151
|
Reid MB. Response of the ubiquitin-proteasome pathway to changes in muscle activity. Am J Physiol Regul Integr Comp Physiol 2005; 288:R1423-31. [PMID: 15886351 DOI: 10.1152/ajpregu.00545.2004] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The ubiquitin-proteasome pathway plays a critical role in the adaptation of skeletal muscle to persistent decreases or increases in muscle activity. This article outlines the basics of pathway function and reviews what we know about pathway responses to altered muscle use. The ubiquitin-proteasome pathway regulates proteolysis in mammalian cells by attaching ubiquitin polymers to damaged proteins; this targets the protein for degradation via the 26S proteasome. The pathway is constitutively active in muscle and continually regulates protein turnover. Conditions of decreased muscle use, e.g., unloading, denervation, or immobilization, stimulate general pathway activity. This activity increase is caused by upregulation of regulatory components in the pathway and leads to accelerated proteolysis, resulting in net loss of muscle protein. Pathway activity is also increased in response to exercise, a two-phase response. An immediate increase in selective ubiquitin conjugation by constitutive pathway components contributes to exercise-stimulated signal transduction. Over hours-to-days, exercise also stimulates a delayed increase in general ubiquitin conjugating activity by inducing expression of key components in the pathway. This increase mediates a late-phase rise in protein degradation that is required for muscle adaptation to exercise. Thus the ubiquitin-proteasome pathway functions as an essential mediator of muscle remodeling, both in atrophic states and exercise training.
Collapse
Affiliation(s)
- Michael B Reid
- Department of Physiology, University of Kentucky, 800 Rose St., Rm. MS-509, Lexington, KY 40536-0298, USA.
| |
Collapse
|
152
|
Abstract
PURPOSE OF REVIEW Although a variety of diverse stimuli induce muscle atrophy, there is a surprising number of similarities in the intracellular responses. One prominent response is an increase in muscle proteolysis resulting from stimulation of the ubiquitin-proteasome pathway. Understanding the intracellular signaling pathways that regulate muscle mass should offer insights into the coordination of cellular responses. This review will discuss recent findings on the molecular signaling pathways regulating proteolysis during muscle atrophy. RECENT FINDINGS The expression of several muscle-specific E3 ubiquitin ligases is consistently increased in conditions causing muscle atrophy. Insulin and insulin-like growth factor-1 act through the phosphoinositide 3-kinase/AKT pathway to suppress the expression of two of these enzymes, MuRF1 and MAFbx/atrogin-1. Efforts to identify targets of the muscle-specific E3 ligases are yielding interesting information. Insulin and insulin-like growth factor-1 also attenuate wasting by inhibiting caspase-3, which cleaves actin to facilitate its destruction by the ubiqutin-proteasome system. Other signaling systems involved in the regulation of muscle mass include the nuclear factor kappa B pathway. SUMMARY The maintenance of muscle mass requires a delicate balance between catabolic factors and anabolic factors. These signals inversely modulate the activity of several key regulatory pathways including the phosphoinositide-3 kinase/AKT and nuclear factor kappa B systems, which control the transcription of components of the ubiquitin-proteasome proteolytic pathway activity, the activity of caspase-3, and perhaps other proteolytic functions. When levels of insulin or insulin-like growth factor-1 are insufficient or inflammatory cytokine production is increased, muscle atrophy ensues.
Collapse
|
153
|
Combaret L, Adegoke OAJ, Bedard N, Baracos V, Attaix D, Wing SS. USP19 is a ubiquitin-specific protease regulated in rat skeletal muscle during catabolic states. Am J Physiol Endocrinol Metab 2005; 288:E693-700. [PMID: 15562254 DOI: 10.1152/ajpendo.00281.2004] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ubiquitin-dependent proteolysis is activated in skeletal muscle atrophying in response to various catabolic stimuli. Previous studies have demonstrated activation of ubiquitin conjugation. Because ubiquitination can also be regulated by deubiquitinating enzymes, we used degenerate oligonucleotides derived from conserved sequences in the ubiquitin-specific protease (UBP) family of deubiquitinating enzymes in RT-PCR with skeletal muscle RNA to amplify putative deubiquitinating enzymes. We identified USP19, a 150-kDa deubiquitinating enzyme that is widely expressed in various tissues including skeletal muscle. Expression of USP19 mRNA increased by approximately 30-200% in rat skeletal muscle atrophying in response to fasting, streptozotocin-induced diabetes, dexamethasone treatment, and cancer. Increased mRNA levels during fasting returned to normal with refeeding, but 1 day later than the normalization of rates of proteolysis and coincided instead with recovery of muscle mass. Indeed, in all catabolic treatments, USP19 mRNA was inversely correlated with muscle mass and provided an index of muscle mass that may be useful in many pathological conditions, using small human muscle biopsies. The increased expression of this deubiquitinating enzyme under conditions of increased proteolysis suggests that it may play a role in regeneration of free ubiquitin either coincident with or after proteasome-mediated degradation of substrates. USP19 may also be involved in posttranslational processing of polyubiquitin produced de novo in response to induction of the polyubiquitin genes seen under these conditions. Deubiquitinating enzymes thus appear involved in muscle wasting and implicate a widening web of regulation of genes in the ubiquitin system in this process.
Collapse
Affiliation(s)
- Lydie Combaret
- Polypeptide Laboratory, Department of Medicine, McGill University, 3640 University St., Montreal, Quebec H3A 2B2, Canada
| | | | | | | | | | | |
Collapse
|
154
|
Stevenson EJ, Koncarevic A, Giresi PG, Jackman RW, Kandarian SC. Transcriptional profile of a myotube starvation model of atrophy. J Appl Physiol (1985) 2005; 98:1396-406. [PMID: 15608089 DOI: 10.1152/japplphysiol.01055.2004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Skeletal muscle wasting is a pervasive phenomenon that can result from a wide range of pathological conditions as well as from habitual muscular inactivity. The present work describes a cell-culture condition that induces significant atrophy in skeletal muscle C2C12 myotubes. The failure to replenish differentiation media in mature myotubes leads to rapid atrophy (53% in diameter), which is referred to here as starvation. Affymetrix microarrays were used to develop a transcriptional profile of control (fed) vs. atrophied (nonfed) myotubes. Myotube starvation was characterized by an upregulation of genes involved in translational inhibition, amino acid biosynthesis and transport, and cell cycle arrest/apoptosis, among others. Downregulated genes included several structural and regulatory elements of the extracellular matrix as well as several elements of Wnt/frizzled and TGF-β signaling pathways. Interestingly, the characteristic transcriptional upregulation of the ubiquitin-proteasome system, calpains, and cathepsins known to occur in multiple in vivo models of atrophy were not seen during myotube starvation. With the exception of the downregulation of extracellular matrix genes, serine protease inhibitor genes, and the upregulation of the translation initiation factor PHAS-I, this model of atrophy in cell culture has a transcriptional profile quite distinct from any study published to date with atrophy in whole muscle. These data show that, although the gross morphology of atrophied muscle fibers may be similar in whole muscle vs. myotube culture, the processes by which this phenotype is achieved differ markedly.
Collapse
Affiliation(s)
- Eric J Stevenson
- Dept. of Health Sciences, Boston Univ., 635 Commonwealth Ave., Boston, MA 02215, USA
| | | | | | | | | |
Collapse
|
155
|
Wang J, Laschinger C, Zhao XH, Mak B, Seth A, McCulloch CA. Mechanical force activates eIF-2α phospho-kinases in fibroblast. Biochem Biophys Res Commun 2005; 330:123-30. [PMID: 15781241 DOI: 10.1016/j.bbrc.2005.02.140] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2005] [Indexed: 10/25/2022]
Abstract
Mechanical forces can induce differentiation of fibroblasts into myofibroblasts, a process which requires activation of the MAP kinase p38. Currently, the identification of other phospho-kinases involved in myofibroblast differentiation has not been explored. We applied static tensile forces to rat cardiac fibroblasts via collagen-coated magnetite beads and examined activation of protein phospho-kinases by the Kinexus phospho-antibody screening system. Of 75 candidate protein kinases screened, 39 were detected and, of these, 31 phospho-kinases were analyzed. Following force application, 12 out of 31 phospho-kinases exhibited increases of phosphorylation including PKR (>4-fold), MKK3 (3-fold), MKK6 ( approximately 2-fold), and p38 ( approximately 2-fold). In several types of mechanically sensitive, contractile fibroblasts including rat cardiac, human gingival, and Rat-2 fibroblasts, tensile forces increased eIF-2alpha phosphorylation, a downstream effector of PKR. We conclude that phospho-antibody screening is an efficient method for discovery of novel mechanical force-induced phospho-kinases and force can activate eIF-2alpha phospho-kinases in fibroblasts.
Collapse
Affiliation(s)
- J Wang
- CIHR Group in Matrix Dynamics, Faculty of Dentistry, University of Toronto, Toronto, Ont., Canada
| | | | | | | | | | | |
Collapse
|
156
|
Abstract
Inflammation occurs in response to tissue injury or the presence of foreign antigens and is important in the mobilization of specific immunologic and nonimmunologic defenses against injury. The vascular endothelium is altered to allow immune competent cells to access the interstitial space. Muscle and visceral proteins are catabolized and the amino acids are used either to supply energy or as substrates for the production of acute-phase proteins that play a role in defense. Restoration of muscle mass is impaired while inflammation is on going. Lipids are mobilized. Although serving a vital role in allowing host survival from acute injury or infection, if unimpeded, or if triggered inappropriately, the acute-phase response may instead lead to increased vascular injury and progressive loss of muscle and visceral protein pools causing malnutrition. Markers of inflammation (C reactive protein [CRP] or interleukin-6 [IL-6] levels) are associated with cardiovascular risk in the general population and in dialysis patients. Hypoalbuminemia also is associated with cardiovascular risk in dialysis patients. Although albumin is considered a marker of nutrition, changes in albumin levels are associated with increased levels of acute-phase proteins. Persistent changes in albumin levels are caused by reduced albumin synthesis associated with inflammation and not decreased normalized protein catabolic rate. The cause(s) of inflammation must be identified and treated to resolve malnutrition and reduce cardiovascular risk.
Collapse
Affiliation(s)
- George A Kaysen
- Division of Nephrology, Department of Medicine, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
157
|
Wyke SM, Tisdale MJ. NF-kappaB mediates proteolysis-inducing factor induced protein degradation and expression of the ubiquitin-proteasome system in skeletal muscle. Br J Cancer 2005; 92:711-21. [PMID: 15714207 PMCID: PMC2361865 DOI: 10.1038/sj.bjc.6602402] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Loss of skeletal muscle in cancer cachexia has a negative effect on both morbidity and mortality. The role of nuclear factor-κB (NF-κB) in regulating muscle protein degradation and expression of the ubiquitin–proteasome proteolytic pathway in response to a tumour cachectic factor, proteolysis-inducing factor (PIF), has been studied by creating stable, transdominant-negative, muscle cell lines. Murine C2C12 myoblasts were transfected with plasmids with a CMV promoter that had mutations at the serine phosphorylation sites required for degradation of I-κBα, an NF-κB inhibitory protein, and allowed to differentiate into myotubes. Proteolysis-inducing factor induced degradation of I-κBα, nuclear accumulation of NF-κB and an increase in luciferase reporter gene activity in myotubes containing wild-type, but not mutant, I-κBα proteins. Proteolysis-inducing factor also induced total protein degradation and loss of the myofibrillar protein myosin in myotubes containing wild-type, but not mutant, plasmids at the same concentrations as those causing activation of NF-κB. Proteolysis-inducing factor also induced increased expression of the ubiquitin–proteasome pathway, as determined by ‘chymotrypsin-like’ enzyme activity, the predominant proteolytic activity of the β-subunits of the proteasome, protein expression of 20S α-subunits and the 19S subunits MSS1 and p42, as well as the ubiquitin conjugating enzyme, E214k, in cells containing wild-type, but not mutant, I-κBα. The ability of mutant I-κBα to inhibit PIF-induced protein degradation, as well as expression of the ubiquitin–proteasome pathway, confirms that both of these responses depend on initiation of transcription by NF-κB.
Collapse
Affiliation(s)
- S M Wyke
- Pharmaceutical Sciences Research Institute, Aston University, Birmingham, B4 7ET, UK
| | - M J Tisdale
- Pharmaceutical Sciences Research Institute, Aston University, Birmingham, B4 7ET, UK
- Pharmaceutical Sciences Research Institute, Aston University, Birmingham, B4 7ET, UK. E-mail:
| |
Collapse
|
158
|
Lin SY, Chen WY, Lee FY, Huang CJ, Sheu WHH. Activation of ubiquitin-proteasome pathway is involved in skeletal muscle wasting in a rat model with biliary cirrhosis: potential role of TNF-alpha. Am J Physiol Endocrinol Metab 2005; 288:E493-501. [PMID: 15522995 DOI: 10.1152/ajpendo.00186.2004] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hepatic cirrhosis is associated with negative nitrogen balance and loss of lean body mass. This study aimed to identify the specific proteolytic pathways activated in skeletal muscles of cirrhotic rats. TNF-alpha can stimulate muscle proteolysis; therefore, a potential relationship between TNF-alpha and muscle wasting in liver cirrhosis was also evaluated. Cirrhosis was induced by bile duct ligation (BDL) in male adult Sprague-Dawley rats. mRNA and protein levels of various targets were determined by RT-PCR and Western blotting, respectively. The proteolytic rate was measured ex vivo using isolated muscles. Compared with sham-operated controls, BDL rats had an increased degradation rate of muscle proteins and enhanced gene expression of ubiquitin, 14-kDa ubiquitin carrier protein E2, and the proteasome subunits C2 and C8 (P < 0.01). The muscle protein levels of free ubiquitin and conjugated ubiquitin levels were also elevated (P < 0.01). However, there was no difference between the two groups with regard to cathepsin and calpain mRNA levels. Cirrhotic muscle TNF-alpha levels were increased and correlated positively with free and conjugated ubiquitin (P < 0.01). We conclude that the ubiquitin-proteasome system is involved in muscle wasting of rats with BDL-induced cirrhosis. TNF-alpha might play a role in mediating activation of this proteolytic pathway, probably through a local mechanism.
Collapse
Affiliation(s)
- Shih-Yi Lin
- Division of Endocrinology and Metabolism, Department of Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | | | | | | | | |
Collapse
|
159
|
Wing SS. Control of ubiquitination in skeletal muscle wasting. Int J Biochem Cell Biol 2004; 37:2075-87. [PMID: 16125111 DOI: 10.1016/j.biocel.2004.11.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2004] [Revised: 11/15/2004] [Accepted: 11/18/2004] [Indexed: 11/23/2022]
Abstract
The ubiquitin proteasome system is now well recognized to play a role in mediating skeletal muscle protein wasting. Ubiquitin exerts its effects by covalent attachment to other proteins. Increased ubiquitination of muscle proteins has been observed in a number of conditions of atrophy suggesting that flux through the pathway may be regulated by controlling availability of ubiquitinated substrates for the proteasome. Therefore the enzymes that control ubiquitination of proteins likely play critical roles in regulating flux through the pathway, are sites of activation by catabolic stimuli and potentially good drug targets in the search for therapies for wasting disorders. In this article, the enzymes that can modulate ubiquitination are briefly reviewed and the current data regarding regulation of these enzymes in skeletal muscle are described. Physiological regulators of muscle size appear to modulate many of these enzymes and several of these regulators appear to do so via signaling pathways that involve Akt or NFkappaB. Further work needs to be done to identify all the enzymes that are involved in controlling ubiquitination in muscle, to characterize their regulation by non-transcriptional mechanisms also, and most importantly to identify their target substrates and to determine how these various pathways of ubiquitination work together to mediate the catabolic stimulus.
Collapse
Affiliation(s)
- Simon S Wing
- Polypeptide Laboratory, Department of Medicine, McGill University, Strathcona Anatomy and Dentistry Bldg, Room W315, 3640 University St., Montreal, Que., Canada H3A 2B2.
| |
Collapse
|
160
|
Cao PR, Kim HJ, Lecker SH. Ubiquitin-protein ligases in muscle wasting. Int J Biochem Cell Biol 2004; 37:2088-97. [PMID: 16125112 DOI: 10.1016/j.biocel.2004.11.010] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2004] [Revised: 11/02/2004] [Accepted: 11/17/2004] [Indexed: 11/18/2022]
Abstract
Muscle wasting occurs when rates of protein degradation outstrip rates of protein synthesis. Accelerated rates of protein degradation develop in atrophying muscle largely through activation of the ubiquitin-proteasome pathway. The complexity of the ubiquitination process, however, has hampered our understanding of how this pathway is activated in atrophying muscles and which enzymes of the ubiquitin conjugation system are responsible. Recent studies demonstrate that two ubiquitin-protein ligases (E3s), atrogin-1/MAFbx and MuRF1 are critical in the development of muscle atrophy. Other experiments implicate E2(14k) and E3alpha, of the N-end rule pathway, as important players in the process. It seems likely that multiple pathways of ubiquitin conjugation are activated in parallel in atrophying muscle, perhaps to target for degradation specific classes of muscle proteins. The emerging challenge will be to define the protein targets for, as well as to develop inhibitors of, these E3s.
Collapse
Affiliation(s)
- Pei Rang Cao
- Renal Unit, DA517, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | | | | |
Collapse
|
161
|
Abstract
Skeletal muscle atrophy has extreme adverse consequences. Molecular mechanisms that mediate the process of atrophy are not well defined. Recent studies have focused on diverse molecular cascades that control the activation of ubiquitin ligases, indicating that the involvement of the ubiquitin proteasome may be common to a range of atrophic stimuli.
Collapse
Affiliation(s)
- Iain W McKinnell
- Ottawa Health Research Institute, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada
| | | |
Collapse
|
162
|
Hunter RB, Kandarian SC. Disruption of either the Nfkb1 or the Bcl3 gene inhibits skeletal muscle atrophy. J Clin Invest 2004. [DOI: 10.1172/jci200421696] [Citation(s) in RCA: 178] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
163
|
Hunter RB, Kandarian SC. Disruption of either the Nfkb1 or the Bcl3 gene inhibits skeletal muscle atrophy. J Clin Invest 2004; 114:1504-11. [PMID: 15546001 PMCID: PMC525738 DOI: 10.1172/jci21696] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2004] [Accepted: 09/21/2004] [Indexed: 11/17/2022] Open
Abstract
The intracellular signals that mediate skeletal muscle protein loss and functional deficits due to muscular disuse are just beginning to be elucidated. Previously we showed that the activity of an NF-kappaB-dependent reporter gene was markedly increased in unloaded muscles, and p50 and Bcl-3 proteins were implicated in this induction. In the present study, mice with a knockout of the p105/p50 (Nfkb1) gene are shown to be resistant to the decrease in soleus fiber cross-sectional area that results from 10 days of hindlimb unloading. Furthermore, the marked unloading-induced activation of the NF-kappaB reporter gene in soleus muscles from WT mice was completely abolished in soleus muscles from Nfkb1 knockout mice. Knockout of the B cell lymphoma 3 (Bcl3) gene also showed an inhibition of fiber atrophy and an abolition of NF-kappaB reporter activity. With unloading, fast fibers from WT mice atrophied to a greater extent than slow fibers. Resistance to atrophy in both strains of knockout mice was demonstrated clearly in fast fibers, while slow fibers from only the Bcl3(-/-) mice showed atrophy inhibition. The slow-to-fast shift in myosin isoform expression due to unloading was also abolished in both Nfkb1 and Bcl3 knockout mice. Like the soleus muscles, plantaris muscles from Nfkb1(-/-) and Bcl3(-/-) mice also showed inhibition of atrophy with unloading. Thus both the Nfkb1 and the Bcl3 genes are necessary for unloading-induced atrophy and the associated phenotype transition.
Collapse
Affiliation(s)
- R Bridge Hunter
- Department of Health Sciences, Boston University, Boston, Massachusetts 02215, USA
| | | |
Collapse
|
164
|
Abstract
Skeletal muscle atrophy attributable to muscular inactivity has significant adverse functional consequences. While the initiating physiological event leading to atrophy seems to be the loss of muscle tension and a good deal of the physiology of muscle atrophy has been characterized, little is known about the triggers or the molecular signaling events underlying this process. Decreases in protein synthesis and increases in protein degradation both have been shown to contribute to muscle protein loss due to disuse, and recent work has delineated elements of both synthetic and proteolytic processes underlying muscle atrophy. It is also becoming evident that interactions among known proteolytic pathways (ubiquitin-proteasome, lysosomal, and calpain) are involved in muscle proteolysis during atrophy. Factors such as TNF-alpha, glucocorticoids, myostatin, and reactive oxygen species can induce muscle protein loss under specified conditions. Also, it is now apparent that the transcription factor NF-kappaB is a key intracellular signal transducer in disuse atrophy. Transcriptional profiles of atrophying muscle show both up- and downregulation of various genes over time, thus providing further evidence that there are multiple concurrent processes involved in muscle atrophy. The purpose of this review is to synthesize our current understanding of the molecular regulation of muscle atrophy. We also discuss how ongoing work should uncover more about the molecular underpinnings of muscle wasting, particularly that due to disuse.
Collapse
Affiliation(s)
- Robert W Jackman
- Boston University, Department of Health Sciences, 635 Commonwealth Ave., Rm. 443, Boston, MA 02215, USA
| | | |
Collapse
|
165
|
Sacheck JM, Ohtsuka A, McLary SC, Goldberg AL. IGF-I stimulates muscle growth by suppressing protein breakdown and expression of atrophy-related ubiquitin ligases, atrogin-1 and MuRF1. Am J Physiol Endocrinol Metab 2004; 287:E591-601. [PMID: 15100091 DOI: 10.1152/ajpendo.00073.2004] [Citation(s) in RCA: 425] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Muscle atrophy results primarily from accelerated protein degradation and is associated with increased expression of two muscle-specific ubiquitin ligases (E3s): atrogin-1 and muscle ring finger 1 (MuRF1). Glucocorticoids are essential for many types of muscle atrophy, and their effects are opposite to those of insulin-like growth factor I (IGF-I) and insulin, which promote growth. In myotubes, dexamethasone (Dex) inhibited growth and enhanced breakdown of long-lived cell proteins, especially myofibrillar proteins (as measured by 3-methylhistidine release), while also increasing atrogin-1 and MuRF1 mRNA. Conversely, IGF-I suppressed protein degradation and prevented the Dex-induced increase in proteolysis. IGF-I rapidly reduced atrogin-1 expression within 1 h by blocking mRNA synthesis without affecting mRNA degradation, whereas IGF-I decreased MuRF1 mRNA slowly. IGF-I and insulin also blocked Dex induction of these E3s and several other atrophy-related genes ("atrogenes"). Changes in overall proteolysis with Dex and IGF-I correlated tightly with changes in atrogin-1 mRNA content, but not with changes in MuRF1 mRNA. IGF-I activates the phosphatidylinositol 3-kinase (PI3K)-Akt pathway, and inhibition of this pathway [but not the calcineurin-nuclear factor of activated T cell (NFAT) or the MEK-ERK pathway] increased proteolysis and atrogin-1 mRNA expression. Thus an important component of growth stimulation by IGF-I, through the PI3K-Akt pathway, is its ability to rapidly suppress transcription of the atrophy-related E3 atrogin-1 and other atrogenes and degradation of myofibrillar proteins.
Collapse
MESH Headings
- Blotting, Northern
- Cells, Cultured
- Dexamethasone/pharmacology
- Enzyme Inhibitors/pharmacology
- Gene Expression Regulation, Enzymologic/drug effects
- Humans
- Hypoglycemic Agents/pharmacology
- Insulin/pharmacology
- Insulin Resistance/physiology
- Insulin-Like Growth Factor I/pharmacology
- Methylhistidines/metabolism
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle Proteins/antagonists & inhibitors
- Muscle Proteins/biosynthesis
- Muscle Proteins/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/growth & development
- Muscular Atrophy/metabolism
- Myoblasts/drug effects
- Myoblasts/metabolism
- Myofibrils/metabolism
- Phosphoinositide-3 Kinase Inhibitors
- RNA, Messenger/biosynthesis
- Reverse Transcriptase Polymerase Chain Reaction
- SKP Cullin F-Box Protein Ligases/antagonists & inhibitors
- SKP Cullin F-Box Protein Ligases/biosynthesis
- Stimulation, Chemical
- Tripartite Motif Proteins
- Ubiquitin/biosynthesis
- Ubiquitin-Protein Ligases/antagonists & inhibitors
- Ubiquitin-Protein Ligases/biosynthesis
Collapse
Affiliation(s)
- Jennifer M Sacheck
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave., Boston, MA 02115, USA
| | | | | | | |
Collapse
|
166
|
Acharyya S, Ladner KJ, Nelsen LL, Damrauer J, Reiser PJ, Swoap S, Guttridge DC. Cancer cachexia is regulated by selective targeting of skeletal muscle gene products. J Clin Invest 2004; 114:370-8. [PMID: 15286803 PMCID: PMC484974 DOI: 10.1172/jci20174] [Citation(s) in RCA: 197] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2003] [Accepted: 06/03/2004] [Indexed: 02/06/2023] Open
Abstract
Cachexia is a syndrome characterized by wasting of skeletal muscle and contributes to nearly one-third of all cancer deaths. Cytokines and tumor factors mediate wasting by suppressing muscle gene products, but exactly which products are targeted by these cachectic factors is not well understood. Because of their functional relevance to muscle architecture, such targets are presumed to represent myofibrillar proteins, but whether these proteins are regulated in a general or a selective manner is also unclear. Here we demonstrate, using in vitro and in vivo models of muscle wasting, that cachectic factors are remarkably selective in targeting myosin heavy chain. In myotubes and mouse muscles, TNF-alpha plus IFN-gamma strongly reduced myosin expression through an RNA-dependent mechanism. Likewise, colon-26 tumors in mice caused the selective reduction of this myofibrillar protein, and this reduction correlated with wasting. Under these conditions, however, loss of myosin was associated with the ubiquitin-dependent proteasome pathway, which suggests that mechanisms used to regulate the expression of muscle proteins may be cachectic factor specific. These results shed new light on cancer cachexia by revealing that wasting does not result from a general downregulation of muscle proteins but rather is highly selective as to which proteins are targeted during the wasting state.
Collapse
Affiliation(s)
- Swarnali Acharyya
- Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus 43210, USA
| | | | | | | | | | | | | |
Collapse
|
167
|
Acharyya S, Ladner KJ, Nelsen LL, Damrauer J, Reiser PJ, Swoap S, Guttridge DC. Cancer cachexia is regulated by selective targeting of skeletal muscle gene products. J Clin Invest 2004. [DOI: 10.1172/jci200420174] [Citation(s) in RCA: 351] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
168
|
Sartorelli V, Fulco M. Molecular and cellular determinants of skeletal muscle atrophy and hypertrophy. Sci Signal 2004; 2004:re11. [PMID: 15292521 DOI: 10.1126/stke.2442004re11] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The maintenance of adult skeletal muscle mass is ensured by physical exercise. Accordingly, physiological and pathological situations characterized by either impaired motor neuron activity, reduced gravity (microgravity during space flights), or reduced physical activity result in loss of muscle mass. Furthermore, a plethora of clinical conditions, including cancer, sepsis, diabetes, and AIDS, are associated with varying degrees of muscle atrophy. The cellular and molecular pathways responsible for maintaining the skeletal muscle mass are not well defined. Nonetheless, studies aimed at the understanding of the mechanisms underlying either muscular atrophy or hypertrophy have begun to identify the physiological determinants and clarify the molecular pathways responsible for the maintenance of muscle mass.
Collapse
Affiliation(s)
- Vittorio Sartorelli
- Muscle Gene Expression Group Laboratory of Muscle Biology, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
169
|
Durham WJ, Li YP, Gerken E, Farid M, Arbogast S, Wolfe RR, Reid MB. Fatiguing exercise reduces DNA binding activity of NF-kappaB in skeletal muscle nuclei. J Appl Physiol (1985) 2004; 97:1740-5. [PMID: 15208298 DOI: 10.1152/japplphysiol.00088.2004] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study tested the hypothesis that skeletal muscle contraction activates nuclear factor-kappaB (NF-kappaB), a putative regulator of muscle protein breakdown. Muscle biopsies were obtained from the vastus lateralis of healthy humans before, immediately after, and 1 h after fatiguing resistance exercise of the lower limbs. Biopsies were analyzed for nuclear NF-kappaB DNA binding activity by using electrophoretic mobility shift assay. NF-kappaB activity, measured immediately after exercise, was less than preexercise activity; after 1-h recovery, activity returned to preexercise levels. In follow-up studies in adult mice, basal NF-kappaB activity varied among individual muscles. NF-kappaB activity in diaphragm fiber bundles was decreased after a 10-min bout of fatiguing tetanic contractions in vitro. NF-kappaB activity in soleus was increased by 12 days of unloading by hindlimb suspension; this increase was reversed by 10 min of fatiguing exercise. These data provide no support for our original hypothesis. Instead, acute fatiguing exercise appears to decrease NF-kappaB activity in muscle under a variety of conditions.
Collapse
Affiliation(s)
- William J Durham
- Pulmonary and Critical Care Medicine Section, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | |
Collapse
|
170
|
Franch HA, Raissi S, Wang X, Zheng B, Bailey JL, Price SR. Acidosis impairs insulin receptor substrate-1-associated phosphoinositide 3-kinase signaling in muscle cells: consequences on proteolysis. Am J Physiol Renal Physiol 2004; 287:F700-6. [PMID: 15161606 DOI: 10.1152/ajprenal.00440.2003] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic acidosis is a stimulus for proteolysis in muscle in vivo, but the mechanism of this response is unknown. We tested the hypothesis that acidosis or TNF-alpha, a cytokine whose production increases in acidosis, regulates proteolysis by inhibiting insulin signaling through phosphoinositide 3-kinase (PI3K). In cultured L6 myotubes, acidified (pH 7.1) media did not accelerate the basal protein degradation rate, but it inhibited insulin's ability to suppress proteolysis. Insulin receptor substrate-1 (IRS-1)-associated PI3K activity was not altered in cells acidified for 10 min but was strongly inhibited in cells incubated at pH 7.1 for 24 h. Phosphorylation of Akt was also suppressed by acidification for 24 h. Acidification did not induce changes in IRS-1 abundance, insulin-stimulated IRS-1 tyrosine phosphorylation, or the amount of PI3K p85 regulatory subunit. In contrast to acidification, TNF-alpha suppressed proteolysis in the presence or absence of insulin but had no effect on IRS-1-associated PI3K activity. To establish that the PI3K pathway can regulate protein degradation in muscle, we measured proteolysis in cells after inhibition of PI3K activity with LY-294002 or infection with an adenovirus encoding a dominant negative PI3K p85alpha-subunit. Both approaches inhibited insulin-induced suppression of proteolysis to a degree similar to that seen with acidification. We conclude that acidosis accelerates protein degradation by impairing insulin signaling through PI3K in muscle cells.
Collapse
Affiliation(s)
- Harold A Franch
- Renal Divisioin, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | | | | | | | | | |
Collapse
|
171
|
Abstract
PURPOSE OF REVIEW Metabolic abnormalities leading to a catabolic syndrome with progressive muscular atrophy are a common final stage of various chronic diseases. Proinflammatory cytokines have been suggested both to induce and mediate local catabolic mechanisms. This review focuses on the role of proinflammatory cytokines in the development of muscular abnormalities resulting in a loss of muscle mass and function. In addition, the underlying molecular signaling pathways, their transcriptional regulation, and the cellular systems contributing to enhanced muscular protein breakdown are discussed. RECENT FINDINGS Using transcriptional screening techniques, specific changes in gene expression have been identified that are characteristic of muscular wasting processes. Of particular interest is the atrophy-related and cytokine-inducible expression of a number of E3 ligases (e.g. atrogin-1, muscle ring finger protein 1), highly specific regulators of the ubiquitin-proteasome pathway, which target proteins for proteolytic breakdown by the proteasome. Furthermore, the activity of several transcription factors (e.g. nuclear factor kappa B) has been involved in specific transcriptional mechanisms of local inflammation and muscular atrophy. Finally, proinflammatory cytokines suppress the expression and function of the local anabolic growth factor insulin-like growth factor 1. SUMMARY Pronounced catabolic effects of proinflammatory cytokines in various tissues contribute to local catabolism, with progressive atrophic alterations of the skeletal muscle in chronic disease states. Recent developments in this field have explored the underlying pathways of muscular wasting and have already resulted in the description of new molecular targets that might lead to new therapeutic options for the treatment of muscular atrophy.
Collapse
Affiliation(s)
- Ulrike Späte
- The Forsyth Institute, Department of Cytokine Biology, Boston, Massachusetts, USA
| | | |
Collapse
|
172
|
Hamel FG, Fawcett J, Bennett RG, Duckworth WC. Control of proteolysis: hormones, nutrients, and the changing role of the proteasome. Curr Opin Clin Nutr Metab Care 2004; 7:255-8. [PMID: 15075915 DOI: 10.1097/00075197-200405000-00003] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The maintenance of protein balance is essential for the proper functioning of a cell. Protein degradation must be controlled to account for the availability of nutrients and hormone signals from the body as a whole. The proteasome is the major cytosolic protein degrading machinery, and is responsible for a considerable proportion of cellular protein degradation. It is thus a prime site for the integration of these various signals. We will examine some recent data regarding the mechanisms for control of the peptidolytic activities of the proteasome, and possible implications for signal transduction and integration. RECENT FINDINGS Nutrients, such as amino acids and fatty acids, have been shown to have effects on proteasome-mediated protein degradation. The ubiquitinylating process is important for the control of protein degradation by the 26S proteasome. Amino acids and hormones control the expression of the necessary components, and can control protein degradation on a relatively longer-term basis. The 20S proteasome has been shown to be capable of degrading proteins without activating subunits. Furthermore, the 20S proteasome is allosterically affected by a number of smaller peptides, suggesting a more immediate mechanism for control. Amino acids and fatty acids have been shown to exert such control in vitro. SUMMARY As more is learned about the functioning of the proteasome, the greater appreciation we have of its vital role in the control of cellular metabolism. Recent evidence shows that the proteasome is central to the integration of various nutrient and hormonal signals that the cell receives that may impact on protein metabolism.
Collapse
Affiliation(s)
- Frederick G Hamel
- Research Service, Department of Veterans Affairs Medical Center, and Department of Internal Medicine, University of Nebraska Medical Center, Omaha, 68105, USA.
| | | | | | | |
Collapse
|
173
|
Kaysen GA, Müller HG, Young BS, Leng X, Chertow GM. The influence of patient- and facility-specific factors on nutritional status and survival in hemodialysis. J Ren Nutr 2004; 14:72-81. [PMID: 15060871 DOI: 10.1053/j.jrn.2004.01.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Parameters of nutritional status, including serum albumin, serum creatinine, and body mass index (BMI), are powerful predictors of mortality and hospitalization in patients with end stage renal disease (ESRD). Patient-specific characteristics and facility-related practice patterns modify certain parameters of nutritional status. We aimed to determine whether patient and facility characteristics modify the risk profiles associated with malnutrition in hemodialysis patients. METHODS We analyzed data on 5,234 prevalent hemodialysis patients from the Dialysis Morbidity and Mortality Study (DMMS) Wave 1 for whom information on demographic, clinical, nutritional, and facility-related characteristics were available. We evaluated the associations among facility characteristics and serum albumin, serum creatinine, and BMI, adjusting for the effects of age, sex, race/ethnicity, diabetes, and dialysis vintage. We determined correlates of mortality and hospitalization, focusing on nutritional parameters, facility effects, and the interactions among patient-specific and facility-specific characteristics, albumin, creatinine, and BMI. RESULTS Serum albumin was lower with older age, diabetes, nonblack race, and hemodialysis using a catheter. Serum albumin was higher with annual vascular access surveillance, higher BMI among women, higher urea reduction ratio, among patients in whom dialyzers were reprocessed (particularly with bleach), among dialysis units in which water purification was used, and when vascular access blood flow rates were > or =350 mL/min. Overall survival was decreased with lower albumin, creatinine, and BMI. There were interactions among albumin, age, and vintage. Whereas lower serum albumin concentrations consistently were associated with an increased risk of death, the differences were attenuated among older patients and accentuated among patients of longer vintage. CONCLUSION Some facility-specific factors are associated with nutritional parameters including serum albumin, serum creatinine, and BMI. The associations of nutritional parameters with mortality and hospitalization vary by age, sex, and vintage but not by facility-specific factors, including those associated with the nutritional parameters themselves.
Collapse
Affiliation(s)
- George A Kaysen
- Division of Nephrology, Department of Medicine, University of California Davis, Davis, CA 94118-1211, USA
| | | | | | | | | |
Collapse
|
174
|
Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, Walsh K, Schiaffino S, Lecker SH, Goldberg AL. Foxo Transcription Factors Induce the Atrophy-Related Ubiquitin Ligase Atrogin-1 and Cause Skeletal Muscle Atrophy. Cell 2004; 117:399-412. [PMID: 15109499 PMCID: PMC3619734 DOI: 10.1016/s0092-8674(04)00400-3] [Citation(s) in RCA: 2273] [Impact Index Per Article: 108.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2004] [Revised: 02/24/2004] [Accepted: 03/17/2004] [Indexed: 11/25/2022]
Abstract
Skeletal muscle atrophy is a debilitating response to fasting, disuse, cancer, and other systemic diseases. In atrophying muscles, the ubiquitin ligase, atrogin-1 (MAFbx), is dramatically induced, and this response is necessary for rapid atrophy. Here, we show that in cultured myotubes undergoing atrophy, the activity of the PI3K/AKT pathway decreases, leading to activation of Foxo transcription factors and atrogin-1 induction. IGF-1 treatment or AKT overexpression inhibits Foxo and atrogin-1 expression. Moreover, constitutively active Foxo3 acts on the atrogin-1 promoter to cause atrogin-1 transcription and dramatic atrophy of myotubes and muscle fibers. When Foxo activation is blocked by a dominant-negative construct in myotubes or by RNAi in mouse muscles in vivo, atrogin-1 induction during starvation and atrophy of myotubes induced by glucocorticoids are prevented. Thus, forkhead factor(s) play a critical role in the development of muscle atrophy, and inhibition of Foxo factors is an attractive approach to combat muscle wasting.
Collapse
Affiliation(s)
- Marco Sandri
- Department of Cell Biology Harvard Medical School Boston, Massachusetts 02115
| | - Claudia Sandri
- Department of Biomedical Sciences Venetian Institute of Molecular Medicine 35129 Padova Italy
| | - Alex Gilbert
- Renal Unit Beth Israel Deaconess Medical Center Boston, Massachusetts 02215
| | - Carsten Skurk
- Boston University Medical School Cardiovascular Research Institute Boston, Massachusetts 02118
| | - Elisa Calabria
- Department of Biomedical Sciences Venetian Institute of Molecular Medicine 35129 Padova Italy
| | - Anne Picard
- Department of Biomedical Sciences Venetian Institute of Molecular Medicine 35129 Padova Italy
| | - Kenneth Walsh
- Boston University Medical School Cardiovascular Research Institute Boston, Massachusetts 02118
| | - Stefano Schiaffino
- Department of Biomedical Sciences Venetian Institute of Molecular Medicine 35129 Padova Italy
| | - Stewart H. Lecker
- Renal Unit Beth Israel Deaconess Medical Center Boston, Massachusetts 02215
- Correspondence: (S.H.L.), (A.L.G.)
| | - Alfred L. Goldberg
- Department of Cell Biology Harvard Medical School Boston, Massachusetts 02115
- Correspondence: (S.H.L.), (A.L.G.)
| |
Collapse
|
175
|
Bhat-Nakshatri P, Campbell RA, Patel NM, Newton TR, King AJ, Marshall MS, Ali S, Nakshatri H. Tumour necrosis factor and PI3-kinase control oestrogen receptor alpha protein level and its transrepression function. Br J Cancer 2004; 90:853-9. [PMID: 14970864 PMCID: PMC2410160 DOI: 10.1038/sj.bjc.6601541] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Oestrogen receptor alpha (ERα) is an oestrogen-activated transcription factor, which regulates proliferation and differentiation of mammary epithelial cells by activating or repressing gene expression. ERα is a critical prognostic indicator and a therapeutic target for breast cancer. Patients with tumours that express higher level of ERα have better prognosis than patients with tumours that are ERα negative or express lower level of ERα. Better prognosis in ERα-positive patients is believed to be due to repression of proinvasive gene expression by ERα. Oestrogen receptor alpha represses gene expression by transrepressing the activity of the transcription factors such as nuclear factor-kappaB or by inducing the expression of transcriptional suppressors such as MTA3. In this report, we show that ERα transrepresses the expression of the proinvasive gene interleukin 6 (IL-6) in ERα-negative MDA-MB-231 breast cancer cells stably overexpressing ERα. Using these cells as well as ERα-positive MCF-7 and ZR-75-1 cells, we show that tumour necrosis factor alpha (TNFα) and the phosphatidylinositol-3-kinase (PI3-kinase) modulate transrepression function of ERα by reducing its stability. From these results, we propose that TNFα expression or PI3-kinase activation lead to reduced levels of ERα protein in cancer cells and corresponding loss of transrepression function and acquisition of an invasive phenotype.
Collapse
Affiliation(s)
- P Bhat-Nakshatri
- Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Walther Cancer Institute, Indianapolis, IN 46208, USA
| | - R A Campbell
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - N M Patel
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - T R Newton
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - A J King
- Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - M S Marshall
- Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - S Ali
- Department of Cancer Medicine, Imperial College School of Medicine, Hammersmith Hospital, London W12 0NN, UK
| | - H Nakshatri
- Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Walther Cancer Institute, Indianapolis, IN 46208, USA
- R4-202, Indiana Cancer Research Institute, 1044 West Walnut Street, Indianapolis, IN 46202, USA. E-mail:
| |
Collapse
|
176
|
|
177
|
Langen RCJ, Van Der Velden JLJ, Schols AMWJ, Kelders MCJM, Wouters EFM, Janssen-Heininger YMW. Tumor necrosis factor-alpha inhibits myogenic differentiation through MyoD protein destabilization. FASEB J 2004; 18:227-37. [PMID: 14769817 DOI: 10.1096/fj.03-0251com] [Citation(s) in RCA: 235] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Tumor necrosis factor alpha (TNFalpha) has been implicated as a mediator of muscle wasting through nuclear factor kappa B (NF-kappaB) -dependent inhibition of myogenic differentiation. The aim of the present study was to identify the regulatory molecule(s) of myogenesis targeted by TNFalpha/NF-kappaB signaling. TNFalpha interfered with cell cycle exit and repressed the accumulation of transcripts encoding muscle-specific genes in differentiating C2C12 myoblasts. Overexpression of a p65 (RelA) mutant lacking the transcriptional activation domain attenuated the TNFalpha-mediated inhibition of muscle-specific gene transcription. The ability of muscle regulatory factor MyoD to induce muscle-specific transcription in 10T1/2 fibroblasts was also disrupted by wild-type p65, demonstrating that NF-kappaB transcriptional activity interferes with the function of MyoD. Inhibition of muscle-specific gene expression by TNFalpha was restored by overexpression of MyoD, whereas endogenous MyoD protein abundance and stability were reduced by TNFalpha through increased proteolysis of MyoD by the ubiquitin proteasome pathway. Last, the inhibitory effects of TNFalpha on myogenic differentiation were demonstrated in a mouse model of skeletal muscle regeneration, in which TNFalpha caused a delay in myoblast cell cycle exit. These results implicate that TNFalpha inhibits myogenic differentiation through destabilizing MyoD protein in a NF-kappaB-dependent manner, which interferes with skeletal muscle regeneration and may contribute to muscle wasting.
Collapse
Affiliation(s)
- Ramon C J Langen
- Department of Respiratory Medicine, Maastricht University, Maastricht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
178
|
Lecker SH, Jagoe RT, Gilbert A, Gomes M, Baracos V, Bailey J, Price SR, Mitch WE, Goldberg AL. Multiple types of skeletal muscle atrophy involve a common program of changes in gene expression. FASEB J 2004; 18:39-51. [PMID: 14718385 DOI: 10.1096/fj.03-0610com] [Citation(s) in RCA: 1187] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Skeletal muscle atrophy is a debilitating response to starvation and many systemic diseases including diabetes, cancer, and renal failure. We had proposed that a common set of transcriptional adaptations underlie the loss of muscle mass in these different states. To test this hypothesis, we used cDNA microarrays to compare the changes in content of specific mRNAs in muscles atrophying from different causes. We compared muscles from fasted mice, from rats with cancer cachexia, streptozotocin-induced diabetes mellitus, uremia induced by subtotal nephrectomy, and from pair-fed control rats. Although the content of >90% of mRNAs did not change, including those for the myofibrillar apparatus, we found a common set of genes (termed atrogins) that were induced or suppressed in muscles in these four catabolic states. Among the strongly induced genes were many involved in protein degradation, including polyubiquitins, Ub fusion proteins, the Ub ligases atrogin-1/MAFbx and MuRF-1, multiple but not all subunits of the 20S proteasome and its 19S regulator, and cathepsin L. Many genes required for ATP production and late steps in glycolysis were down-regulated, as were many transcripts for extracellular matrix proteins. Some genes not previously implicated in muscle atrophy were dramatically up-regulated (lipin, metallothionein, AMP deaminase, RNA helicase-related protein, TG interacting factor) and several growth-related mRNAs were down-regulated (P311, JUN, IGF-1-BP5). Thus, different types of muscle atrophy share a common transcriptional program that is activated in many systemic diseases.
Collapse
Affiliation(s)
- Stewart H Lecker
- Renal Unit, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Kaysen GA, Greene T, Daugirdas JT, Kimmel PL, Schulman GW, Toto RD, Levin NW, Yan G. Longitudinal and cross-sectional effects of C-reactive protein, equilibrated normalized protein catabolic rate, and serum bicarbonate on creatinine and albumin levels in dialysis patients. Am J Kidney Dis 2003; 42:1200-11. [PMID: 14655192 DOI: 10.1053/j.ajkd.2003.08.021] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Loss of muscle mass and hypoalbuminemia each may result in part from either malnutrition, inflammation, or a combination of both. Short-term acidosis increases muscle protein catabolism and inhibits albumin synthesis. METHODS We analyzed albumin and creatinine levels as outcome variables and their association with C-reactive protein (CRP) level, equilibrated normalized protein catabolic rate (enPCR), and serum bicarbonate level as independent variables from laboratory data obtained from patients in the Hemodialysis Study. Analyses controlled for race, sex, age, body mass index, and randomized treatment group. RESULTS Albumin level correlated with both enPCR and CRP level, but not serum bicarbonate level, in both cross-sectional and longitudinal analyses. Effects of CRP level and enPCR were not linear. Albumin level correlated positively with enPCR for an enPCR less than 1.0 g/kg/d, but not for a greater enPCR, and correlated inversely with CRP level for a CRP level greater than 13 mg/L. Similarly, creatinine level correlated with both enPCR and CRP level. As in the case of albumin level, effects were not linear. Creatinine level correlated positively with enPCR for values less than 1.0 g/kg/d, but not for greater enPCR values. In contrast to albumin level, creatinine level correlated negatively with serum bicarbonate level, even when adjusted for enPCR. CONCLUSION Albumin and creatinine levels are independently associated with nutrition (enPCR) and inflammation (CRP level). The cross-sectional relationship with enPCR is apparent only at values less than 1.0 g/kg/d. CRP level is associated with reduced albumin and creatinine values when increased to values greater than 5.6 mg/dL. CRP may be increased to levels associated with increased cardiovascular risk with little or no effect on either serum albumin or creatinine level. Thus, a normal albumin level does not exclude elevated CRP levels.
Collapse
Affiliation(s)
- George A Kaysen
- Veterans Affairs Northern California Health Care System, Mather, USA.
| | | | | | | | | | | | | | | |
Collapse
|
180
|
Razeghi P, Sharma S, Ying J, Li YP, Stepkowski S, Reid MB, Taegtmeyer H. Atrophic Remodeling of the Heart In Vivo Simultaneously Activates Pathways of Protein Synthesis and Degradation. Circulation 2003; 108:2536-41. [PMID: 14610007 DOI: 10.1161/01.cir.0000096481.45105.13] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Mechanical unloading of the heart results in atrophic remodeling. In skeletal muscle, atrophy is associated with inactivation of the mammalian target of rapamycin (mTOR) pathway and upregulation of critical components of the ubiquitin proteosome proteolytic (UPP) pathway. The hypothesis is that mechanical unloading of the mammalian heart has differential effects on pathways of protein synthesis and degradation.
Methods and Results—
In a model of atrophic remodeling induced by heterotopic transplantation of the rat heart, we measured gene transcription, protein expression, polyubiquitin content, and regulators of the mTOR pathway at 2, 4, 7, and 28 days. In atrophic hearts, there was an increase in polyubiquitin content that peaked at 7 days and decreased by 28 days. Furthermore, gene and protein expression of UbcH2, a ubiquitin conjugating enzyme, was also increased early in the course of unloading. Transcript levels of TNF-α, a known regulator of UbcH2-dependent ubiquitin conjugating activity, were upregulated early and transiently in the atrophying rat heart. Unexpectedly, p70S6K and 4EBP1, downstream components of mTOR, were activated in atrophic rat heart. This activation was independent of Akt, a known upstream regulator of mTOR. Rapamycin treatment of the unloaded rat hearts inhibited the activation of p70S6K and 4EBP1 and subsequently augmented atrophy in these hearts compared with vehicle-treated, unloaded hearts.
Conclusions—
Atrophy of the heart, secondary to mechanical unloading, is associated with early activation of the UPP. The simultaneous activation of the mTOR pathway suggests active remodeling, involving both protein synthesis and degradation.
Collapse
Affiliation(s)
- Peter Razeghi
- Division of Cardiology, University of Texas Houston-Medical School, Houston, Tex 77030, USA
| | | | | | | | | | | | | |
Collapse
|
181
|
Li YP, Chen Y, Li AS, Reid MB. Hydrogen peroxide stimulates ubiquitin-conjugating activity and expression of genes for specific E2 and E3 proteins in skeletal muscle myotubes. Am J Physiol Cell Physiol 2003; 285:C806-12. [PMID: 12773310 DOI: 10.1152/ajpcell.00129.2003] [Citation(s) in RCA: 240] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Reactive oxygen species (ROS) are thought to promote muscle atrophy in chronic wasting diseases, but the underlying mechanism has not been determined. Here we show that H2O2 stimulates ubiquitin conjugation to muscle proteins through transcriptional regulation of the enzymes (E2 and E3 proteins) that conjugate ubiquitin to muscle proteins. Incubation of C2C12 myotubes with 100 microM H2O2 increased the rate of 125I-labeled ubiquitin conjugation to muscle proteins in whole cell extracts. This response required at least 4-h exposure to H2O2 and persisted for at least 24 h. Preincubating myotubes with cycloheximide or actinomycin D blocked H2O2 stimulation of ubiquitin-conjugating activity, suggesting that gene transcription is required. Northern blot analyses revealed that H2O2 upregulates expression of specific E3 and E2 proteins that are thought to regulate muscle catabolism, including atrogin1/MAFbx, MuRF1, and E214k. These results suggest that ROS stimulate protein catabolism in skeletal muscle by upregulating the ubiquitin conjugation system.
Collapse
Affiliation(s)
- Yi-Ping Li
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | |
Collapse
|