• Non ci sono risultati.

Transcription Factors and Nuclear Cofactors in Muscle Wasting

N/A
N/A
Protected

Academic year: 2022

Condividi "Transcription Factors and Nuclear Cofactors in Muscle Wasting"

Copied!
9
0
0

Testo completo

(1)

in Muscle Wasting

P.-O. Hasselgren

Introduction

Muscle wasting is commonly seen in patients with sepsis, severe injury, and cancer [1, 2]. The loss of muscle mass in these conditions mainly reflects ubiquitin-protea- some-dependent degradation of myofibrillar proteins although other proteolytic mechanisms may be involved as well [3]. Muscle atrophy is regulated by multiple factors, including glucocorticoids [4], the pro-inflammatory cytokines, interleukin (IL)-1q and tumor necrosis factor (TNF)- [ [5, 6], and myostatin [7]. In addition to these catabolic factors, a lack of anabolic signals, such as insulin-like growth factor (IGF)-1 and insulin, is probably also important for the development of muscle wast- ing in various catabolic conditions.

Although it may be argued that the increased peripheral release of amino acids that is associated with muscle wasting may be beneficial to the organism by providing energy and substrates to various organs and tissues, including the liver, gut mucosa, and cells in the immune system, during prolonged and severe catabolic conditions, the negative effects of muscle atrophy clearly outweigh any potentially beneficial effects.

Thus, muscle wasting results in weakness and fatigue that in turn delays ambulation of bed-ridden patients, increasing the risk for thromboembolic and pulmonary complica- tions. The risk for pulmonary complications is further increased when the muscle wast- ing process affects respiratory muscles [8]. In patients with cancer, the accompanying muscle wasting can significantly impair quality of life and may even reduce the effec- tiveness of chemotherapy [9]. It has been estimated that approximately 20 % of deaths in cancer patients can be attributed to the catabolic response in skeletal muscle [10].

Thus, it is obvious that muscle wasting is a significant clinical problem with sometimes devastating consequences. Despite substantial progress during the past decade in our understanding of mechanisms regulating the development of muscle atrophy, we still do not have any effective treatment by which muscle wasting can be prevented or reversed in critically ill patients. Continued efforts to understand the molecular mechanisms behind the loss of muscle mass in these patients will hope- fully help in the development of therapeutic strategies for subjects with this debili- tating condition. In this chapter, novel insights into the role of various transcription factors and nuclear cofactors in the development of muscle atrophy are discussed.

Gene Transcription in Muscle Wasting

In previous studies, we and others found evidence that mRNA levels for various genes in the ubiquitin-proteasome pathway are increased in muscle wasting condi-

(2)

tions. For example, in research from our laboratory, the gene expression for ubiqui- tin as well as different components of the 26S proteasome was increased in muscle from septic [11, 12] and burned rats [13] and in muscle from patients with sepsis [14] or cancer [15]. Results in other studies suggested that the increased mRNA lev- els for various components of the ubiquitin-proteaosme pathway reflected upregula- ted transcription of the genes rather than increased stability of the mRNA [16].

In more recent studies, Lecker et al. [17] reported that multiple catabolic condi- tions (uremia, fasting, muscle inactivity and denervation) were characterized by a common set of genes that were significantly upregulated. They called these genes

“atrogins” and found that the mRNA levels for two ubiquitin ligases, atrogin-1 and muscle ring finger 1 (MuRF1), were particularly increased [18]. The dramatic increase in the expression of these genes in atrophying muscle has been confirmed by others as well [19] and in studies in our laboratory, atrogin-1 and MuRF1 mRNA levels were increased almost 20-fold in skeletal muscle during sepsis [20]. Other studies provided evidence that the atrogin-1 and MuRF1 gene products regulate the development of muscle atrophy caused by various catabolic conditions and the mRNA levels for atrogin-1 and MuRF1 are frequently used as ‘molecular markers’ of muscle wasting.

Because the transcription of multiple genes is upregulated in atrophying muscle [17] it is not surprising that much attention has been given to the role of various transcription factors in the regulation of muscle mass. Here, the potential roles of the transcription factors C/EBPq and · , nuclear factor-kappa B (NF-κB) and Foxo1 and 3a in the development of muscle wasting are discussed. In addition, recent observations in our laboratory [21, 22] indicating an important role of the nuclear cofactor p300 and its histone actetyl transferase (HAT) activity in the regulation of muscle mass are described.

C/EBP Transcription Factors in Atrophying Muscle

The C/EBP (CCAAT/enhancer binding protein) family of transcription factors con- sists of at least six members: C/EBP[ , q , * , · , 5 , and C/EBP-homologous protein-10 (CHOP-10), also called Gadd 153 [23]. The different isoforms form homo- or hetero- dimers that influence the transcription of multiple genes in various organs and tis- sues. Among the different C/EBP family members, there is evidence that C/EBPq and · are particularly important for the inflammatory response [24].

We found that the expression of C/EBPq and · was upregulated in the nuclear fraction of muscles from septic rats [25]. This finding was accompanied by increased C/EBPq and · DNA binding activity determined by electrophoretic mobility shift assay (EMSA) with supershift analysis. Interestingly, the sepsis-induced increase in the expression and activity of C/EBPq and · was inhibited by the glucocorticoid receptor antagonist RU38486, supporting the role of glucocorticoids as an important mediator of sepsis-induced muscle wasting [4, 25]. Although we did not provide direct evidence that C/EBPq and · participate in the regulation of sepsis-induced muscle proteolysis, a sequence analysis demonstrated multiple putative binding sites for C/EBP in the promoter regions of genes that are upregulated in atrophying mus- cle, including genes for calpains and various components of the ubiquitin-protea- some proteolytic pathway [25].

Further support for a role of glucocorticoids in the upregulation of C/EBP tran- scription factors in atrophying skeletal muscle was found in experiments in which

(3)

we treated cultured myotubes in vitro or rats in vivo with dexamethasone [26]. In those experiments, treatment with dexamethasone resulted in increased protein and mRNA expression and DNA binding activity of C/EBPq and · . Because dexametha- sone treatment did not influence the expression of C/EBP[ , * , or 5 , it is possible that C/EBPq and · play a specific role among the C/EBP family members in atrophying muscle. The increase in C/EBPq and · mRNA levels in dexamtheasone-treated myo- tubes was not affected by the protein synthesis inhibitor, cycloheximide, indicating that the expression of the transcription factors was regulated directly by dexametha- sone. This differed from the dexamethasone-induced upregulation of atrogin-1 mRNA levels that was blocked by cycloheximide, consistent with a model in which the atrogin-1 gene is activated in skeletal muscle secondary to the upregulation of another gene or genes, possibly C/EBPq and · [26].

Thus, our recent observations suggest that C/EBPq and · may be involved in the development of sepsis- and glucocorticoid-induced muscle protein breakdown and atrophy. Interestingly, other members of the C/EBP family may regulate lipid metab- olism in atrophying muscle. Thus, in a recent study, muscle atrophy caused by denervation was associated with fatty degeneration and upregulated C/EBP[ expres- sion in the interstitium of the muscles, suggesting a role of C/EBP[ in lipid metabo- lism in atrophying muscle [27]. This observation supports previous reports that C/

EBP[ is a transcription factor involved in the regulation of lipid metabolism in vari- ous cell types, including muscle cells [28].

NF- ␬B Plays a Role in Muscle Wasting

NF-κB is probably the most extensively studied transcription factor in the field of inflammation. It is beyond the scope of this chapter to review the mechanisms that regulate NF-κB activation. Recent extensive review articles describe the molecular biology of this important transcription factor, its activation, and the mechanisms by which it upregulates gene transcription [29, 30].

Although the exact mechanisms by which NF-κB regulates muscle protein break- down are not completely understood at present, there is strong pharmacological and genetic evidence that NF-κB is involved in the development of muscle wasting in various catabolic conditions. We reported that NF-κB DNA binding activity was upregulated in skeletal muscle during early sepsis but was subsequently inhibited during the later course of an experimental model of sepsis in rats [31]. Interestingly, a biphasic response of NF-κB activity was observed in TNF- [ /interferon (IFN) * - treated myotubes as well [32]. Although the mechanisms for this biphasic response of NF-κB, and the biological implications with regards to which genes are activated or repressed, are not known at present, it is possible that the early activation and the late inhibition of NF-κB in septic muscle [31] reflects the role of different mediators involved in muscle wasting. For example, the early increase in NF-κB activity may reflect the influence of pro-inflammatory cytokines (most notably TNF-[ ) and the subsequent downregulation of NF-κB activity may reflect the effect of glucocorti- coids. We and others have reported several lines of evidence that both cytokines and glucocorticoids are important mediators of muscle wasting [4 – 6]. Other studies have shown that NF-κB activity is upregulated by pro-inflammatory cytokines, including TNF-[ [32, 33] and IL-1 q [34] in skeletal muscle cells and that NF-κB activity may be inhibited by glucocorticoids in myocytes [35]. This may seem para- doxical because treatment of cultured myotubes with either cytokines or catabolic

(4)

concentrations of dexamethasone results in increased protein degradation, activa- tion of the ubiquitin-proteaosme proteolytic pathway, and muscle atrophy [32, 33, 36, 37].

An additional potential mechanism to explain some of these apparently conflict- ing observations may be that NF-κB influences individual muscle wasting-related genes differentially. For example, there is evidence that NF-κB activates the expres- sion of the ubiquitin ligase, MuRF1, [38] but may act as a repressor of some of the proteaosme subunit genes [39]. An alternative explanation why the role of NF-κB may seem confusing (having a biphasic response; being activated or inhibited by mediators that all induce muscle atrophy) is that different pathways may be involved in the activation of NF-κB. The classic pathway, activated by pro-inflammatory cyto- kines, involves the activation of a p50/p65(RelA) heterodimer by ubiquitin-protea- some-dependent degradation of NF-κB inhibitor (IκB), triggered by its phosphory- lation by the IκB kinase, IKK q . An alternative pathway for NF-κB activation is the IKK[ -mediated phosphorylation and proteolytic processing of p100, resulting in activation of the non-canonical NF-κB pathway involving p52/RelB heterodimer [29]. The exact role of the different pathways activating NF-κB and the influence of cytokines and glucocorticoids on the regulation of these pathways in atrophying muscle are areas for future study.

Although important questions remain to be addressed with regards to the role of NF-κB in muscle wasting, in recent studies we have found strong molecular evidence that NF-κB activation results in muscle wasting [38]. In those experiments, trans- genic mice created in the laboratory of Dr Steven Shoelson were used. Mice with a muscle-specific overexpression of activated IKKq displayed upregulated NF-κB activity, increased expression of MuRF1 (but, interestingly enough, not atrogin-1), atrophy of muscle fibers, and a substantial loss of muscle mass. In other experiments in the same study, muscle-specific expression of an IκB [ superrepressor blocked NF-κB activation and prevented denervation- and tumor-induced muscle loss. A similar prevention of muscle atrophy was seen in mice treated with pharmacological inhibitors of NF-κB further supporting a role of NF-κB in the development of mus- cle wasting. In recent (unpublished) experiments in our laboratory we found that treatment of rats with the NF-κB inhibitor, curcumin, prevented sepsis-induced muscle proteolysis, suggesting that inhibition of NF-κB may prevent muscle wasting in different conditions characterized by muscle cachexia.

It should be noted that although there is evidence that NF-κB is activated in cata- bolic muscle in vivo [31] and in cytokine-treated muscle cells in vitro [32 – 34], and that muscle-specific activation of NF-κB in transgenic mice results in muscle atrophy [38], several important questions remain to be answered. First, it will be important in future studies to determine the mechanisms and biological consequences of the biphasic response of NF-κB in catabolic muscle and in cytokine-treated myotubes.

Second, the apparent contradiction between stimulation of protein degradation by both glucocorticoids and cytokines on the one hand, and the inhibition of NF-κB by glucocorticoids and activation of NF-κB by cytokines on the other hand, needs to be resolved. Third, it will be important to determine in greater detail which muscle wasting-related genes are upregulated and inhibited by NF-κB. Finally, the role of an interaction between glucocorticoids and cytokines, both at a systemic and a cellular level, in the regulation of NF-κB activity needs to be determined.

(5)

Activation of Foxo Transcription Factors Results in Muscle Atrophy

Recent studies support an important role of some of the Foxo (Forkhead box O) transcription factors in the development of muscle atrophy [40, 41]. The Foxo sub- family of the forkhead transcription factors consists of three members: Foxo1, 3a, and 4. These transcription factors are downstream targets of Akt and are inactivated by Akt-mediated phosphorylation. Phosphorylated Foxo transcription factors are retained in the cytoplasm in their inactive form. Dephosphorylation results in trans- port into the nucleus of the activated Foxo transcription factors. In recent studies in cultured myotubes, treatment with dexamethasone activated Foxo1 and 3a and upre- gulated the atrogin-1 and MuRF1 genes, resulting in myotube atrophy [40, 41].

These observations are in line with in vivo observations of an association between Foxo transcription factor activation and muscle atrophy in diabetes, fasting, cachexia, and aging [reviewed in 42]. The influence of sepsis, severe injury, and can- cer on Foxo transcription factors in skeletal muscle remains to be determined but considering the common response of multiple atrogins in different catabolic condi- tions, it is probably safe to predict that the Foxo transcription factors are involved in muscle wasting seen in those conditions as well. It should be noted that Foxo tran- scription factors do not regulate the catabolic response only in skeletal muscle but may also activate an atrogene transcriptional program in cardiomyocytes [43].

p300/HAT Expression and Activity Regulate Protein Degradation in Muscle Cells

In recent years, it has become increasingly clear that in addition to transcription fac- tors, so called nuclear cofactors participate in the regulation of gene activation [44].

Among nuclear cofactors, p300 has attracted much attention [45]. p300 exerts some of its effects through its HAT activity. Although it was initially believed that the major mechanism by which HAT activity regulates gene activation was by increasing the acetylation of histones, disrupting chromatin and enhancing the accessibility of transcription factors to their DNA binding sites, there is evidence that p300 acety- lates other proteins as well, including transcription factors and other nuclear cofac- tors. Another important mechanism by which nuclear cofactors influence transcrip- tional activity is protein-protein interactions with transcription factors, nuclear cofactors, and other nuclear proteins that are components of the basal transcription machinery. The interaction with other proteins can result in the acetylation of those proteins but may also serve as a mechanism by which other proteins are recruited for regulation of gene transcription.

It should be noted that protein acetylation is determined not only by HAT activity but by histone deacetylase (HDAC) activity as well [46, 47], and the degree of acety- lation is the result of the balance between ongoing acetylation and deacetylation of any given protein (analogous to the phosphorylation of proteins that is regulated by the balance between kinase and phosphatase activities).

Recent studies in our laboratory have focused on the potential role of p300 in the development of muscle wasting. In several of those studies we used glucocorticoid- treated cultured L6 myotubes, a rat skeletal muscle cell line. Glucocorticoid-treated myotubes have been used in a large number of studies, in our and other laborato- ries, as an in vitro model of muscle wasting characterized by increased ubiquitin- proteaosme-dependent proteolysis and reflecting the important role of glucocorti-

(6)

coids as a mediator of muscle wasting [36, 37, 40, 41]. In initial experiments, we found that p300 protein and mRNA levels were increased in a time- and dose-depen- dent manner in dexamethasone-treated myotubes [21]. In the same study, co-immu- noprecipitation experiments provided evidence for a protein-protein interaction between p300 and C/EBPq . Because there was no interaction between p300 and C/

EBP· , it is possible that the interaction with C/EBP q is specific for this transcription factor among the C/EBP family members. In more recent experiments, we have found evidence that C/EBPq is hyperacetylated in dexamethasone-treated myotubes (unpublished observations), suggesting that the p300-C/EBPq interaction may influ- ence gene transcription at least in part by hyperacetylation.

In subsequent experiments, we observed that p300-associated HAT activity was increased and HDAC3 and 6 expression and activity were reduced after treatment of the myotubes with dexamethasone [22]. In the same study, treatment of the myotu- bes with the HDAC inhibitor, trichostatin A (TSA), resulted in increased protein degradation, further supporting the role of hyperacetylation as a regulator of muscle protein breakdown. Interestingly, the increase in protein degradation caused by TSA was similar to the increase caused by dexamethasone and the combined treatment of the myotubes with dexamethasone and TSA gave rise to the same stimulation of proteolysis as was noticed after treatment with either drug alone, suggesting (but not proving) that they share a common mechanism in their actions.

Additional experiments in the same study [22] provided strong genetic evidence for a role of p300/HAT activity in the regulation of glucocorticoid-induced muscle protein degradation. In those experiments, silencing of p300 expression using a small interfering RNA (siRNA) technique blocked the dexamethasone-induced increase in protein degradation and a similar inhibition of dexamethasone-induced proteolysis was seen when muscle cells were transfected with a plasmid expressing p300 that had been mutated in its HAT activity domain and therefore lacked HAT activity.

Thus, our recent studies provide evidence for a role of hyperacetylation, mediated by increased HAT and decreased HDAC3 and 6 activities, in the regulation of gluco- corticoid-induced muscle proteolysis. It will be important in future experiments to test whether similar mechanisms are involved in the in vivo regulation of muscle wasting seen in catabolic conditions such as sepsis, severe injury, and cancer. It will also be important to determine the exact role of C/EBPq acetylation in the develop- ment of muscle atrophy and to test whether other transcription factors (or other nuclear proteins) are acetylated in catabolic muscle. In that respect, it is interesting to note that studies suggest that the activities of both Foxo transcription factors [48]

and NF-κB [30] may be regulated by acetylation/deacetylation. For example, recent studies have provided evidence that the NF-κB p65 subunit is acetylated by p300 after its transport into the nucleus. This acetylation, which occurs on lysine residues 218, 221, and 310, results in increased NF-κB DNA binding and upregulated tran- scription of NF-κB target genes [30]. p65 is subsequently deacetylated by HDACs, in particular HDAC3, which facilitates the binding of p65 to the inhibitory IκB result- ing in nuclear export of p65 and inhibition of NF-κB activity. Thus, the acetylation and deacetylation of p65 seem to act as an important molecular switch regulating NF-κB activity. Ongoing studies in our laboratories are designed to test whether acetylation of p65 (in addition to acetylation of C/EBPq ) plays a role in sepsis- and glucocorticoid-induced muscle wasting. It should be noticed that in other recent experiments we did not find evidence for acetylation of Foxo1 or 3a in dexametha- sone-treated myotubes (unpublished observations) suggesting that transcription fac- tors involved in muscle wasting may be differentially regulated by p300/HAT activity.

(7)

Fig 1. Schematic illustration of the role of tran- scription factors and nuclear cofactors in the development of glucocorticoid- and cytokine-regu- lated muscle wasting in sepsis, severe injury and cancer. Although the roles of CCAAT/enhancer binding protein (C/EBP) transcription factors, nuclear factor-kappa B (NF-κB), forkhead box O (Foxo) 1 and 3a, p300, and histone deacetylases (HDACs) are discussed in this chapter, it is likely that other transcription factors and nuclear cofac- tors also participate in the regulation of protein degradation in atrophying muscle.

Conclusion

Muscle wasting in a number of catabolic conditions, such as sepsis, severe injury, and cancer, is associated with increased transcription of multiple genes, including (but not limited to) genes in the ubiquitin-proteasome proteolyitc pathway. There is increasing evidence that the expression and activity of transcription factors and nuclear cofactors play an important role in the regulation of muscle wasting-associ- ated genes. Among transcription factors, C/EBPq and · , NF-κB, and Foxo1 and 3a seem to be particularly important for the development of muscle atrophy. Recent studies suggest that the expression and HAT activity of the nuclear cofactor, p300, are essential for the regulation of muscle protein breakdown, possibly by acetylating C/EBPq and the NF-κB p65 subunit. The roles of transcription factors and nuclear cofactors in the regulation of muscle proteolysis and the development of muscle wasting are summarized in Fig 1. An increased understanding of the molecular reg- ulation of catabolic events in skeletal muscle is important for the development of novel and targeted therapeutic strategies for the care of patients with muscle wast- ing.

Acknowledgment: Studies performed in the author’s laboratory and described in this chapter were supported in part by NIH grants R01 DK37908 and R01 NR8545.

References

1. Hasselgren PO, Fischer JE (2001) Muscle cachexia: Current concepts of intracellular mecha- nisms and molecular regulation. Ann Surg 233:9 – 17

2. Lecker SH, Solomon V, Mitch WE, Goldberg AL (1999) Muscle protein breakdown and the critical role of the ubiquitin-proteasome pathway in normal and disease states. J Nutr 129 (Suppl):227S-237S

(8)

3. Hasselgren PO, Wray C, Mammen J (2002) Molecular regulation of muscle cachexia: It may be more than the proteasome. Biochem Biophys Res Commun 290:1 – 10

4. Hasselgren PO (1999) Glucocorticoids and muscle catabolism. Curr Opin Clin Nutr Metab Care 2:201 – 205

5. Tracey KJ, Wei H, Manogue KR, et al (1998) Cachectin/tumor necrosis factor induces cachexia, anemia, and inflammation. J Exp Med 167:1211 – 1227

6. Zamir O, Hasselgren PO, Kunkel SL, Frederick JA, Higashiguchi T, Fischer JE (1992) Evidence that tumor necrosis factor participates in the regulation of muscle proteolysis during sepsis.

Arch Surg 127:170 – 174

7. Reisz-Porszasz S, Bhasin S, Artaza JN, et al (2003) Lower skeletal muscle mass in male trans- genic mice with muscle-specific overexpression of myostatin. Am J Physiol 285:E876-E888 8. Reid WP, MacGowan NA (1998) Respiratory muscle injury in animal models and humans.

Mol Cell Biochem 179:63 – 80

9. Andreyev HJ, Norman AR, et al (1998) Why do patients with weight loss have a worse out- come when undergoing chemotherapy for gastrointestinal malignancies? Eur J Cancer 34:503 – 509

10. Skipworth RJ, Stewart GD, Ross JA, Guttridge DC, Fearon KC (2006) The molecular mecha- nisms of skeletal muscle wasting: implications for therapy. Surgeon 4:273 – 283

11. Tiao G, Fagan JM, Samuels N, et al (1994) Sepsis stimulates nonlysosomal, energy-dependent proteolysis and increases ubiquitin mRNA levels in rats skeletal muscle. J Clin Invest 94:2255 – 2264

12. Hobler SC, Williams AB, Fischer D, et al (1999) The activity and expression of the 20S protea- some are increased in skeletal muscle during sepsis. Am J Physiol 277:R434-R440

13. Fang CH, Tiao G, James JH, Ogle CK, Fischer JE, Hasselgren PO (1995) Burn injury stimu- lates multiple proteolytic pathways in skeletal muscle, including the ubiquitin-energy-depen- dent pathway. J Am Coll Surg 180:161 – 170

14. Tiao G, Hobler S, Wang JJ, et al (1997) Sepsis is associated with increased mRNAs of the ubi- quitin-proteasome proteolytic pathway in human skeletal muscle. J Clin Invest 99:163 – 168 15. Williams A, Sun X, Fischer JE, Hasselgren PO (1999) The expression of genes in the ubiqui-

tin-proteasome proteolytic pathway is increased in skeletal muscle from patients with cancer.

Surgery 126:744 – 750

16. Bailey JL, Wang X, England BK, Price SR, Ding X, Mitch WE (1996) The acidosis of chronic renal failure activates muscle proteolysis in rats by augmenting transcription of genes encod- ing proteins of the ATP-dependent ubiquitin-proteasome pathway. J Clin Invest 97:1447 – 1453 17. Lecker SH, Jagoe RT, Gilbert A, et al (2004) Multiple types of skeletal muscle atrophy involve

a common program of changes in gene expression. FASEB J 18:39 – 51

18. Gomes MD, Lecker SH, Jagoe RT, Navon A, Goldberg AL (2001) Atrogin-1, a muscle specific F- box protein highly expressed during muscle atrophy. Proc Natl Acad Sci USA 98:14440 – 14445 19. Bodine SC, Latres E, Baumheuter S, et al (2001) Identification of ubiquitin ligases required

for skeletal muscle atrophy. Science 294:1704 – 1708

20. Wray CJ, Mammen JM, Hershko DD, Hasselggren PO (2003) Sepsis upregulates the gene expression of multiple ubiquitin ligases in skeletal muscle. Int J Biochem Cell Biol 35:698 – 705

21. Yang H, Menconi M, Wei W, Petkova V, Hasselgren PO (2005) Dexamethasone upregulates the expression of the nuclear cofactor p300 and its interaction with C/EBPq in cultured myo- tubes. J Cell Biochem 94:1058 – 1067

22. Yang H, Wei W, Menconi M, Hasselgren PO (2007) Dexamethasone-induced protein degrada- tion in cultured myotubes is p300/HAT-dependent. Am J Physiol (in press)

23. Lekstrom-Himes J, Xanthopoulos KG (1998) Biological role of the CCAAT/enhancer-binding family of transcription factors. J Biol Chem 273:28545 – 28548

24. Poli V (1998) The role of C/EBP isoforms in the control of inflammatory and and native immune functions. J Biol Chem 273:29279 – 29282

25. Penner G, Gang G, Sun X, Wray C, Hasselgren PO (2002) C/EBP DNA-binding activity is uoregulated by a glucocorticoid-dependent mechanism in septic muscle. Am J Physiol 282:R439-R444

26. Yang H, Mammen J, Wei W, et al (2005) Expression and activity of C/EBPq and · are upregu- lated by dexamethasone in skeletal muscle. J Cell Phsyiol 204:219 – 226

(9)

27. Wagatsuma A (2006) Upregulation of genes encoding adipogenic transcriptional factors C/

EBP[ and PPAR * 2 in denervated muscle. Exp Physiol 91:747–753

28. Hu E, Tontonoz P, Spiegelman BM (1995) Transdifferentiation of myoblasts by the adipogenic factors PPAR* and C/EBP [ . Proc Natl Acad Sci USA 92:9856–9860

29. Ghosh S, May MJ, Kopp EB (1998) NF-kappaB and Rel proteins: evolutionarily conserved mediators of immune responses. Annu Rev Immunol 16:225 – 260

30. Chen LF, Greene WC (2003) Regulation of distinct biological activities of the NF-kB tran- scription factor complex by acetylation. J Mol Med 81:549 – 557

31. Penner CG, Gang G, Wray C, Fischer JE, Hasselgren PO (2001) The transcription factors NF- kB and AP-1 are differentially regulated in skeletal muscle during sepsis. Bioochem Biophys Res Commun 281:1331 – 1336

32. Ladner KJ, Caligiuri MA, Guttridge DC (2003) Tumor necrosis factor-regulated biphasic acti- vation of NF-kB is required for cytokine-induced loss of skeletal muscle gene products. J Biol Chem 278:2294 – 2303

33. Li YP, Reid MB (2000) NF-kB mediates the protein loss induced by TNF-[ in differentiated skeletal muscle myotubes. Am J Physiol 279:R1165-R1170

34. Luo GJ, Hershko DD, Robb BW, Wray CJ, Hasselgren PO (2003) IL-1q stimulates IL-6 produc- tion in cultured skeletal muscle cells through activation of MAP kinase signaling pathway and NFkB. Am J Physiol 284:R1249-R1254

35. Luo GJ, Sun X, Hungness E, Hasselgren PO (2001) Heat shock protects L6 myotubes from cat- abolic effects of dexamethasone and prevents downregulation of NF-kB. Am J Physiol 281:R1193-R1200

36. Hong DH, Forsberg NE (1995) Effects of dexamethasone on protein degradation and protease gene expression in rat L8 myotube cultures. Mol Cell Endocrinol 108:199 – 209

37. Wang L, Luo GJ, Wang JJ, Hasselgren PO (1998) Dexamethasone stimulates proteasome- and calcium-dependent proteolysis in cultured L6 myotubes. Shock 10:298 – 306

38. Cai D, Frantz JD, Tawa NE, et al (2004) IKKq /NF-kB activation causes severe muscle wasting in mice. Cell 119:285 – 298

39. Du J, Mitch WE, Wang X, Price SR (2000) Glucocorticoids induce proteasome C3 subunit expression in L6 muscle cells by opposing the suppression of its transcription by NF-kappa B. J Biol Chem 275:19661 – 19666

40. Sandri M, Sandri C, Gilbert A, et al (2004) Foxo transcription factors induce the atrophy- related ubiquitin-ligase atrogin-1 and cause skeletal muscle atrophy. Cell 117:399 – 412 41. Stitt TN, Drijan D, Clarke BA, et al (2004) The IGF-1/PI3K/Akt pathway prevents expression

of muscle atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors. Mol Cell 14:395 – 403

42. Kandarian SC, Jackman RW (2006) Intracellular signaling during skeletal muscle atrophy.

Muscle Nerve 33:155 – 165

43. Skurk C, Izumiya Y, Maatz H, et al (2005) The FOXO3a transcription factor regulates cardiac myocyte size downstream of AKT signaling. J Biol Chem 280:20814 – 20823

44. Janknecht R, Hunter T (1996) Transcription: a growing coactivator network. Nature 383:22 – 23

45. Polesskaya A, Naguibneva I, Fritsch L, et al (2001) CBP/p300 and muscle differentiation: no HAT, no muscle. EMBO J 20:6816 – 6825

46. DeRuijter AJM, van Gennip AH, Caron HN, Kemp S, Kuilenburg ABP (2003) Histone deace- tylases (HDACs): characterization of the classical HDAC family. Biochem J 370:737 – 749 47. Kuo MH, Allis CD (1998) Roles of histone acetyltransferases and deacetylases in gene regula-

tion. BioEssays 20:615 – 626

48. Van der Heide LP, Smidt MP (2005) Regulation of FoxO activity by CBP/p300-mediated acety- lation. TRENDS Biochem Sci 30:81 – 86

Riferimenti

Documenti correlati

Ten years ago a precursor study in an animal cancer model was published in the European Journal of Translation Myology (back then, Basic and Applied Myology), highlighting that

17,18 In addition, cisplatin treatment of healthy mice, which was observed to cause equivalent reduction of muscle mass as tumor- bearing mice (Figure 1), showed no

The main finding of this study is that neural agrin, a nerve derived factor, boosts the in vitro proliferative performance of human myoblasts from elderly subjects. Our

In questo repertorio si assiste invece, tra gli anni Settanta e Ottanta dell’Ottocento, all’affermazione di un principio compositivo a scapito di un altro, oltretutto con

Auditorium Loyola Room Marinetti Room Trilussa Room Belli Room Ungaretti Room Verga Room Saba Room Quasimodo. COLLOQUIUM: Multimodality 2.0: New theories, new methods,

(1998) Crystal structure of a vertebrate smooth muscle myosin motor domain and its complex with the essential light chain: visualization of the pre-power stroke state..

MVC force, and 2) compare muscle activation patterns and differences during each frequency, when additional voluntary activation was superimposed over ES-induced 25%MVC

into the mitochondrial matrix is able to stimulate the respiratory chain; b) we have recently discovered that, in adult skeletal muscle fibers, mitochondria and CRUs are