Sarcopenia Age-Related Muscle Wasting and Weakness: Mechanisms and Treatments P21 docx

10 212 0
Sarcopenia Age-Related Muscle Wasting and Weakness: Mechanisms and Treatments P21 docx

Đang tải... (xem toàn văn)

Thông tin tài liệu

186 S.E. Alway and P.M. Siu Karin 2009). However, NF-kB can also promote apoptosis when activated by pro-apoptotic proteins including p53, Fas and Fas ligand (Burstein and Duckett 2003; Dutta et al. 2006; Fan et al. 2008). p53 upregulated modulator of apoptosis (PUMA) is a downstream target of p53 and a BH3-only Bcl-2 family member(Lee et al. 2009; Chipuk and Green 2009; Ghosh et al. 2009b). It is induced by p53 following exposure to DNA- damaging agents, such as gamma-irradiation and commonly used chemothera- peutic drugs or oxidative stress (Lee et al. 2009; Chipuk and Green 2009; Ghosh et al. 2009a). It is also activated by a variety of nongenotoxic stimuli indepen- dent of p53, such as serum starvation, kinase inhibitors, glucocorticoids, endo- plasmic reticulum stress, and ischemia/reperfusion (Nickson et al. 2007; Yu and Zhang 2008). The pro-apoptotic function of PUMA is mediated by its interac- tions with anti-apoptotic Bcl-2 family members such as Bcl-2 and Bcl-XL which lead to Bax/Bak-dependent mitochondrial dysfunction mitochondria permeabil- ity and caspase activation (Chipuk and Green 2009). In addition, PUMA is directly activated by NF-kB and contributes to TNF-a-induced apoptosis (Wang et al. 2009). Fig. 4 The extrinsic (death receptor) pathway is activated in aging and contributes to sarcopenia. A ligand (e.g., TNF-a) binds to the death receptor and TNFR1, activates procaspase 8 and caspase 8 which in turn activates caspase 3 and DNA fragmentation 187Nuclear Apoptosis and Sarcopenia Based on the well-documented increase in circulating TNF-a levels with aging (Bruunsgaard et al. 1999, 2001, 2003a, b; Bruunsgaard 2002; Visser et al. 2002; Pedersen et al. 2003; Sandmand et al. 2003; Schaap et al. 2006, 2009) and increases in apoptosis of myonuclei in aged skeletal muscles (Allen et al. 1997; Siu et al. 2005c; Pistilli et al. 2006b), we examined whether apoptotic signalling via the extrinsic pathway contributed to sarcopenia. Our data show that pro- and anti- apoptotic proteins in the extrinsic apoptotic pathway are affected by aging in fast (plantaris) and slow (soleus) skeletal muscles of rats (Pistilli et al. 2006b). Similarly, Marzetti et al. (2009a, b) report elevated TNF-a and TNF-receptor 1 in muscles of old rodents. Together, these data suggest that TNF-a mediated signalling may be an important element triggering the extrinsic apoptotic pathway in and leading to sarcopenia in aging muscles. Muscles from aged rats are significantly smaller and exhibit a larger incidence in fragmented DNA. This suggests that there is a higher level of nuclear apoptosis in muscles from aged animals. In addition, muscles from aged rodents have higher TNFR and FADD mRNA content (measured by semi-quantitative RT-PCR) and protein contents for FADD, Bid, and FLIP, and enzymatic activities of caspase 8 and caspase 3, when compared to muscles from young adult rodents. Although there is an increase in mRNA expression for the TNFR as measured by the semi- quantitative approach, the protein content for the TNFR remains unchanged (Pistilli et al. 2006a, b). This may be explained by the fact that the TNFR antibody utilized in western immunoblots recognizes the soluble form of the receptor. Thus, the changes in the membrane bound form of the receptor, measured by PCR, and the amount of the soluble TNFR may not be equivalent. While fast contracting muscles are generally more susceptible to apoptosis and sarcopenic muscle loss, the pro- apoptotic changes have been reported to be expressed in a similar fashion in both plantaris and soleus muscles; however strong relationships were observed between markers of apoptosis and muscle loss in the fast plantaris muscle that were not observed in the soleus (Pistilli et al. 2006a). These data extend the previous dem- onstration that type II fibres are preferentially affected by aging and suggest that type II fibre containing skeletal muscles may be more susceptible to muscle mass loses via the extrinsic apoptotic pathway (Pistilli et al. 2006b). We have found activation of the extrinsic apoptotic signalling pathway in muscles of old rats (Pistilli et al. 2006a, 2007; Siu et al. 2008), and therefore we speculate that circulating TNF-a may be the initiator of this pathway in skeletal muscle. Nevertheless, we cannot rule out the possibility that other pathways that we did not examine may have been activated by circulating TNF-a in aging muscle. For exam- ple, TNF-a has been shown to directly promote protein degradation (Garcia- Martinez, et al. 1993a, b; Llovera et al. 1997, 1998) and apoptosis within skeletal muscle (Carbo et al. 2002; Figueras et al. 2005). Furthermore, intravenous injection of recombinant TNF-a increases protein degradation in rat skeletal muscles and this is associated with the increased activity of the ubiquitin-dependent proteolytic path- way (Garcia-Martinez et al. 1993a, 1995; Llovera et al. 1997, 1998). In addition, elevated TNF-a concentrations in cell culture for 24–48 h increases apoptosis in skeletal myoblasts as determined by DNA fragmentation (Meadows et al. 2000; 188 S.E. Alway and P.M. Siu Foulstone et al. 2001). A reduction of procaspase 8 occurs within 6h of incubating myoblasts in vitro with recombinant TNF-a, suggesting a TNF-a mediated cleavage and activation of this initiator caspase in myoblast cultures (Stewart et al. 2004). Lees and co-workers (Lees et al. 2009) have recently shown that satellite cells (i.e., MPCs) isolated from hindlimb muscles of old rats have increased TNF-a- induced nuclear factor-kappa B (NF-kB) activation and expression of mRNA levels for TRAF2 and the cell death-inducing receptor, Fas (CD95), in response to pro- longed (24 h) TNF-a treatment compared to in MPCs isolated from muscles of young animals. These findings suggest that age-related differences may exist in the regulatory mechanisms responsible for NF-kB inactivation, which may in turn have an effect on TNF-a-induced apoptotic signalling. Systemic and muscle levels of TNF-a increase with aging, and this should have an even more profound increase in activation of apoptotic gene targets through the extrinsic pathway, as compared to MPCs in muscles of young adult rats (Krajnak et al. 2006; Lees et al. 2009). The effects of TNF-a on apoptosis are not limited to in vitro conditions, because a systemic elevation of TNF-a in vivo increases DNA fragmentation within rodent skeletal muscle (Carbo et al. 2002). Based on the observation that TNF-a mRNA was not different between muscles from young adult and aged rats, it is reasonable to assume that muscle-derived TNF-a does not act in an autocrine manner to stimulate the pro-apoptotic signalling observed in this study. Data from Pistilli and co-workers (Pistilli et al. 2006b) are consistent with the hypothesis that the well- documented systemic elevation of TNF-a with age, may increase the likelihood of ligand binding to the TNFR and stimulate apoptotic signalling of the extrinsic pathway downstream of the TNFR and contribute to sarcopenia in skeletal muscle of old rats. 5.2 Cross-talk Between Extrinsic and Intrinsic Apoptotic Signalling Cross-talk between extrinsic and intrinsic apoptotic pathways was recently reviewed (Sprick and Walczak 2004). Cross-talk between these pathways is the result of the cleavage of the pro-apoptotic BCL-2 family member Bid. Cleaved and activated caspase 8 cannot only serve to activate caspase 3, which is the execu- tioner caspase, but also cleave full-length Bid into a truncated version (tBid) (Tang et al. 2000). tBid then interacts with pro-apoptotic Bax, to stimulate apoptotic sig- nalling from the mitochondria (Grinberg et al. 2005). As has been previously shown, apoptotic signalling from the mitochondria stimulates cleavage of procas- pase 9, which then serves to activate caspase 3 (Johnson and Jarvis 2004). Thus, both the extrinsic and intrinsic apoptotic pathways converge on caspase 3, which then fully engages pro-apoptotic signalling. Skeletal muscles from aged rodents contained a greater protein expression of full-length Bid, which raises the possibil- ity that cross talk between the extrinsic pathway and the intrinsic pathway may occur in aged skeletal muscles (Fig. 5). 189Nuclear Apoptosis and Sarcopenia 6 Exercise Modulation of Apoptosis in Sarcopenia Various perturbations have been used to determine if aging increases the sensitivity of skeletal muscle to apoptosis and apoptosis signalling cascades. These include increases in muscle loading, loading followed by a period of unloading, disuse, denervation or muscle unloading, and aerobic exercise. 6.1 Interventions by Muscle Loading The evidence presented above indicates that mitochondrial dysfunction is a major contributing factor to the path physiology of aging including sarcopenia. While muscle disuse decreases mitochondria function leading to apoptosis (Adhihetty et al. 2003; Siu and Alway 2005a; Bourdel-Marchasson et al. 2007), chronic exer- cise improves mitochondria function (Daussin et al. 2008; Lanza et al. 2008) and reduces apoptotic signalling (Siu et al. 2004). Fig. 5 The potential cross talk between the extrinsic and intrinsic apoptotic signalling pathways are shown 190 S.E. Alway and P.M. Siu Adaptation to chronic loading has been shown to improve anti-apoptotic proteins in skeletal muscle including XIAP (Siu et al. 2005d), Bcl2 (Song et al. 2006), and reduce DNA fragmentation (Siu and Alway 2006a) (Song et al. 2006) and lower pro-apoptotic proteins including Bax (Song et al. 2006), ARC (Siu and Alway 2006a), AIF (Siu and Alway 2006a). In contrast, models of muscle unloading show most of the appositive apoptotic signalling such as elevations in Bax, Apaf1, AIF (Pistilli et al. 2006b), cyto- solic levels of Id2 and p53 (Siu et al. 2006) and the Bax/Bcl2 ratio (Song et al. 2006). Reduced levels of pro-apoptotic proteins may provide one mechanism to explain the improvements in muscle mass and force that are observed in humans after a period of resistance exercise. Our lab (Roman et al. 1993; Ferketich et al. 1998) and others (Charette et al. 1991; Welle et al. 1995; Parise and Yarasheski 2000; Deschenes and Kraemer 2002; Mayhew et al. 2009) have shown that resistance exercise is an effective tool to reduce but not eliminate sarcopenia in aging humans. Although aging has gen- erally been shown to attenuate the absolute extent of muscle adaptations that are pos- sible with increased loading (Alway et al. 2002a; Degens and Alway 2003; Degens 2007; Degens et al. 2007), it is not known how much of this might be the result of increased nuclear apoptosis in skeletal muscle. Interestingly, several studies have reported unexpected improvements in mitochondrial function in both young adult and aged subjects as a result of resistance exercise training. For example, the mitochondrial capacity for ATP synthesis increases after resistance training (Jubrias et al. 2001; Conley et al. 2007b; Tarnopolsky 2009). Resistance exercise also increases antioxidant enzymes and decreases oxidative stress (Parise et al. 2005; Johnston et al. 2008). Furthermore, 26 weeks of whole body resistance exercise was shown to reverse the gene expression of mitochondrial proteins that were associated with normal aging, to that observed in young subjects (Melov et al. 2007). Although we have found that resistance training did not increase the relative volume of mitochondria in muscle fibres of young adults, resistance exercise stimulated mitochondria biogenesis to main- tain the myofibrillar to mitochondria volume (Alway et al. 1989; Alway 1991). In addition, aging attenuates the adaptive response to improve the muscle’s ability to buf- fer pro-oxidants in response to chronic muscle loading (Ryan et al. 2008). Nevertheless, there is some improvement in antioxidant enzymes and the ability to buffer oxidative stress in response to loading conditions (Ryan et al. 2008). Therefore, it is possible that, resistance training could also improve mitochondria function and stimulate mito- chondrial biogenesis in aged individuals. If muscle loading improves not only antioxi- dant enzymes levels but it also reduces Bax accumulation in mitochondria, we would expect that apoptosis signalling should be decreased. This would lead to improved muscle recovery following disuse and reduce sarcopenia. 6.2 Apoptotic Elimination of MPCs Reduces Muscle Hypertrophic Adaptation to Loading It is thought that myonuclei maintain a constant cytoplasm to nuclei ratio, (i.e. “nuclear domain”, see Fig. 1), and that hypertrophy requires that fibres add new 191Nuclear Apoptosis and Sarcopenia nuclei (Schultz 1989, 1996; Schultz and McCormick 1994). Because myonuclei are post mitotic (Schultz 1989, 1996; Schultz and McCormick 1994), satellite cells/ MPCs provide the only important source for adding new nuclei to initiate muscle regeneration, muscle hypertrophy, and postnatal muscle growth in muscles of both young and aged animals (Rosenblatt et al. 1994; Phelan and Gonyea 1997; McCall et al. 1998; Allen et al. 1999; Hawke and Garry 2001; Adams et al. 2002). MPCs are critical for muscle growth because muscle hypertrophy is markedly reduced or eliminated completely after irradiation to prevent MPC activation (Rosenblatt et al. 1994; Hawke and Garry 2001). Growth of adult skeletal muscle requires activation and differentiation of satellite cells/MPCs and increased protein synthesis and accu- mulation of proteins, and this necessitates increased transcription of muscle genes (Dirks and Leeuwenburgh 2002; Pollack et al. 2002; Alway et al. 2002b; Leeuwenburgh 2003; Dirks and Leeuwenburgh 2004; Siu et al. 2005c). Thus, there is little doubt that MPC activation and differentiation are critical components in determining muscle adaptation and growth. If MPCs are activated normally, but they either do not differentiate or do not survive to participate in increased protein synthesis, then muscle adaptation would be compromised. Elevation of apoptosis (lower MPC survival) in muscles from aged animals (Renault et al. 2002; Siu et al. 2005c) could explain the poorer adapta- tion to repetitive loading in aging. We have shown that the most recently activated satellite cells/MPCs during loading are also the most susceptible to apoptosis (Pollack et al. 2002; Alway et al. 2002a, b; Leeuwenburgh 2003; Dirks and Leeuwenburgh 2004). Based on these data, we hypothesize that MPC contribution to chronic loading-induced adaptation (hypertrophy) is lower in muscles of old animals because apoptosis is higher (Degens and Alway 2003), and fewer MPCs survive to contribute to muscle adaptation (Chakravarthy et al. 2001). 6.3 Regulation of Apoptotic Signalling by Aerobic Exercise Although acute endurance exercise has been shown to increase apoptotic signalling under some conditions including dystrophies and other pathologies (Sandri et al. 1997; Podhorska-Okolow et al. 1998, 1999) long-term adaptation to endurance exercise has been shown to lower mitochondria-associated apoptosis in heart and skeletal muscle of rodents (Siu et al. 2004; Kwak et al. 2006; Song et al. 2006; Peterson et al. 2008); however, it does not improve muscle mass or act as a coun- termeasure to sarcopenia (Alway et al. 1996; Marzetti et al. 2008a). This might be in part due to aerobically-induced pathways that are generally inhibitory to muscle growth (e.g., AMPK). Apoptosis has been shown to occur in cardiac (Dalla et al. 2001; Hu et al. 2008; Molina et al. 2009) and skeletal muscles (Dalla et al. 2001; Vescovo and Dalla 2006; Libera et al. 2009) of experimental models of chronic heart failure. Apoptosis in skeletal muscle has been linked to elevated circulating levels of TNF-a (Adams et al. 1999; Vescovo et al. 2000). Although nuclear apoptosis has been detected in 192 S.E. Alway and P.M. Siu muscles of humans with severe chronic heart failure (Conraads et al. 2009), it does not appear to be a large component of muscle loss associated when the disease is less severe (Dirks and Jones 2006; Yu et al. 2009a). Complicating the treatment of heart failure and related cardiovascular diseases is the likelihood that drugs includ- ing statins which are routinely prescribed to reduce hypercholesterolemia, may themselves have a pro-apoptotic role in skeletal muscle (Adams et al. 2008). Such increases in apoptosis are likely to have devastating effects when statins are combined with sarcopenia, where muscle loss is already high. Although aerobic exercise appears to reduce several skeletal muscle problems of persons suffering from severe chronic heart failure (Linke et al. 2005) and an exercise-induced improvement in antioxidant enzymes is correlated to reduced apoptosis in muscles of patients with chronic heart failure (Siu et al. 2004, 2005a; Song et al. 2006), currently there are no data to definitively address if aerobic exercise reduces apop- tosis in heart failure patients. The role or aerobic exercise on nuclear apoptosis of skeletal muscle has not been well-studied but limited data suggest that apoptosis signalling is reduced by aerobic exercise in cardiac and skeletal muscle of young, diseased and aged animals (Siu et al. 2004; Kwak et al. 2006; Song et al. 2006; Peterson et al. 2008; Marzetti et al. 2008a, b). 7 Summary and Conclusions Sarcopenia involves complex of several cellular mechanisms which together con- tribute to muscle loss during aging. Among them, nuclear apoptosis has recently emerged as an important factor involved in the pathophysiology of sarcopenia. Several lines of evidence support the hypothesis that mitochondrial (intrinsic), extrinsic (death receptor) and endoplasmic reticulum-calcium stress activated apop- totic signalling, occurs in skeletal muscles of old mammals. Nevertheless, it has not been determined to what extent sarcopenia would be reduced, if apoptotic signal- ling could be fully blocked. Although there is evidence that reducing Bax markedly reduces apoptosis associated muscle loss with denervation (Siu and Alway 2006b), it is not known if this is also the case with aging. We cannot rule the possibility that the apoptotic signalling events may occur to simply eliminate dysfunctional nuclei and/or damaged muscle fibres, whose perseverance would be detrimental for organ function. Even though a cause and effect relationship between apoptosis and sarcopenia has not been unequivocally determined, evidence that muscle loss is reduced in Bax null mice (Siu and Alway 2006b), and experimental interventions to accelerate muscle loss in aged animals also elevates apoptosis (Siu and Alway 2005a; Siu et al. 2005b, c, d, 2006, 2008; Pistilli et al. 2007) strongly suggests that a causal relationship likely exists between nuclear apoptosis and muscle loss, and this may also extend to aging associated muscle loss. Furthermore, activation of mitochon- drial apoptotic signalling during the early phases of disuse muscle atrophy (Siu and Alway 2005b; Siu and Alway 2009) suggests that this may exist to balance muscle 193Nuclear Apoptosis and Sarcopenia size and the metabolic or functional needs of the animal. If this is true, nuclear apoptosis may be a fundamentally important mechanism that regulates myonuclei number and, therefore controls the extent of muscle growth (or atrophy) in aging. Apoptotic signalling may be modified by loading and aerobic forms of exercise, but it remains to be seen how effective exercise might be in slowing or preventing apoptosis in sarcopenia. Clearly further research is required to better understand the complex cellular mechanisms underlying muscle atrophy that occurs in sarcopenia, and the importance of apoptosis in this process. Unravelling the regulatory factors in the apoptotic pathways will be a necessary step prior to having the ability to design effective interventions and countermeasures for sarcopenia. References Abmayr, S., Crawford, R. W., Chamberlain, J. S. (2004). Characterization of ARC, apoptosis repressor interacting with CARD, in normal and dystrophin-deficient skeletal muscle. Human Molecular Genetics, 13, 213–221. Acehan, D., Jiang, X., Morgan, D. G., Heuser, J. E., Wang, X., Akey, C. W. (2002). Three- dimensional structure of the apoptosome: implications for assembly, procaspase-9 binding, and activation. Molecular Cell, 9, 423–432. Adams, G. R., Caiozzo, V. J., Haddad, F., Baldwin, K. M. (2002). Cellular and molecular responses to increased skeletal muscle loading after irradiation. American Journal of Physiology. Cell Physiology, 283, C1182–C1195. Adams, V., Jiang, H., Yu, J., Mobius-Winkler, S., Fiehn, E., Linke, A., Weigl, C., Schuler, G., Hambrecht, R. (1999). Apoptosis in skeletal myocytes of patients with chronic heart failure is associated with exercise intolerance. Journal of the American College of Cardiology, 33, 959–965. Adams, V., Doring, C., Schuler, G. (2008). Impact of physical exercise on alterations in the skeletal muscle in patients with chronic heart failure. Frontiers in Bioscience, 13, 302–311. Adhihetty, P. J., Hood, D. A. (2003). Mechanisms of apoptosis in skeletal muscle. Basic and Applied Myology, 13, 171–179. Adhihetty, P. J., Irrcher, I., Joseph, A. M., Ljubicic, V., Hood, D. A. (2003). Plasticity of skeletal muscle mitochondria in response to contractile activity. Experimental Physiology, 88, 99–107. Adhihetty, P. J., Ljubicic, V., Hood, D. A. (2007a). Effect of chronic contractile activity on SS and IMF mitochondrial apoptotic susceptibility in skeletal muscle. American Journal of Physiology. Endocrinology and Metabolism, 292, E748–E755. Adhihetty, P. J., O’Leary, M. F., Chabi, B., Wicks, K. L., Hood, D. A. (2007b). Effect of denerva- tion on mitochondrially mediated apoptosis in skeletal muscle. Journal of Applied Physiology, 102, 1143–1151. Adhihetty, P. J., O’Leary, M. F., Hood, D. A. (2008). Mitochondria in skeletal muscle: adaptable rheostats of apoptotic susceptibility. Exercise and Sport Sciences Reviews, 36, 116–121. Adhihetty, P. J., Uguccioni, G., Leick, L., Hidalgo, J., Pilegaard, H., Hood, D. A. (2009). The role of PGC-1{alpha} on mitochondrial function and apoptotic susceptibility in muscle. American Journal of Physiology. Cell Physiology, 297(1), C217–C225. Allen, D. L., Linderman, J. K., Roy, R. R., Bigbee, A. J., Grindeland, R. E., Mukku, V., Edgerton, V. R. (1997). Apoptosis: a mechanism contributing to remodeling of skeletal muscle in response to hindlimb unweighting. The American Journal of Physiology, 273, C579–C587. 194 S.E. Alway and P.M. Siu Allen, D. L., Roy, R. R., Edgerton, V. R. (1999). Myonuclear domains in muscle adaptation and disease. Muscle & Nerve, 22, 1350–1360. Alway, S. E. (1991). Is fiber mitochondrial volume density a good indicator of muscle fatigability to isometric exercise? Journal of Applied Physiology, 70, 2111–2119. Alway, S. E., Siu, P. M. (2008). Nuclear apoptosis contributes to sarcopenia. Exercise and Sport Sciences Reviews, 36, 51–57. Alway, S. E., MacDougall, J. D., Sale, D. G. (1989). Contractile adaptations in the human triceps surae after isometric exercise. Journal of Applied Physiology, 66, 2725–2732. Alway, S. E., Coggan, A. R., Sproul, M. S., Abduljalil, A. M., Robitaille, P. M. (1996). Muscle torque in young and older untrained and endurance-trained men. The Journals of Gerontology. Series A: Biological Sciences and Medical Sciences, 51, B195–B201. Alway, S. E., Degens, H., Krishnamurthy, G., Smith, C. A. (2002a). Potential role for Id myogenic repressors in apoptosis and attenuation of hypertrophy in muscles of aged rats. American Journal of Physiology. Cell Physiology, 283, C66–C76. Alway, S. E., Degens, H., Lowe, D. A., Krishnamurthy, G. (2002b). Increased myogenic repressor Id mRNA and protein levels in hindlimb muscles of aged rats. American Journal of Physiology: Regulatory, Integrative and Comparative Physiology, 282, R411–R422. Alway, S. E., Degens, H., Krishnamurthy, G., Chaudhrai, A. (2003a). Denervation stimulates apoptosis but not Id2 expression in hindlimb muscles of aged rats. The Journals of Gerontology. Series A: Biological Sciences and Medical Sciences, 58, 687–697. Alway, S. E., Martyn, J. K., Ouyang, J., Chaudhrai, A., Murlasits, Z. S. (2003b). Id2 expression during apoptosis and satellite cell activation in unloaded and loaded quail skeletal muscles. American Journal of Physiology: Regulatory, Integrative and Comparative Physiology, 284, R540–R549. Antonsson, B., Montessuit, S., Lauper, S., Eskes, R., Martinou, J. C. (2000). Bax oligomerization is required for channel-forming activity in liposomes and to trigger cytochrome c release from mitochondria. The Biochemical Journal, 345(Pt 2), 271–278. Attaix, D., Ventadour, S., Codran, A., Bechet, D., Taillandier, D., Combaret, L. (2005). The ubiq- uitin-proteasome system and skeletal muscle wasting. Essays in Biochemistry, 41, 173–186. Basanez, G., Nechushtan, A., Drozhinin, O., Chanturiya, A., Choe, E., Tutt, S., Wood, K. A., Hsu, Y., Zimmerberg J., Youle R.J. (1999). Bax, but not Bcl-xL, decreases the lifetime of planar phos- pholipid bilayer membranes at subnanomolar concentrations. Proceedings of the National Academy of Sciences of the United States of America, 96, 5492–5497. Benayoun, B., Baghdiguian, S., Lajmanovich, A., Bartoli, M., Daniele, N., Gicquel, E., Bourg, N., Raynaud, F., Pasquier, M. A., Suel, L., Lochmuller, H., Lefranc, G., Richard, I. (2008). NF-kappaB-dependent expression of the antiapoptotic factor c-FLIP is regulated by calpain 3, the protein involved in limb-girdle muscular dystrophy type 2A. The FASEB Journal, 22, 1521–1529. Bernardi, P. (1999). Mitochondria in muscle cell death. Italian Journal of Neurological Sciences, 20, 395–400. Bhuiyan, M. S. & Fukunaga, K. (2007). Inhibition of HtrA2/Omi ameliorates heart dysfunction following ischemia/reperfusion injury in rat heart in vivo. European Journal of Pharmacology, 557, 168–177. Blink, E., Maianski, N. A., Alnemri, E. S., Zervos, A. S., Roos, D., Kuijpers, T. W. (2004). Intramitochondrial serine protease activity of Omi/HtrA2 is required for caspase-independent cell death of human neutrophils. Cell Death and Differentiation, 11, 937–939. Bourdel-Marchasson, I., Biran, M., Dehail, P., Traissac, T., Muller, F., Jenn, J., Raffard, G., Franconi, J. M., Thiaudiere, E. (2007). Muscle phosphocreatine post-exercise recovery rate is related to functional evaluation in hospitalized and community-living older people. The Journal of Nutrition, Health & Aging, 11, 215–221. Brack, A. S. & Rando, T. A. (2007). Intrinsic changes and extrinsic influences of myogenic stem cell function during aging. Stem Cell Reviews, 3, 226–237. Brack, A. S., Bildsoe, H., Hughes, S. M. (2005). Evidence that satellite cell decrement contributes to preferential decline in nuclear number from large fibres during murine age-related muscle atrophy. Journal of Cell Science, 118, 4813–4821. 195Nuclear Apoptosis and Sarcopenia Brack, A. S., Conboy, M. J., Roy, S., Lee, M., Kuo, C. J., Keller, C., Rando, T. A. (2007). Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science, 317, 807–810. Bruunsgaard, H. (2002). Effects of tumor necrosis factor-alpha and interleukin-6 in elderly popu- lations. European Cytokine Network, 13, 389–391. Bruunsgaard, H., Andersen-Ranberg, K., Jeune, B., Pedersen, A. N., Skinhoj, P., Pedersen, B. K. (1999). A high plasma concentration of TNF-alpha is associated with dementia in centenari- ans. The Journals of Gerontology. Series A: Biological Sciences and Medical Sciences, 54, M357–M364. Bruunsgaard, H., Pedersen, M., Pedersen, B. K. (2001). Aging and proinflammatory cytokines. Current Opinion in Hematology, 8, 131–136. Bruunsgaard, H., Ladelund, S., Pedersen, A. N., Schroll, M., Jorgensen, T., Pedersen, B. K. (2003a). Predicting death from tumour necrosis factor-alpha and interleukin-6 in 80-year-old people. Clinical and Experimental Immunology, 132, 24–31. Bruunsgaard, H., Andersen-Ranberg, K., Hjelmborg, J. B., Pedersen, B. K., Jeune, B. (2003b). Elevated levels of tumor necrosis factor alpha and mortality in centenarians. The American Journal of Medicine, 115, 278–283. Bruusgaard, J. C., Liestol, K., Gundersen, K. (2006). Distribution of myonuclei and microtubules in live muscle fibers of young, middle-aged, and old mice. Journal of Applied Physiology, 100, 2024–2030. Burstein, E. & Duckett, C. S. (2003). Dying for NF-kappaB? Control of cell death by transcrip- tional regulation of the apoptotic machinery. Current Opinion in Cell Biology, 15, 732–737. Cacciapaglia, F., Spadaccio, C., Chello, M., Gigante, A., Coccia, R., Afeltra, A., Amoroso, A. (2009). Apoptotic molecular mechanisms implicated in autoimmune diseases. European Review for Medical and Pharmacological Sciences, 13, 23–40. Campisi, J. & Sedivy, J. (2009). How does proliferative homeostasis change with age? What causes it and how does it contribute to aging? The Journals of Gerontology. Series A: Biological Sciences and Medical Sciences, 64, 164–166. Cande, C., Cecconi, F., Dessen, P., Kroemer, G. (2002a). Apoptosis-inducing factor (AIF): key to the conserved caspase-independent pathways of cell death? Journal of Cell Science, 115, 4727–4734. Cande, C., Cohen, I., Daugas, E., Ravagnan, L., Larochette, N., Zamzami, N., Kroemer, G. (2002b). Apoptosis-inducing factor (AIF): a novel caspase-independent death effector released from mitochondria. Biochimie, 84, 215–222. Carbo, N., Busquets, S., van, R. M., Alvarez, B., Lopez-Soriano, F. J., Argiles, J. M. (2002). TNF- alpha is involved in activating DNA fragmentation in skeletal muscle. British Journal of Cancer, 86, 1012–1016. Cartron, P. F., Moreau, C., Oliver, L., Mayat, E., Meflah, K., Vallette, F. M. (2002). Involvement of the N-terminus of Bax in its intracellular localization and function. FEBS Letters, 512, 95–100. Chabi, B., Ljubicic, V., Menzies, K. J., Huang, J. H., Saleem, A., Hood, D. A. (2008). Mitochondrial function and apoptotic susceptibility in aging skeletal muscle. Aging Cell, 7, 2–12. Chakravarthy, M. V., Spangenburg, E. E., Booth, F. W. (2001). Culture in low levels of oxygen enhances in vitro proliferation potential of satellite cells from old skeletal muscles. Cellular and Molecular Life Sciences, 58, 1150–1158. Chang, H. Y. & Yang, X. (2000). Proteases for cell suicide: functions and regulation of caspases. Microbiology and Molecular Biology Reviews, 64, 821–846. Chao, D. T., Korsmeyer, S. J. (1998). BCL-2 family: regulators of cell death. Annual Review of Immunology, 16, 395–419. Chao, D. T., Linette, G. P., Boise, L. H., White, L. S., Thompson, C. B., Korsmeyer, S. J. (1995). Bcl-XL and Bcl-2 repress a common pathway of cell death. The Journal of Experimental Medicine, 182, 821–828. Charette, S. L., McEvoy, L., Pyka, G., Snow-Harter, C., Guido, D., Wiswell, R. A., Marcus, R. (1991). Muscle hypertrophy response to resistance training in older women. Journal of Applied Physiology, 70, 1912–1916. . Schultz and McCormick 1994), satellite cells/ MPCs provide the only important source for adding new nuclei to initiate muscle regeneration, muscle hypertrophy, and postnatal muscle growth in muscles. activation and differentiation of satellite cells/MPCs and increased protein synthesis and accu- mulation of proteins, and this necessitates increased transcription of muscle genes (Dirks and Leeuwenburgh. phases of disuse muscle atrophy (Siu and Alway 2005b; Siu and Alway 2009) suggests that this may exist to balance muscle 193Nuclear Apoptosis and Sarcopenia size and the metabolic or functional

Ngày đăng: 07/07/2014, 10:20

Tài liệu cùng người dùng

Tài liệu liên quan