Skeletal Muscle and Stress Proteins: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor:

All organisms and cells respond to various stress conditions by up-regulating the expression and/or activation of a group of proteins called heat shock proteins (HSPs). Although their expression is induced by several stimuli, they are commonly recognized as HSPs due to the first experiments showing their increased transcription after application of heat shock. These proteins are molecular chaperones mainly involved in assisting protein transport and folding, assembling multimolecular complexes, and triggering protein degradation by proteasome. In addition, they play a crucial role in gene expression regulation, DNA replication, signal transduction, cell differentiation, apoptosis, cellular senescence or immortalization, and intercellular communications. Heat shock proteins are classified according to their molecular weight in super heavy, 100, 90, 70, 60, 40, and small HSPs. Although they are the most highly conserved, ubiquitous, and abundant proteins in all organisms, their cellular stress response can depend on the class and stimulus.

  • skeletal muscle
  • physical exercise
  • heat shock protein 60

1. Fiber-Types of Skeletal Muscle and Its Adaptation to Physical Exercise

Skeletal muscle is a heterogeneous tissue containing fibers with different morphological, metabolic, and functional properties. Muscle fibers are typically identified by the expression of a multigene family of Myosin Heavy Chains (MHCs). Their sequence variability has been associated with their specific structure and function, but the codified actin-based motor proteins are conserved [1][2]. In the skeletal muscle they are classified as MHC-I, MHC-IIa, MHC-IIx, and MHC-IIb [1][2][3]. The MHC-I, IIa, and IIx fiber-types are expressed to variable degrees in both small animals (mice, rats, and rabbits) and human skeletal muscle. Instead, MHC-IIb fibers are solely expressed in small animals’ skeletal muscle [4]. In mammals, ‘‘hybrid’’ fibers (i.e., type I/IIa, IIa/x, IIx/b) can occur when different MHC transcripts coexist in a single fiber [5][6][7].

Although the structural and functional muscle requests define privileged associations between MHC isoforms, other common fiber-type classifications are used. One of them is related to the contractile response (or speed of contraction) distinguishing between slow or fast muscle fibers. In addition, cellular metabolism has been used as another parameter to distinguish the different types of myofibers with glycolytic metabolism and fibers with a massive mitochondrial presence and prevalent aerobic metabolism. Notably, MHC isoform expression correlates with fiber-type morphology, metabolism, and function [8][9]. In general terms, MHC-I-expressing fibers are small, rich in oxidative enzymes, slow in contraction, and have a greater resistance to fatigue, while MHC-IIb-expressing fibers are large, rich in glycolytic enzymes, and fast in contraction due to the developed sarcoplasmic reticulum that allows the rapid release of calcium ions, and a predominantly anaerobic metabolism [10][11]. Specifically, fiber contraction correlates with myosin ATPase activity with relative velocities of I < IIa < IIx < IIb [12][13]. Furthermore, skeletal muscle can be classified as postural or non-postural according to its function and the percentage of each fiber-type [14]. Although the four main types of skeletal muscle fibers are present in different percentages among the mammal species and within the same species therein, they are represented differently in the various structures of the musculoskeletal system (Table 1) [15][16][17][18]. Moreover, basal mitochondrial content has been shown to vary between species and is fiber-type-specific in mouse, rat, and human skeletal muscles [19].

Table 1. Fiber-type distribution and mitochondrial content in different skeletal muscles.

One of the most surprising characteristics of the myofibers of the skeletal muscle is the high degree of plasticity as an adaptive response to physiological and non-physiological requests [20]. Plasticity is the ability of a tissue to modify its composition by adapting it to changing functional needs. Repeated, prolonged, or simple changes in functional requests can both quantitatively and qualitatively modify muscle tissue. These changes can affect the myofibrillary system, the sarcoplasmic reticulum, the proteins involved in regulating the concentration of intracellular calcium as well as the enzymatic systems involved in energy metabolism. The mechanism of adaptation of the functional requests is based primarily on the transformation of the MHC content, with a shift of the MHC fibers that affects the overall speed of contraction of the muscle. However, it is also possible that an increase in the speed of the same cellular type occurs in the absence of variations of the expressed MHC isoforms [21][22]. The increased contractile activity following physical exercise activates several signal pathways that lead to significant phenotypic changes such as MHC fiber transitions, enhanced mitochondrial biogenesis, and angiogenesis. Changes in the expression pattern of MHC isoforms and in the cross-sectional area (CSA) of the skeletal muscle cells, in response to different training protocols, are related to the changes in strength and power that the muscle undergoes [23]. For these reasons, physical exercise induces muscle hypertrophy that is followed by the upregulation of contractile elements’ synthesis. Literature data have shown the hypertrophic response for all three major types of fibers (MCH-I, IIa, and IIx) following resistance training, both in young subjects and in elderly subjects [24]. However, exercise-induced hypertrophy seems to affect fast muscle fibers more than type I fibers [25]. Exercise can therefore induce changes in the expression of MHCs, thus causing a switch from type IIb to IIx and IIa and, in rare cases, also to type I. In most cases, physiological adaptations to increased activity induce a switch to a more oxidative fast phenotype [3]. Allen et al. showed a significant increase in the percentage of fibers expressing MHC-IIa and a concomitant decrease in the percentage of fibers expressing MHC-IIb in mouse fast muscles after some weeks of wheel exercise [26]. The switch in the range IIb–IIx–IIa in mouse and rat muscle or in the range IIx–IIa in human muscle likely reflects the total amount of activity [27].

Human studies have shown that strength training induces an increase in type IIa and hybrid IIa/x fibers at the expense of fast IIx fibers. At the same time, there is an increase in the CSA in all types of myofibers, indicating a hypertrophic effect [28]. In fact, Kesidis et al. showed that type IIa muscle fibers in human skeletal muscle seem to have an enzymatic profile and a rate of contraction that makes these fibers more suitable for strength performance than fibers containing the MHC I isoform [29]. On the other hand, endurance training seems to induce fast-to-slow fiber transitions (from IIx to IIa, and in rare cases type I), while the CSA values remain unchanged. The physiological advantage is the greater transduction efficiency of the mechanical energy associated with the IIa fibers compared to the IIx fibers. However, the increased proportion of type I fibers could derive from different training protocols, causing a significant turnover and regeneration of fibers, and also including a regeneration of the peripheral nerve [30]. The absence of cellular damage can be explained by the lack of the expression of embryonic myosin isoforms and supports the results of the studies in which no variations in fiber-type I following training protocols were detected [31].

2. Stress Proteins: Heat Shock Protein 60

The HSPD1 gene comprises ~17 kb with 12 exons and it is localized at chromosome locus 2q33.1. This gene encodes a protein of 573aa corresponding to a molecular weight of 61.05 kDa known as HSP60 or Hsp60, also commonly referred to as Cpn60 [32]. Hsp60 belongs to group I of chaperonins [33]. Its ATP-dependent chaperon mechanism was thoroughly investigated for the prokaryotic homolog GroEL. Three structural domains were identified for GroEL: apical, intermediate, and equatorial. To carry out its chaperoning function, GroEL needs to generate a tetradecamer complex with its co-chaperon GroES (the homolog of Hsp10) [34]. Thus, the chaperon complex is made up of GroEL, structured in two rings with seven identical subunits, and GroES, which binds to the apical domains of GroEL to close the cage [35]. The chaperon mechanism is a multistep process that involves the unfolded protein binding to GroEL apical domains. Concomitantly, ATP binds to GroEL’s equatorial domain and its hydrolysis allows the conformational change (from trans to cis) of the GroEL apical and intermediate domains for the substrate encapsulation in the central cavity of the chaperon [36]. Consecutively, the dissociation of the cis-complex and the release of the folded protein, ADP, and GroES occurs [37]. Although the chaperon mechanism of the mammalian mitochondrial Hsp60-Hsp10 complex is similar, the solid-state structure appears as a symmetrical football-shaped complex by X-ray [38]. In humans, the mitochondrial Hsp60 exists as a homo-oligomer of seven subunits in equilibrium with very minor populations of monomers and double-ring tetradecamers [39][40]. In addition, it was demonstrated that Hsp60 single ring could perform chaperonin-mediated folding activity in vivo [41].

Hsp60 is constitutively expressed under philological conditions, so much so that its knockout is incompatible with life [42][43][44]. At the same time, Hsp60 expression is related to numerous etio-pathological conditions [45]. Hsp60 is mainly localized in the mitochondria. The mitochondrial import signal (MIS) at the N-terminus drives Hsp60 from the cytoplasm to the mitochondria [46]. Nevertheless, one third of Hsp60 is localized to the extra-mitochondrial sites, such as cytosol, plasma-cell membrane, inside exosomes, extracellular space, and circulation [47][48]. Inside the mitochondria, Hsp60 guarantees the correct folding of other mitochondrial proteins [49][50] as well as its “unfoldase” activity to stabilize misfolded and aggregated proteins, making provision for the mitochondrial biogenesis and protein homeostasis [51][52]. In addition, the mitochondrial Hsp60 directs the replication and transmission of mitochondrial DNA (mtDNA) [53][54]. Otherwise, the extramitochondrial Hsp60 is involved in intracellular protein trafficking [55] and peptide-hormone signaling [56]. Interestingly, the mitochondrial and the cytosolic Hsp60 have a contradictory role in pro-apoptotic and pro-survival mechanisms [57]. Whether Hsp60 is associated with carcinogenesis, specifically with tumor cell survival and proliferation, for certain tumors is used as a good diagnostic marker [58][59][60][61]. Therefore, Hsp60 exerts divergent roles in several physiological and pathological processes, and an understanding of its structural and functional biology aspires to draw novel pathways and to develop therapeutic strategies.

3. Stress Proteins: αB-Crystallin

HSPB1, or the CRYAB gene, encodes a 175-amino acid protein with a molecular mass of ~20 kDa [62]. CRYAB is a ubiquitous sHSP with highly conserved stretch that adopts a β-sandwich, immunoglobulin-like fold called the “α-crystallin domain (ACD),” which is a characteristic hallmark of the sHSPs family [63]. The ACD region is flanked by a less conserved and flexible N-terminal domain (NTD) and a C-Terminal extension (CTE), which are variable in length and sequence except for few conserved stretches [63][64].

Based on all findings related to CRYAB missense, truncating, and frame-shift mutations, specific functional roles of these domains have been hypothesized. Indeed, mutations within ACD domain (i.e., D109H/D109A, R120G) seem to interfere with the CRYAB oligomerization processes [65][66][67], while those within the CTE domain (i.e., 464delCT, R151X, G154S, L155fs_163X, R157H) seem to compromise CRYAB chaperone function [68][69][70][71][72][73]. Finally, mutations within the NTD domain seem to prevent the building of higher order oligomeric structures [74]. Thus, oligomeric assembly and chaperone activity of CRYAB is inter-dependent on its NTD, ACD and CTE domains.

As all sHSPs do, CRYAB shares in the property to form globular oligomer structures that in mammalian cells are characterized by molecular masses ranging from 50 to about 800 kDa. This ability, together with the well-known hetero-oligomerization property, is crucial factor in regulating the activity of this protein [75]. This hetero oligomer is probably unable to play an efficient protective role in stress conditions. However, the dissociation of the complex after-exposure to heat shock or oxidative stress suggests that it could bear new protein target recognition abilities and/or modulate those of the parental molecules [76].

Another intriguing property of sHSPs concerns its ability to be phosphorylated and therefore susceptible of control by several transduction pathways. Depending on the type and/or duration of various stimuli, the fraction of phosphorylated CRYAB ranges between 10% and 27% [77][78]. Different studies demonstrate that the phosphorylation of CRYAB shows a dual role that leads to either beneficial or deleterious outcomes depending on the extent and duration of stress and subsequent degree of phosphorylation; a phosphorylation at initial stage of stress is usually reversible and seems to provide a beneficial outcome, while prolonged stress can induce an irreversible phosphorylation which may lead to a deleterious outcome [79]. The CRYAB has three phosphorylation sites (S19, S45, and S59) at the NTD, which play a critical role in the protein functions. While the phosphorylation on S59 is mediated by p38 mitogen-activated protein kinase (p38 MAPK) and phosphorylation on S45 by the extracellular signal-regulated kinase 1/2 (ERK1/2) [78][80], the kinase responsible for phosphorylation on S19 is still unknown. Nevertheless, both unphosphorylated and phosphorylated forms of CRYAB are reported to be equally effective in preventing in vitro assembly of glial fibrillary acidic protein and vimentin in an ATP-independent manner [81]. In fact, during physiological or pathological stress both CRYAB content and phosphorylation can be modulated [82][83][84][85][86].

All aforementioned serine residues can be phosphorylated after various stimuli [78], but only a few studies have reported their contemporary involvement in muscle tissues [87][86][88][89][90]. To date, most of the available data are related to CRYAB expression and/or activation at Ser59 [82][83][91][92][93][94][95][96]. Moreover, the relationship between the phosphorylation of CRYAB and its chaperone activity is contradictory. Though in general the phosphorylation has an augmentative effect, it is possible that modulation of the activity upon phosphorylation might depend on the target protein and its interactions [97]. Further details about the phosphorylation of CRYAB in various physiological or pathological conditions can be found elsewhere [79][98].

In addition to being overexpressed in stress conditions, CRYAB shares the ability of having a tissue/cell-specific expression in the absence of stress, which can be detected in healthy adults as well as during organism development [99]. In mammalian cells, CRYAB is constitutively expressed in tissue with high rates of oxidative metabolism, including the cardiac and skeletal muscle [100]. The significance of the constitutive expression of this sHSP is probably linked to the protection of the cells against chronic stress or to a specific function in a particular tissue.

4. Hsp60 in Skeletal Muscle Fibers

The chaperoning systems that participate in controlling cellular homeostasis have been detected in skeletal muscle. Small Hsp, Hsp60, Hsp70, and Hsp90 play a significant role in muscle adaptation [101][102]. Nevertheless, Hsp60 was not deeply investigated after physical exercise, which, as we have already discussed, influences muscle homeostasis [47]. Hsp60 and exercise correlation appears to be rational, but literature data are restricted and sometimes controversial (Table 2). Morton et al. [103] demonstrated that aerobically trained men had significantly higher resting levels of Hsp60 in the vastus lateralis muscle (high percentage of fibers I and IIa) compared to untrained subjects, suggesting Hsp60 as a molecular marker of physiological adaptation to aerobic exercise. However, it has been demonstrated that in the human vastus lateralis muscle the highest mitochondrial content is showed by type I fibers followed by type IIa > IIx (Table 1) [19]. At the same time, an acute single bout of endurance training that is considered an aerobic exercise did not increase the basal Hsp60 protein levels in the same muscle [103]. Thus, chronic training has the capacity to increase Hsp60 expression, whereas a single bout of exercise does not. Hsp60 expression is stimulated by endurance, resistance, and mixed training, but its fiber specificity is still debated. Hsp60 expression in the vastus lateralis of healthy active people with different training backgrounds was considered not to be fiber-type specific [104]. In agreement, Ogata et al. [105] and Soares Moura et al. [106] did not show significant differences in Hsp60 levels in the plantaris and gastrocnemius, both rich fiber IIx muscles, of male rats after endurance training. On the other hand, several groups, including ours, demonstrated fiber-type specificity after training in specific muscles. Mattson et al. [107] showed that female rats trained with an endurance protocol for 8 weeks displayed significantly higher levels of Hsp60 in the muscle plantaris, which is rich in fiber-type IIb [16]. No difference of Hsp60 levels was detected in the rich fiber I muscle soleus in endurance-trained rats compared to the untrained group [17][107]. Hsp60 fiber-type I specificity was reported by Samelman [108], who showed increased basal levels of Hsp60 in the soleus and not in the lateral gastrocnemius of endurance trained rats. In agreement, our group noted Hsp60 fiber-specific expression in healthy male BALB/c mice trained for 45 days on the treadmill. Specifically, higher levels of Hsp60 were observed in type I and IIa muscle fibers, while type IIx and IIb fibers showed a constitutive expression of this chaperonin [109]. Therefore, increased levels of Hsp60 were reported after six weeks of endurance training, mainly in red gastrocnemius and soleus muscles, which are particularly rich in type I and IIa fibers [109]. We also correlated this physiological adaptation to an increased expression of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α), which triggers the mitochondrial biogenesis and thereby avoiding the cytotoxic effects [109][110][111]. Finally, increased levels of Hsp60 were observed in the soleus muscle of male mice and the Extensor Digitorum Longus (EDL) muscle of female mice after an acute single bout of endurance training [18].

Table 2. Hsp60 expression levels in different skeletal muscles after physical exercise.

This entry is adapted from the peer-reviewed paper 10.3390/biology10020077

References

  1. Weiss, A.; McDonough, D.; Wertman, B.; Acakpo-Satchivi, L.; Montgomery, K.; Kucherlapati, R.; Leinwand, L.; Krauter, K. Organization of human and mouse skeletal myosin heavy chain gene clusters is highly conserved. Proc. Natl. Acad. Sci. USA 1999, 96, 2958–2963.
  2. Weiss, A.; Schiaffino, S.; Leinwand, L.A. Comparative sequence analysis of the complete human sarcomeric myosin heavy chain family: Implications for functional diversity. J. Mol. Biol. 1999, 290, 61–75.
  3. Schiaffino, S.; Reggiani, C. Fiber types in mammalian skeletal muscles. Physiol. Rev. 2011, 91, 1447–1531.
  4. Morton, J.P.; MacLaren, D.P.; Cable, N.T.; Bongers, T.; Griffiths, R.D.; Campbell, I.T.; Evans, L.; Kayani, A.; McArdle, A.; Drust, B. Time course and differential responses of the major heat shock protein families in human skeletal muscle following acute nondamaging treadmill exercise. J. Appl. Physiol. (1985) 2006, 101, 176–182.
  5. Schiaffino, S.; Reggiani, C. Myosin isoforms in mammalian skeletal muscle. J. Appl. Physiol. 1994, 77, 493–501.
  6. Staron, R.S.; Pette, D. The continuum of pure and hybrid myosin heavy chain-based fibre types in rat skeletal muscle. Histochemistry 1993, 100, 149–153.
  7. Schiaffino, S. Fibre types in skeletal muscle: A personal account. Acta Physiol. 2010, 199, 451–463.
  8. Bottinelli, R.; Canepari, M.; Pellegrino, M.A.; Reggiani, C. Force-velocity properties of human skeletal muscle fibres: Myosin heavy chain isoform and temperature dependence. J. Physiol. 1996, 495, 573–586.
  9. Ranatunga, K.W.; Thomas, P.E. Correlation between shortening velocity, force-velocity relation and histochemical fibre-type composition in rat muscles. J. Muscle Res. Cell Motil. 1990, 11, 240–250.
  10. Edgerton, V.R.; Roy, R.R. Regulation of skeletal muscle fiber size, shape and function. J. Biomech. 1991, 24, 123–133.
  11. Talmadge, R.J.; Roy, R.R.; Edgerton, V.R. Muscle fiber types and function. Curr. Opin. Rheumatol. 1993, 5, 695–705.
  12. Resnicow, D.I.; Deacon, J.C.; Warrick, H.M.; Spudich, J.A.; Leinwand, L.A. Functional diversity among a family of human skeletal muscle myosin motors. Proc. Natl. Acad. Sci. USA 2010, 107, 1053–1058.
  13. Reiser, P.J.; Moss, R.L.; Giulian, G.G.; Greaser, M.L. Shortening velocity in single fibers from adult rabbit soleus muscles is correlated with myosin heavy chain composition. J. Biol. Chem. 1985, 260, 9077–9080.
  14. Armstrong, R.B.; Phelps, R.O. Muscle fiber type composition of the rat hindlimb. Am. J. Anat. 1984, 171, 259–272.
  15. Bloemberg, D.; Quadrilatero, J. Rapid determination of myosin heavy chain expression in rat, mouse, and human skeletal muscle using multicolor immunofluorescence analysis. PLoS ONE 2012, 7, e35273.
  16. Haddad, F.; Arnold, C.; Zeng, M.; Baldwin, K. Interaction of thyroid state and denervation on skeletal myosin heavy chain expression. Muscle Nerve 1997, 20, 1487–1496.
  17. Drzymala-Celichowska, H.; Karolczak, J.; Redowicz, M.J.; Bukowska, D. The content of myosin heavy chains in hindlimb muscles of female and male rats. J. Physiol. Pharmacol. Off. J. Pol. Physiol. Soc. 2012, 63, 187–193.
  18. D’Amico, D.; Marino Gammazza, A.; Macaluso, F.; Paladino, L.; Scalia, F.; Spinoso, G.; Dimauro, I.; Caporossi, D.; Cappello, F.; Di Felice, V.; et al. Sex-based differences after a single bout of exercise on PGC1α isoforms in skeletal muscle: A pilot study. FASEB J. 2021, 35, e21328.
  19. Gouspillou, G.; Sgarioto, N.; Norris, B.; Barbat-Artigas, S.; Aubertin-Leheudre, M.; Morais, J.A.; Burelle, Y.; Taivassalo, T.; Hepple, R.T. The relationship between muscle fiber type-specific PGC-1alpha content and mitochondrial content varies between rodent models and humans. PLoS ONE 2014, 9, e103044.
  20. Meissner, J.D.; Umeda, P.K.; Chang, K.C.; Gros, G.; Scheibe, R.J. Activation of the beta myosin heavy chain promoter by MEF-2D, MyoD, p300, and the calcineurin/NFATc1 pathway. J. Cell. Physiol. 2007, 211, 138–148.
  21. Widrick, J.J.; Romatowski, J.G.; Bain, J.L.; Trappe, S.W.; Trappe, T.A.; Thompson, J.L.; Costill, D.L.; Riley, D.A.; Fitts, R.H. Effect of 17 days of bed rest on peak isometric force and unloaded shortening velocity of human soleus fibers. Am. J. Physiol. 1997, 273, C1690–C1699.
  22. Widrick, J.J.; Knuth, S.T.; Norenberg, K.M.; Romatowski, J.G.; Bain, J.L.; Riley, D.A.; Karhanek, M.; Trappe, S.W.; Trappe, T.A.; Costill, D.L.; et al. Effect of a 17 day spaceflight on contractile properties of human soleus muscle fibres. J. Physiol. 1999, 516, 915–930.
  23. Bottinelli, R.; Pellegrino, M.A.; Canepari, M.; Rossi, R.; Reggiani, C. Specific contributions of various muscle fibre types to human muscle performance: An in vitro study. J. Electromyogr. Kinesiol. Off. J. Int. Soc. Electrophysiol. Kinesiol. 1999, 9, 87–95.
  24. Hikida, R.S.; Staron, R.S.; Hagerman, F.C.; Walsh, S.; Kaiser, E.; Shell, S.; Hervey, S. Effects of high-intensity resistance training on untrained older men. II. Muscle fiber characteristics and nucleo-cytoplasmic relationships. J. Gerontol. Ser. A Biol. Sci. Med Sci. 2000, 55, B347–B354.
  25. Campos, G.E.; Luecke, T.J.; Wendeln, H.K.; Toma, K.; Hagerman, F.C.; Murray, T.F.; Ragg, K.E.; Ratamess, N.A.; Kraemer, W.J.; Staron, R.S. Muscular adaptations in response to three different resistance-training regimens: Specificity of repetition maximum training zones. Eur. J. Appl. Physiol. 2002, 88, 50–60.
  26. Allen, D.L.; Harrison, B.C.; Maass, A.; Bell, M.L.; Byrnes, W.C.; Leinwand, L.A. Cardiac and skeletal muscle adaptations to voluntary wheel running in the mouse. J. Appl. Physiol. 2001, 90, 1900–1908.
  27. Ausoni, S.; Gorza, L.; Schiaffino, S.; Gundersen, K.; Lomo, T. Expression of myosin heavy chain isoforms in stimulated fast and slow rat muscles. J. Neurosci. Off. J. Soc. Neurosci. 1990, 10, 153–160.
  28. Liu, Y.; Schlumberger, A.; Wirth, K.; Schmidtbleicher, D.; Steinacker, J.M. Different effects on human skeletal myosin heavy chain isoform expression: Strength vs. combination training. J. Appl. Physiol. 2003, 94, 2282–2288.
  29. Kesidis, N.; Metaxas, T.I.; Vrabas, I.S.; Stefanidis, P.; Vamvakoudis, E.; Christoulas, K.; Mandroukas, A.; Balasas, D.; Mandroukas, K. Myosin heavy chain isoform distribution in single fibres of bodybuilders. Eur. J. Appl. Physiol. 2008, 103, 579–583.
  30. Staron, R.S. Correlation between myofibrillar ATPase activity and myosin heavy chain composition in single human muscle fibers. Histochemistry 1991, 96, 21–24.
  31. Putman, C.T.; Xu, X.; Gillies, E.; MacLean, I.M.; Bell, G.J. Effects of strength, endurance and combined training on myosin heavy chain content and fibre-type distribution in humans. Eur. J. Appl. Physiol. 2004, 92, 376–384.
  32. Mukherjee, K.; Conway de Macario, E.; Macario, A.J.; Brocchieri, L. Chaperonin genes on the rise: New divergent classes and intense duplication in human and other vertebrate genomes. BMC Evol. Biol. 2010, 1, 64.
  33. Caruso Bavisotto, C.; Alberti, G.; Vitale, A.M.; Paladino, L.; Campanella, C.; Rappa, F.; Gorska, M.; Conway de Macario, E.; Cappello, F.; Macario, A.J.L.; et al. Hsp60 Post-translational Modifications: Functional and Pathological Consequences. Front. Mol. Biosci. 2020, 7, 95.
  34. Ishii, N. GroEL and the GroEL-GroES Complex. Sub Cell. Biochem. 2017, 83, 483–504.
  35. Vilasi, S.; Bulone, D.; Caruso Bavisotto, C.; Campanella, C.; Marino Gammazza, A.; San Biagio, P.L.; Cappello, F.; Conway de Macario, E.; Macario, A.J.L. Chaperonin of Group I: Oligomeric Spectrum and Biochemical and Biological Implications. Front. Mol. Biosci. 2017, 4, 99.
  36. Horwich, A.L. Protein folding in the cell: An inside story. Nat. Med. 2011, 17, 1211–1216.
  37. Henderson, B.; Fares, M.A.; Lund, P.A. Chaperonin 60: A paradoxical, evolutionarily conserved protein family with multiple moonlighting functions. Biol. Rev. Camb. Philos. Soc. 2013, 88, 955–987.
  38. Nisemblat, S.; Parnas, A.; Yaniv, O.; Azem, A.; Frolow, F. Crystallization and structure determination of a symmetrical ‘football’ complex of the mammalian mitochondrial Hsp60-Hsp10 chaperonins. Acta Crystallogr. Sect. F Struct. Biol. Commun. 2014, 70, 116–119.
  39. Viitanen, P.V.; Lorimer, G.H.; Seetharam, R.; Gupta, R.S.; Oppenheim, J.; Thomas, J.O.; Cowan, N.J. Mammalian mitochondrial chaperonin 60 functions as a single toroidal ring. J. Biol. Chem. 1992, 267, 695–698.
  40. Vilasi, S.; Carrotta, R.; Mangione, M.R.; Campanella, C.; Librizzi, F.; Randazzo, L.; Martorana, V.; Marino Gammazza, A.; Ortore, M.G.; Vilasi, A.; et al. Human Hsp60 with its mitochondrial import signal occurs in solution as heptamers and tetradecamers remarkably stable over a wide range of concentrations. PLoS ONE 2014, 9, e97657.
  41. Nielsen, K.L.; Cowan, N.J. A single ring is sufficient for productive chaperonin-mediated folding in vivo. Mol. Cell 1998, 2, 93–99.
  42. Christensen, J.H.; Nielsen, M.N.; Hansen, J.; Fuchtbauer, A.; Fuchtbauer, E.M.; West, M.; Corydon, T.J.; Gregersen, N.; Bross, P. Inactivation of the hereditary spastic paraplegia-associated Hspd1 gene encoding the Hsp60 chaperone results in early embryonic lethality in mice. Cell Stress Chaperones 2010, 15, 851–863.
  43. Berger, E.; Rath, E.; Yuan, D.; Waldschmitt, N.; Khaloian, S.; Allgauer, M.; Staszewski, O.; Lobner, E.M.; Schottl, T.; Giesbertz, P.; et al. Mitochondrial function controls intestinal epithelial stemness and proliferation. Nat. Commun. 2016, 7, 13171.
  44. Tang, H.; Li, J.; Liu, X.; Wang, G.; Luo, M.; Deng, H. Down-regulation of HSP60 Suppresses the Proliferation of Glioblastoma Cells via the ROS/AMPK/mTOR Pathway. Sci. Rep. 2016, 6, 28388.
  45. Cappello, F.; Marino Gammazza, A.; Palumbo Piccionello, A.; Campanella, C.; Pace, A.; Conway de Macario, E.; Macario, A.J. Hsp60 chaperonopathies and chaperonotherapy: Targets and agents. Expert Opin. Ther. Targets 2014, 18, 185–208.
  46. Singh, B.; Patel, H.V.; Ridley, R.G.; Freeman, K.B.; Gupta, R.S. Mitochondrial import of the human chaperonin (HSP60) protein. Biochem. Biophys. Res. Commun. 1990, 169, 391–396.
  47. Campanella, C.; Bucchieri, F.; Merendino, A.M.; Fucarino, A.; Burgio, G.; Corona, D.F.; Barbieri, G.; David, S.; Farina, F.; Zummo, G.; et al. The odyssey of Hsp60 from tumor cells to other destinations includes plasma membrane-associated stages and Golgi and exosomal protein-trafficking modalities. PLoS ONE 2012, 7, e42008.
  48. Cappello, F.; Conway de Macario, E.; Marasa, L.; Zummo, G.; Macario, A.J. Hsp60 expression, new locations, functions and perspectives for cancer diagnosis and therapy. Cancer Biol. Ther. 2008, 7, 801–909.
  49. Voos, W. Chaperone-protease networks in mitochondrial protein homeostasis. Biochim. Biophys. Acta 2013, 1833, 388–399.
  50. Cheng, M.Y.; Hartl, F.U.; Martin, J.; Pollock, R.A.; Kalousek, F.; Neupert, W.; Hallberg, E.M.; Hallberg, R.L.; Horwich, A.L. Mitochondrial heat-shock protein hsp60 is essential for assembly of proteins imported into yeast mitochondria. Nature 1989, 337, 620–625.
  51. Lin, Z.; Madan, D.; Rye, H.S. GroEL stimulates protein folding through forced unfolding. Nat. Struct. Mol. Biol. 2008, 15, 303–311.
  52. Marino Gammazza, A.; Bavisotto, C.C.; Barone, R.; de Macario, E.C.; Macario, A.J. Alzheimer’s Disease and Molecular Chaperones: Current Knowledge and the Future of Chaperonotherapy. Curr. Pharm. Des. 2016, 22, 4040–4049.
  53. Kaufman, B.A.; Newman, S.M.; Hallberg, R.L.; Slaughter, C.A.; Perlman, P.S.; Butow, R.A. In organello formaldehyde crosslinking of proteins to mtDNA: Identification of bifunctional proteins. Proc. Natl. Acad. Sci. USA 2000, 97, 7772–7777.
  54. Kaufman, B.A.; Kolesar, J.E.; Perlman, P.S.; Butow, R.A. A function for the mitochondrial chaperonin Hsp60 in the structure and transmission of mitochondrial DNA nucleoids in Saccharomyces cerevisiae. J. Cell Biol. 2003, 163, 457–461.
  55. Deocaris, C.C.; Kaul, S.C.; Wadhwa, R. On the brotherhood of the mitochondrial chaperones mortalin and heat shock protein 60. Cell Stress Chaperones 2006, 11, 116–128.
  56. Sigal, L.H.; Williams, S.; Soltys, B.; Gupta, R. H9724, a monoclonal antibody to Borrelia burgdorferi’s flagellin, binds to heat shock protein 60 (HSP60) within live neuroblastoma cells: A potential role for HSP60 in peptide hormone signaling and in an autoimmune pathogenesis of the neuropathy of Lyme disease. Cell. Mol. Neurobiol. 2001, 21, 477–495.
  57. Chandra, D.; Choy, G.; Tang, D.G. Cytosolic accumulation of HSP60 during apoptosis with or without apparent mitochondrial release: Evidence that its pro-apoptotic or pro-survival functions involve differential interactions with caspase-3. J. Biol. Chem. 2007, 282, 31289–31301.
  58. Cappello, F.; Bellafiore, M.; Palma, A.; David, S.; Marciano, V.; Bartolotta, T.; Sciume, C.; Modica, G.; Farina, F.; Zummo, G.; et al. 60KDa chaperonin (HSP60) is over-expressed during colorectal carcinogenesis. Eur. J. Histochem. 2003, 47, 105–110.
  59. Cappello, F.; David, S.; Rappa, F.; Bucchieri, F.; Marasa, L.; Bartolotta, T.E.; Farina, F.; Zummo, G. The expression of HSP60 and HSP10 in large bowel carcinomas with lymph node metastase. BMC Cancer 2005, 5, 139.
  60. Caruso Bavisotto, C.; Cipolla, C.; Graceffa, G.; Barone, R.; Bucchieri, F.; Bulone, D.; Cabibi, D.; Campanella, C.; Marino Gammazza, A.; Pitruzzella, A.; et al. Immunomorphological Pattern of Molecular Chaperones in Normal and Pathological Thyroid Tissues and Circulating Exosomes: Potential Use in Clinics. Int. J. Mol. Sci. 2019, 20, 4496.
  61. Faried, A.; Sohda, M.; Nakajima, M.; Miyazaki, T.; Kato, H.; Kuwano, H. Expression of heat-shock protein Hsp60 correlated with the apoptotic index and patient prognosis in human oesophageal squamous cell carcinoma. Eur. J. Cancer 2004, 40, 2804–2811.
  62. Dubin, R.A.; Ally, A.H.; Chung, S.; Piatigorsky, J. Human alpha B-crystallin gene and preferential promoter function in lens. Genomics 1990, 7, 594–601.
  63. Kappé, G.; Franck, E.; Verschuure, P.; Boelens, W.C.; Leunissen, J.A.; de Jong, W.W. The human genome encodes 10 alpha-crystallin-related small heat shock proteins: HspB1-10. Cell Stress Chaperones 2003, 8, 53–61.
  64. Kriehuber, T.; Rattei, T.; Weinmaier, T.; Bepperling, A.; Haslbeck, M.; Buchner, J. Independent evolution of the core domain and its flanking sequences in small heat shock proteins. FASEB J. 2010, 24, 3633–3642.
  65. Fichna, J.P.; Potulska-Chromik, A.; Miszta, P.; Redowicz, M.J.; Kaminska, A.M.; Zekanowski, C.; Filipek, S. A novel dominant D109A CRYAB mutation in a family with myofibrillar myopathy affects αB-crystallin structure. BBA Clin. 2016, 11, 1–7.
  66. Sacconi, S.; Féasson, L.; Antoine, J.C.; Pécheux, C.; Bernard, R.; Cobo, A.M.; Casarin, A.; Salviati, L.; Desnuelle, C.; Urtizberea, A. A novel CRYAB mutation resulting in multisystemic disease. Neuromuscul. Disord. 2012, 22, 66–72.
  67. Vicart, P.; Caron, A.; Guicheney, P.; Li, Z.; Prévost, M.C.; Faure, A.; Chateau, D.; Chapon, F.; Tomé, F.; Dupret, J.M.; et al. A missense mutation in the alphaB-crystallin chaperone gene causes a desmin-related myopathy. Nat. Genet. 1998, 20, 92–95.
  68. Gabai, V.L.; Sherman, M.Y. Invited review: Interplay between molecular chaperones and signaling pathways in survival of heat shock. J. Appl. Physiol. (1985) 2002, 92, 1743–1748.
  69. Del Bigio, M.R.; Chudley, A.E.; Sarnat, H.B.; Campbell, C.; Goobie, S.; Chodirker, B.N.; Selcen, D. Infantile muscular dystrophy in Canadian aboriginals is an αB-crystallinopathy. Ann. Neurol. 2011, 69, 866–871.
  70. Inagaki, N.; Hayashi, T.; Arimura, T.; Koga, Y.; Takahashi, M.; Shibata, H.; Teraoka, K.; Chikamori, T.; Yamashina, A.; Kimura, A. Alpha B-crystallin mutation in dilated cardiomyopathy. Biochem. Biophys Res. Commun. 2006, 7, 379–386.
  71. Pilotto, A.; Marziliano, N.; Pasotti, M.; Grasso, M.; Costante, A.M.; Arbustini, E. alphaB-crystallin mutation in dilated cardiomyopathies: Low prevalence in a consecutive series of 200 unrelated probands. Biochem. Biophys Res. Commun. 2006, 346, 1115–1117.
  72. Reilich, P.; Schoser, B.; Schramm, N.; Krause, S.; Schessl, J.; Kress, W.; Müller-Höcker, J.; Walter, M.C.; Lochmuller, H. The p.G154S mutation of the alpha-B crystallin gene (CRYAB) causes late-onset distal myopathy. Neuromuscul. Disord. 2010, 20, 255–259.
  73. Selcen, D.; Engel, A.G. Myofibrillar myopathy caused by novel dominant negative alpha B-crystallin mutations. Ann. Neurol. 2003, 54, 804–810.
  74. Jehle, S.; Vollmar, B.S.; Bardiaux, B.; Dove, K.K.; Rajagopal, P.; Gonen, T.; Oschkinat, H.; Klevit, R.E. N-terminal domain of alphaB-crystallin provides a conformational switch for multimerization and structural heterogeneity. Proc. Natl. Acad. Sci. USA 2011, 108, 6409–6414.
  75. Zantema, A.; Verlaan-De Vries, M.; Maasdam, D.; Bol, S.; van der Eb, A. Heat shock protein 27 and alpha B-crystallin can form a complex, which dissociates by heat shock. J. Biol. Chem. 1992, 25, 12936–12941.
  76. Fontaine, J.M.; Sun, X.; Benndorf, R.; Welsh, M.J. Interactions of HSP22 (HSPB8) with HSP20, alphaB-crystallin, and HSPB3. Biochem. Biophys Res. Commun. 2005, 337, 1006–1011.
  77. Eaton, P.; Fuller, W.; Bell, J.R.; Shattock, M.J. AlphaB crystallin translocation and phosphorylation: Signal transduction pathways and preconditioning in the isolated rat heart. J. Mol. Cell Cardiol. 2001, 33, 1659–1671.
  78. Ito, H.; Okamoto, K.; Nakayama, H.; Isobe, T.; Kato, K. Phosphorylation of alphaB-crystallin in response to various types of stress. J. Biol. Chem. 1997, 272, 29934–29941.
  79. Bakthisaran, R.; Tangirala, R.; Rao, C.M. Small heat shock proteins: Role in cellular functions and pathology. Biochim. Biophys Acta 2015, 1854, 291–319.
  80. Kato, K.; Ito, H.; Kamei, K.; Inaguma, Y.; Iwamoto, I.; Saga, S. Phosphorylation of alpha beta-crystallin in mitotic cells and identification enzymatic activities responsible for phosphorylation. J. Biol. Chem. 1998, 273, 28346–28354.
  81. Nicholl, I.D.; Quinlan, R.A. Chaperone activity of alpha-crystallins modulates intermediate filament assembly. EMBO J. 1994, 13, 945–953.
  82. Adhikari, A.S.; Singh, B.N.; Rao, K.S.; Rao, C.M. αB-crystallin, a small heat shock protein, modulates NF-κB activity in a phosphorylation-dependent manner and protects muscle myoblasts from TNF-α induced cytotoxicity. Biochim. Biophys Acta 2011, 1813, 1532–1542.
  83. Fittipaldi, S.; Mercatelli, N.; Dimauro, I.; Jackson, M.J.; Paronetto, M.P.; Caporossi, D. Alpha B-crystallin induction in skeletal muscle cells under redox imbalance is mediated by a JNK-dependent regulatory mechanism. Free Radic. Biol. Med. 2015, 86, 331–342.
  84. Morrison, L.E.; Hoover, H.E.; Thuerauf, D.J.; Glembotski, C.C. Mimicking phosphorylation of αB-Crystallin on Serine-59 is necessary and sufficient to provide maximal protection of cardiac myocytes from apoptosis. Circ. Res. 2003, 92, 203–211.
  85. Morrison, L.E.; Whittaker, R.J.; Klepper, R.E.; Wawrousek, E.F.; Glembotski, C.C. Roles for alphaB-crystallin and HSPB2 in protecting the myocardium from ischemia-reperfusion-induced damage in a KO mouse model. Am. J. Physiol. Heart Circ. Physiol. 2004, 286, 847–855.
  86. Reddy, V.S.; Jakhotia, S.; Reddy, P.Y.; Reddy, G.B. Hyperglycemia induced expression, phosphorylation, and translocation of αB-crystallin in rat skeletal muscle. IUBMB Life 2015, 67, 291–299.
  87. Dimauro, I.; Antonioni, A.; Mercatelli, N.; Grazioli, E.; Fantini, C.; Barone, R.; Macaluso, F.; Di Felice, V.; Caporossi, D. The early response of αB-crystallin to a single bout of aerobic exercise in mouse skeletal muscles depends upon fiber oxidative features. Redox Biol. 2019, 24, 101183.
  88. Den Engelsman, J.; Gerrits, D.; de Jong, W.W.; Robbins, J.; Kato, K.; Boelens, W.C. Nuclear import of B-crystallin is phosphorylation-dependent and hampered by hyperphosphorylation of the myopathy-related mutant R120G. J. Biol. Chem. 2005, 280, 37139–37148.
  89. Launay, N.; Goudeau, B.; Kato, K.; Vicart, P.; Lilienbaum, A. Cell signaling pathways to alphaB-crystallin following stresses of the cytoskeleton. Expt. Cell Res. 2006, 312, 3570–3584.
  90. Antonioni, A.; Dimauro, I.; Fantini, C.; Barone, R.; Macaluso, F.; Di Felice, V.; Caporossi, D. αB-crystallin response to a pro-oxidant non-cytotoxic environment in murine cardiac cells: An “in vitro” and “in vivo” study. Free Radic. Biol. Med. 2020, 152, 301–312.
  91. Aggeli, I.K.; Beis, I.; Gaitanaki, C. Oxidative stress and calpain inhibition induce alpha B-crystallin phosphorylation via p38-MAPK and calcium signalling pathways in H9c2 cells. Cell. Signal. 2008, 20, 1292–1302.
  92. Beltran Valls, M.R.; Wilkinson, D.J.; Narici, M.V.; Smith, K.; Phillips, B.E.; Caporossi, D.; Atherton, P.J. Protein carbonylation and heat shock proteins in human skeletal muscle: Relationships to age and sarcopenia. J. Gerontol. A Biol. Sci. Med. Sci. 2015, 70, 174–181.
  93. Ivanov, O.; Chen, F.; Wiley, E.L.; Keswani, A.; Diaz, L.K.; Memmel, H.C.; Rademaker, A.; Gradishar, W.J.; Morrow, M.; Khan, S.A.; et al. alphaB-crystallin is a novel predictor of resistance to neoadjuvant chemotherapy in breast cancer. Breast Cancer Res. Treat. 2008, 111, 411–417.
  94. Neppl, R.L.; Kataoka, M.; Wang, D.Z. Crystallin-αB regulates skeletal muscle homeostasis via modulation of argonaute2 activity. J. Biol. Chem. 2014, 289, 17240–17248.
  95. Magi, F.; Dimauro, I.; Margheritini, F.; Duranti, G.; Mercatelli, N.; Fantini, C.; Ripani, F.R.; Sabatini, S.; Caporossi, D. Telomere length is independently associated with age, oxidative biomarkers, and sport training in skeletal muscle of healthy adult males. Free Radic. Res. 2018, 52, 639–647.
  96. Pereira, M.B.; Santos, A.M.; Gonçalves, D.C.; Cardoso, A.C.; Consonni, S.R.; Gozzo, F.C.; Oliveira, P.S.; Pereira, A.H.; Figueiredo, A.R.; Tiroli-Cepeda, A.O.; et al. Corrigendum: αB-crystallin interacts with and prevents stress-activated proteolysis of focal adhesion kinase by calpain in cardiomyocytes. Nat. Commun. 2015, 23, 6508.
  97. Koteiche, H.A.; McHaourab, H.S. Mechanism of chaperone function in small heat shock proteins. Phosphorylation-induced activation of two modes binding in alphaB-crystallin. J. Biol. Chem. 2003, 278, 10361–10367.
  98. Caporossi, D.; Parisi, A.; Fantini, C.; Grazioli, E.; Cerulli, C.; Dimauro, I. AlphaB-crystallin and breast cancer: Role and possible therapeutic strategies. Cell Stress Chaperones 2020.
  99. Richter, K.; Haslbeck, M.; Buchner, J. The heat shock response: Life on the verge of death. Mol. Cell 2010, 22, 253–266.
  100. Bhat, S.P.; Nagineni, C.N. αB subunit of lens-specific protein α-crystallin is present in other ocular and non-ocular tissue. Biochem. Biophys. Res. Commun. 1989, 158, 319–325.
  101. Morton, J.P.; Kayani, A.C.; McArdle, A.; Drust, B. The exercise-induced stress response of skeletal muscle, with specific emphasis on humans. Sports Med. 2009, 39, 643–662.
  102. Bornman, L.; Polla, B.S.; Lotz, B.P.; Gericke, G.S. Expression of heat-shock/stress proteins in Duchenne muscular dystrophy. Muscle Nerve 1995, 18, 23–31.
  103. Morton, J.P.; Maclaren, D.P.; Cable, N.T.; Campbell, I.T.; Evans, L.; Kayani, A.C.; McArdle, A.; Drust, B. Trained men display increased basal heat shock protein content of skeletal muscle. Med. Sci. Sports Exerc. 2008, 40, 1255–1262.
  104. Folkesson, M.; Mackey, A.L.; Langberg, H.; Oskarsson, E.; Piehl-Aulin, K.; Henriksson, J.; Kadi, F. The expression of heat shock protein in human skeletal muscle: Effects of muscle fibre phenotype and training background. Acta Physiol. 2013, 209, 26–33.
  105. Ogata, T.; Oishi, Y.; Higashida, K.; Higuchi, M.; Muraoka, I. Prolonged exercise training induces long-term enhancement of HSP70 expression in rat plantaris muscle. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2009, 296, R1557–R1563.
  106. Moura, C.S.; Lollo, P.C.; Morato, P.N.; Nisishima, L.H.; Carneiro, E.M.; Amaya-Farfan, J. Whey protein hydrolysate enhances HSP90 but does not alter HSP60 and HSP25 in skeletal muscle of rats. PLoS ONE 2014, 9, e83437.
  107. Mattson, J.P.; Ross, C.R.; Kilgore, J.L.; Musch, T.I. Induction of mitochondrial stress proteins following treadmill running. Med. Sci. Sports Exerc. 2000, 32, 365–369.
  108. Samelman, T.R. Heat shock protein expression is increased in cardiac and skeletal muscles of Fischer 344 rats after endurance training. Exp. Physiol. 2000, 85, 92–102.
  109. Barone, R.; Macaluso, F.; Sangiorgi, C.; Campanella, C.; Marino Gammazza, A.; Moresi, V.; Coletti, D.; Conway de Macario, E.; Macario, A.J.; Cappello, F.; et al. Skeletal muscle Heat shock protein 60 increases after endurance training and induces peroxisome proliferator-activated receptor gamma coactivator 1 alpha1 expression. Sci. Rep. 2016, 6, 19781.
  110. Barone, R.; Sangiorgi, C.; Marino Gammazza, A.; D’Amico, D.; Salerno, M.; Cappello, F.; Pomara, C.; Zummo, G.; Farina, F.; Di Felice, V.; et al. Effects of Conjugated Linoleic Acid Associated with Endurance Exercise on Muscle Fibres and Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1 alpha Isoforms. J. Cell. Physiol. 2017, 232, 1086–1094.
  111. Hood, D.A.; Takahashi, M.; Connor, M.K.; Freyssenet, D. Assembly of the cellular powerhouse: Current issues in muscle mitochondrial biogenesis. Exerc. Sport Sci. Rev. 2000, 28, 68–73.
More
This entry is offline, you can click here to edit this entry!
ScholarVision Creations