) ; 103(1): 378-87, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17332274

ABSTRACT

Although the boundaries of skeletal muscle size are fundamentally determined by genetics, this dynamic tissue also demonstrates great plasticity in response to environmental and hormonal factors. Recent work indicates that contractile activity, nutrients, growth factors, and cytokines all contribute to determining muscle mass. Muscle responds not only to endocrine hormones but also to the autocrine production of growth factors and cytokines. Skeletal muscle synthesizes anabolic growth factors such as insulin-like growth factor (IGF)-I and potentially inhibitory cytokines such as interleukin (IL)-6, tumor necrosis factor (TNF)-alpha, and myostatin. These self-regulating inputs in turn influence muscle metabolism, including the use of nutrients such as glucose and amino acids. These changes are principally achieved by altering the activity of the protein kinase known as protein kinase B or Akt. Akt plays a central role in integrating anabolic and catabolic responses by transducing growth factor and cytokine signals via changes in the phosphorylation of its numerous substrates. Activation of Akt stimulates muscle hypertrophy and antagonizes the loss of muscle protein. Here we review the many signals that funnel through Akt to alter muscle mass.


Subject(s)
Cytokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Muscle Contraction , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Adenosine Triphosphate/metabolism , Animals , Autocrine Communication , Cell Proliferation , Cell Size , Humans , Hypertrophy , Insulin-Like Growth Factor I/metabolism , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscular Atrophy/enzymology , Muscular Atrophy/pathology , Myostatin , NF-kappa B/metabolism , Paracrine Communication , Phosphatidylinositol 3-Kinases/metabolism , Transforming Growth Factor beta/metabolism
16.
Am J Physiol Endocrinol Metab ; 292(6): E1555-67, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17264220

ABSTRACT

The hypothesis of the present study was that exposure of differentiated muscle cells to agonists of the AMP-activated protein kinase (AMPK) would increase the mRNA content of the muscle-specific ubiquitin ligases muscle atrophy F-box (MAFbx) and muscle RING finger 1 (MuRF1). C(2)C(12) cells were incubated with incremental doses of 5-aminoimidazol-4-carboximide ribonucleoside (AICAR) or metformin for 24 h. Both MAFbx and MuRF1 mRNA increased dose dependently in response to these AMPK activators. AICAR, metformin, and 2-deoxy-d-glucose produced time-dependent alterations in ubiquitin ligase expression, typified by a biphasic pattern of expression marked by an acute repression followed by a sustained induction. AMPK-activating treatments in conjunction with dexamethasone produced a pronounced synergistic effect on ligase mRNA expression at later time points. This cooperative response occurred in the absence of a dexamethasone-dependent increase in AMPK expression or activity, as determined by immunoblotting for phosphorylation and expression of AMPKalpha and its downstream target acetyl-CoA carboxylase (ACC). These responses elicited by AMPK activation singly or in combination with dexamethasone did not extend to the mRNA expression of the UBR box family E3s UBR1/E3alphaI and UBR2/E3alphaII. Treatment with the AMPK inhibitor compound C prevented increases in MAFbx and MuRF1 mRNA in response to serum deprivation, as well as AICAR and dexamethasone treatment individually or jointly. Stimulation of AMPK activity in vivo via AICAR injection increased both MAFbx and MuRF1 mRNA in murine skeletal muscle. These data suggest that activation of AMPK in skeletal muscle results in a specific upregulation of MAFbx and MuRF1, responses that are reminiscent of the proposed atrophic transcriptional program executed under various conditions of skeletal muscle wasting. Therefore, AMPK may be a critical component of the intercalated network of signaling pathways governing skeletal muscle atrophy, where its input acts to modify anti- and proatrophic signals to influence gene expression in reaction to catabolic perturbations.


Subject(s)
Enzyme Activators/pharmacology , Multienzyme Complexes/metabolism , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , SKP Cullin F-Box Protein Ligases/genetics , Ubiquitin-Protein Ligases/genetics , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/administration & dosage , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cell Line , Deoxyglucose/pharmacology , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Energy Metabolism/physiology , Glucocorticoids/pharmacology , Homeostasis/physiology , Metformin/administration & dosage , Metformin/pharmacology , Multienzyme Complexes/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Ribonucleotides/administration & dosage , Ribonucleotides/pharmacology , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/metabolism
17.
Am J Physiol Endocrinol Metab ; 292(6): E1497-506, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17264221

ABSTRACT

Epidemiological evidence suggests alcoholic myopathy is more severe in females than males, but comparable animal studies are lacking that make elucidating the biochemical locus for this defect problematic. The present study determined whether skeletal muscle protein synthesis and markers of degradation exhibit a sexual dimorphic response to either chronic alcohol consumption or acute intoxication. Male and female rats were fed an alcohol-containing diet, pair-fed for 26 wk (chronic), or received an intraperitoneal injection of alcohol (acute). In males, chronic alcohol decreased gastrocnemius protein synthesis by 20%. This reduction was associated with a twofold increase in the inactive eukaryotic initiation factor (eIF) 4E.4E-binding protein 1 (4E-BP1) complex and a 60% reduction in the active eIF4E.eIF4G complex. This redistribution of eIF4E was associated with decreased phosphorylation of both 4E-BP1 and eIF4G (50-55%). The phosphorylation of ribosomal protein S6 was also reduced 60% in alcohol-consuming male rats. In contrast, neither rates of protein synthesis nor indexes of translation initiation in muscle were altered in alcohol-fed female rats despite blood alcohol levels comparable to males. Chronic alcohol ingestion did not alter atrogin-1 or muscle RING finger-1 mRNA content (biomarkers of muscle proteolysis) in males but increased their expression in females 50-100%. Acute alcohol intoxication produced a comparable decrease in muscle protein synthesis and translation initiation in both male and female rats. Our data demonstrate a sexual dimorphism for muscle protein synthesis, translation initiation, and proteolysis in response to chronic, but not acute, alcohol intoxication; however, they do not support evidence indicating females are more sensitive toward the development of alcoholic skeletal muscle myopathy.


Subject(s)
Alcohol Drinking/metabolism , Alcoholic Intoxication/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Sex Characteristics , Acute Disease , Alcohol Drinking/blood , Alcoholic Intoxication/blood , Animals , Carrier Proteins/metabolism , Ethanol/blood , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4F/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Female , Hormones/blood , Insulin-Like Growth Factor I/metabolism , Intracellular Signaling Peptides and Proteins , Male , Muscle Proteins/biosynthesis , Peptide Hydrolases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Biosynthesis , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6/metabolism , Time Factors
18.
Am J Physiol Regul Integr Comp Physiol ; 292(1): R328-36, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16946078

ABSTRACT

The present study determined whether thermal injury increases the expression of the ubiquitin (Ub) E3 ligases referred to as muscle ring finger (MuRF)-1 and muscle atrophy F-box (MAFbx; aka atrogin-1), which are muscle specific and responsible for the increased protein breakdown observed in other catabolic conditions. After 48 h of burn injury (40% total body surface area full-thickness scald burn) gastrocnemius weight was reduced, and this change was associated with an increased mRNA abundance for atrogin-1 and MuRF-1 (3.1- to 8-fold, respectively). Similarly, burn increased polyUb mRNA content in the gastrocnemius twofold. In contrast, there was no burn-induced atrophy of the soleus and no significant change in atrogin-1, MuRF-1, or polyUb mRNA. Burns also did not alter E3 ligase expression in heart. Four hours after administration of the anabolic agent insulin-like growth factor (IGF)-I to burned rats, the mRNA content of atrogin-1 and polyUb in gastrocnemius had returned to control values and the elevation in MuRF-1 was reduced 50%. In contrast, leucine did not alter E3 ligase expression. In a separate study, in vivo administration of the proteasome inhibitor Velcade prevented burn-induced loss of muscle mass determined at 48 h. Finally, administration of the glucocorticoid receptor antagonist RU-486 did not prevent burn-induced atrophy of the gastrocnemius or the associated elevation in atrogin-1, MuRF-1, or polyUb. In summary, the acute muscle wasting accompanying thermal injury is associated with a glucocorticoid-independent increase in the expression of several Ub E3 ligases that can be downregulated by IGF-I.


Subject(s)
Burns/metabolism , Glucocorticoids/physiology , Insulin-Like Growth Factor I/pharmacology , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , SKP Cullin F-Box Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Animals , Blotting, Northern , Boronic Acids/pharmacology , Bortezomib , Burns/complications , Down-Regulation/drug effects , Glucocorticoids/antagonists & inhibitors , Hormone Antagonists/pharmacology , Immunoblotting , Leucine/pharmacology , Male , Mifepristone/pharmacology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Myocardium/metabolism , Organ Size/drug effects , Polyubiquitin/metabolism , Protein Synthesis Inhibitors/pharmacology , Pyrazines/pharmacology , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , SKP Cullin F-Box Protein Ligases/genetics , Tripartite Motif Proteins , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Wasting Syndrome/etiology , Wasting Syndrome/metabolism
19.
Am J Physiol Endocrinol Metab ; 292(2): E501-12, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17003238

ABSTRACT

Various atrophic stimuli increase two muscle-specific E3 ligases, muscle RING finger 1 (MuRF1) and atrogin-1, and knockout mice for these "atrogenes" display resistance to denervation-induced atrophy. The present study determined whether increased atrogin-1 and MuRF1 mRNA are mediated by overproduction of endogenous glucocorticoids or inflammatory cytokines in adult rats and whether atrogene expression can be downregulated by anabolic agents such as insulin-like growth factor (IGF)-I and the nutrient-signaling amino acid leucine. Both atrogin-1 and MuRF1 mRNA in gastrocnemius was upregulated dose and time dependently by endotoxin. Additionally, peritonitis produced by cecal ligation and puncture increased atrogin-1 and MuRF1 mRNA in gastrocnemius (but not soleus or heart) by 8 h, which was sustained for 72 and 24 h, respectively. Whereas the sepsis-induced increase in atrogin-1 expression was completely prevented by IGF-I, the increased MuRF1 was not altered. In contrast to the IGF-I effect, the sepsis-induced increased mRNA of both atrogenes was unresponsive to either acute or repetitive administration of leucine. Whereas exogenous infusion of TNF-alpha increased atrogin-1 and MuRF1 in gastrocnemius, pretreatment of septic rats with the TNF antagonist TNF-binding protein did not prevent increased expression of either atrogene. Similarly, whereas dexamethasone increased atrogene expression, pretreatment with the glucocorticoid receptor antagonist RU-486 failed to ameliorate the sepsis-induced increase in atrogin-1 and MuRF1. Thus, under in vivo conditions in mature adult rats, the sepsis-induced increase in muscle atrogin-1 and MuRF1 mRNA appears both glucocorticoid and TNF independent and is unresponsive to leucine.


Subject(s)
Cytokines/pharmacology , Diet , Glucocorticoids/pharmacology , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , SKP Cullin F-Box Protein Ligases/metabolism , Sepsis/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Corticosterone/metabolism , Insulin/metabolism , Male , Muscle, Skeletal/metabolism , Rats , Rats, Sprague-Dawley , Tripartite Motif Proteins , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/physiology
20.
Metabolism ; 56(1): 49-57, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17161226

ABSTRACT

Inhibition of translational efficiency is responsible at least in part for the sepsis-induced decrease in protein synthesis observed in skeletal muscle. Moreover, infusion of the inflammatory cytokine tumor necrosis factor alpha (TNF-alpha) into naive rats produces a comparable decrement. Therefore, the purpose of the present study was to determine whether inhibition of TNF action under in vivo conditions could prevent the sepsis-induced decrease in translation initiation observed in the postabsorptive state. To address this aim, sepsis was produced by cecal ligation and puncture (CLP) and rats were studied in the fasted condition 20 to 24 hours thereafter. Both septic and time-matched nonseptic control rats were pretreated with TNF-binding protein (TNF(BP)) before CLP or sham surgery to neutralize endogenously produced TNF. Sepsis altered the distribution of eukaryotic initiation factor 4E (eIF4E) in the gastrocnemius by increasing the amount associated with 4E-BP1 (inactive complex) and decreasing the amount bound to eIF4G (active complex). This change in eIF4E availability was associated with a decreased phosphorylation of 4E-BP1. Furthermore, the phosphorylation of ribosomal protein S6 and mammalian target of rapamycin (mTOR) was also decreased in the gastrocnemius from septic rats. Pretreatment of septic rats with TNF(BP) largely ameliorated the altered distribution of eIF4E as well as the reduced phosphorylation of 4E-BP1, S6, and mTOR. In contrast, sepsis did not change either the total amount or the phosphorylation state of eIF2alpha or eIF2Bepsilon. Furthermore, no sepsis-induced change in eIFs was detected in the slow-twitch soleus muscle. The ability of TNF(BP) to prevent the sepsis-induced alterations in translation initiation was independent of change in plasma insulin and proportional to the insulinlike growth factor I content in blood and muscle but was associated with a reduction in plasma corticosterone. Hence, the decreased constitutive protein synthesis observed in fast-twitch skeletal muscle in response to peritonitis is mediated by a TNF-dependent mechanism affecting mTOR regulation of translation initiation.


Subject(s)
Muscle Proteins/metabolism , Protein Biosynthesis/physiology , Sepsis/metabolism , Tumor Necrosis Factor-alpha/physiology , Animals , Carrier Proteins/metabolism , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2B/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Immunoblotting , Insulin/blood , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Intracellular Signaling Peptides and Proteins , Male , Muscle, Skeletal/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Kinases/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor/administration & dosage , Receptors, Tumor Necrosis Factor/metabolism , Ribosomal Protein S6/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...