HIGHLIGHTED TOPICSExercise Effects on Muscle Insulin Signaling and Action

Invited Review: Autocrine/paracrine IGF-I and skeletal muscle adaptation

Abstract

This brief review presents the basic premises suggesting that insulin-like growth factor I (IGF-I), functioning in an autocrine/paracrine mode, is an important mediator of skeletal muscle adaptation. Key intracellular signaling mechanisms associated with ligation of the primary IGF-I receptor are highlighted to illustrate the mechanisms by which IGF-I may promote muscle hypertrophy. In addition, a number of recent findings are presented that highlight the potential for interactions between IGF-I-related signaling pathways and intracellular signaling mechanisms activated by cytokines or hormonal systems.

insulin-like growth factor I (IGF-I) is the primary mediator of many of the responses regulated by growth hormone in tissues throughout the body (15,116). In addition, it has long been recognized that IGF-I and IGF-II are important for the pre- and postnatal development of skeletal muscle (13, 38). In the specific context of IGF-I as it relates to muscle, the objectives of this review are twofold:1) to briefly outline some of the key factors that have led to the continued interest in IGF-I as a potential mediator of loading-induced skeletal muscle adaptation and 2) to widen the discourse on IGF-I via the inclusion of topics that appear to have received less notice in the muscle-related IGF-I literature. The format will be that of a brief review and thus will not be exhaustive in nature. For readers seeking more depth, there are a number of excellent reviews available in the literature (e.g., Ref. 96).

LOCAL CONTROL OF SKELETAL MUSCLE ADAPTATION

It has become increasingly clear that skeletal muscle is constantly adapting to the functional demands imposed by the load-bearing activities of the individual. In mammalian skeletal muscle, this adaptation process can include changes in both the size and the structural/functional properties of the myofibers. The focus on IGF-I, as well as a number of other growth factors, has been driven in part by the recognition that activity-induced skeletal muscle adaptation is largely mediated by intrinsic mechanisms. Interestingly, a number of studies have demonstrated that adaptations such as muscle hypertrophy can occur even when the somatic milieu would be considered nonanabolic. For example, in rats, the circulating hormone and growth factor milieu can be drastically depressed via surgical hypophysectomy (Hx), which prevents further somatic growth. However, despite this depression of the somatic growth factor environment, the muscles of Hx rats can respond to increased loading with substantial compensatory hypertrophy (40). In this model, the circulating and tissue levels of IGF-I are substantially decreased in Hx rats, but the compensatory hypertrophy process includes a robust increase in the expression of IGF-I mRNA and peptide in the overloaded muscles (2, 30).

MUSCLE REGENERATION

To understand the importance of intrinsic regulation via autocrine/paracrine signaling, it is instructive to consider some cellular processes, such as myofiber regeneration, which appear to be modulated by IGF-I. In models of severe muscle injury, the death of myofibers leaves behind the basal lamina and some satellite cells. Satellite cells are small mononucleated skeletal muscle stem cells1 located between the basal lamina of the muscle and the sarcolemma of myofibers. As a result of the injury to myofibers, these satellite cells are mobilized to begin the regeneration process (26, 75, 77, 93). The initial events after satellite cell activation have been reported to be a proliferative response in which some or all of the activated satellite cells undergo at least one mitotic cycle (75, 83,94). After this initial phase, some of the activated cells and/or their progeny are thought to differentiate into myoblast-like cells. In regenerating muscle, these myoblasts can either fuse with each other to form new myofibers or become incorporated into damaged but surviving myofibers (11, 57, 65, 77, 78). If the capacity of satellite cells to proliferate is eliminated, for example via irradiation, the regeneration process is inhibited (26, 42,75). There is evidence that locally produced, i.e., autocrine/paracrine IGF-I, may be important in this regeneration process. Jennische et al. demonstrated that increased IGF-I immunoreactivity can be detected in the cytoplasm of myoblasts and myotubes (44) as well as in satellite cells (45) during muscle regeneration. Furthermore, the introduction of neutralizing antibodies, which prevent either IGF-I or fibroblast growth factor (FGF-2) activity, has been shown to reduce the number and size of regenerating myofibers after muscle injury with anti-IGF-I treatment demonstrating a higher potency (53).

INTRACELLULAR IGF-I SIGNALING

With the muscle regeneration process in mind, an examination of the known effects of IGF-I on skeletal muscle cells provides insight into potential mechanisms by which this growth factor may contribute to muscle repair or adaptation. In studies involving both established cell lines and primary satellite cell cultures, ligation of the type 1 IGF-I receptor (IGFR1) has been shown to initiate intracellular signaling cascades involved in key mitogenic and myogenic responses (25,38, 79). One pathway activated by IGF-I involves Ras-Raf signaling to extracellular response kinases (ERKs), which can activate a number of transcription factors as well as other protein kinases. In muscle cell cultures, this pathway has been shown to promote increased cell proliferation (Fig. 1) (e.g., Ref.25). A second pathway involves phosphorylation of insulin receptor substrate and leads to the activation of phosphatidylinositol 3-kinase (PI3K) (Fig. 2). PI3K activation is central to a number of important cellular processes, including protection from apoptosis, increased translation, and alteration in intracellular calcium. PI3K activation increases the initiation of translation via alterations in the phosphorylation state of eukaryotic initiation factor 4 binding protein and the p70 S6 kinase (Fig. 2). The activation of p70 S6-kinase is of particular interest in that it enhances the translation of mRNAs encoding ribosomal proteins and elongation factors, integral components of the protein synthesis machinery (106). In addition to generalized anabolic effects, activation of portions of the PI3K signaling cascade appear to be particularly important for the differentiation of muscle cell lines in culture (16, 17, 25, 52, 105, 107, 110).

Fig. 1.

Fig. 1.The Ras-ERK signaling cascade. A simplified diagram of one intracellular signaling pathway associated with tyrosine kinase activity of the type 1 insulin-like growth factor receptor (IGFR1). A number of studies have linked this pathway with the control of muscle cell proliferation in vitro. The phosphorylation targets of ERKs include transcription factors and additional protein kinases. ERK, extracellular signal-regulated kinase; MEK, mitogen-activated protein kinase (MAPK)/ERK kinase; Raf, MAPK kinase kinase; Ras protein, member of the Ras GTPase family; Shc, SH2-containing collagen-related proteins (couples IGFR1 tyrosine kinase to Ras).


Fig. 2.

Fig. 2.The IRS-PI3K signaling cascade. Signaling through PI3K is central to a large number of processes in mammalian cells. In this greatly simplified diagram, one of the primary pathways leads to increased translation initiation and increased production of components of the transnational system. Also shown is the pathway for protection from apoptosis and that which can mediate increased cytoplasmic calcium levels. DAG-induced increases in PKC activity have the potential to feed back and prevent the phosphorylation of IRS-1. For clarity, potential interactions between the Ras/ERK pathway (Fig. 1) and calcineurin and G-protein receptor signaling have been omitted. Akt, protein kinase B; BAD, proapoptotic regulator of programmed cell death; Bcl2, regulator of programmed cell death, promotes cell survival; DAG, diacylglycerol; 4E-BP1, eukaryotic initiation factor 4 binding protein; eEF2, eukaryotic elongation factor-2 (k = kinase); GSK3, glycogen synthase kinase 3; IRS, insulin receptor substrate; mTOR, mammalian target of rapamycin; PI3-kinase (PI3K), phosphotidylinositol 3-kinase; PIP2, phosphatidylinositol 3,4-bisphosphate; PIP3, phosphatidylinositol 3,4,5-trisphosphate; PDK1, PI3K-dependent kinase; PKC, protein kinase C; S6K1, p70 S6 kinase.


It is important to note that the activities of the pathways depicted in Figs. 1 and 2 are conditional, i.e., the outcomes are based on a complex set of interactions yet to be comprehensively identified. For example, there are reports that both ERK and PI3K activity act in concert in some cell types (e.g., Refs. 62,117) and that both may be required for the differentiation of myoblasts (89). In contrast, others have reported that the activity of one pathway may actually inhibit the other (80,88, 117, 119). As an example of the conditional nature of the effects of signaling through the PI3K pathway, Chakravarthy et al. (19) recently reported that inhibition of PI3K signaling in satellite cell cultures can prevent the completion of the cell cycle, inducing arrest in the G1 phase. Under physiological conditions, G1 arrest would be expected to lead to either cell differentiation or apoptosis. The finding that inhibition of this signaling pathway stimulates cell cycle arrest would suggest that, under some conditions, signaling through PI3K is important for the continuation of cellular proliferation as opposed to the commonly ascribed differentiation response.

Interestingly, the processes of cellular proliferation (i.e., mitotic activity) and differentiation (i.e., expression of muscle-specific proteins) are generally thought to be mutually exclusive. In fact, in a number of cell types, activation of one of the two primary signaling pathways associated with ligation of growth factor receptors (e.g., Fig. 1 vs. Fig. 2) will generally inactivate portions of the other (80, 88). Among the well-characterized growth factors, IGF-I is relatively unique in that it has been reported to stimulate both proliferation and differentiation2, depending on timing and intracellular conditions (84, 108).

The intracellular signaling pathways that subserve IGFR1 ligation also represent potential points for interactions between IGF-I-induced responses and those initiated by other mediators. For example, there is evidence that signaling via G-protein receptors may interact with IGF-I receptor-related pathways modulating or even blocking some responses (e.g., Refs. 48, 58). There is also a growing body of data that suggests that there are interactions between the calcineurin- and IGF-I-signaling pathways in skeletal muscle (e.g., Refs. 29, 67). This is of particular interest in that calcineurin-IGF-I interactions would provide a another mechanism linking cellular calcium homeostasis to IGF-I signaling.

MUSCLE ADAPTATION TO INCREASED LOADING

There is evidence that the mitogenic and myogenic effects of IGF-I that render it useful for muscle regeneration might also be important for the adaptation of muscle to increased loading as well. A number of in vivo activity models, such as increased loading, stretch, and “eccentric contraction,” are known to result in increased IGF-I and/or IGF-I mRNA expression in muscle cells (2, 3, 9, 30, 46,86, 92, 97, 114, 115). Furthermore, experimental manipulations of muscle IGF-I levels have been shown to induce muscle hypertrophy both in vitro and in vivo (4, 24, 109). For example, overexpression (24) or direct infusion (4) of IGF-I in muscle results in hypertrophy, whereas inhibition of intracellular signaling components associated with IGFR1 ligation can prevent this response (14). Overexpression of IGF-I in muscle has also been shown to prevent some of the age-related effects on skeletal muscle, such as the decline in muscle mass (10,68). However, muscle IGF-I overexpression in a transgenic model did not prevent atrophy due to acute muscle unloading (27).

THE AUTOCRINE/PARACRINE IGF-I SYSTEM

One of the more interesting recent developments in the IGF-I story has been the identification of a unique IGF-I isoform that is expressed in response to changes in the loading state of skeletal muscles (115). This isoform, mechanogrowth factor (MGF), has been shown to be markedly upregulated in response to both stretch and increased loading (61, 70). It appears that skeletal muscles produce both a generalized tissue-type IGF-I (1) and the loading-sensitive MGF isoform with differing time courses, suggesting distinct roles for these two growth factor isoforms (70). Expression of both IGF-I and MGF appears to be very sensitive to the loading state of the muscle. For example, we recently found that IGF-I and MGF mRNA increased significantly within a few hours after a single bout of resistance-type exercise in rat muscles (unpublished observations).

In addition to IGF-I itself, there is evidence that cells in muscle produce other components of the IGF-I regulatory system (8, 12,60). For example, Awede et al. (8) reported that overloaded muscles in mice increased the expression of IGF binding protein 4 (IGFBP4) mRNA, whereas that of IGFBP5 was decreased. In contrast, unloading of mouse muscle resulted in an increase in IGFBP5 mRNA, whereas that of IGFBP4 was unchanged (8). In general, IGFBP4 and IGFBP5 would be expected to modulate the effects of IGF-I via regulation of the free IGF-I concentration in muscle and possibly via competition with IGF receptors for IGF-I (11). These findings provide further support of the idea that the autocrine/paracrine IGF-I system is active in skeletal muscle and sensitive to the loading state of the muscle.

THE “MYOGENIC” COMPONENT OF MUSCLE ADAPTATION

As with myofiber regeneration, a number of processes that IGF-I is known to stimulate would also promote skeletal muscle hypertrophy. The utility of the insulin-like anabolic effects for promoting muscle hypertrophy is obvious (71, 85). However, the importance of IGF-I-induced actions on muscle satellite cells may be less evident. In the case of the hypertrophy response, there appears to be a “myogenic” component wherein satellite cell-derived myoblasts are thought to fuse with existing myofibers much as they would with damaged but still viable myofibers after injury (22, 66, 87, 91,98). The importance of this response stems from the observations that mature mammalian skeletal muscle fibers appear to maintain a relatively finite, fiber type-specific relationship between the size of the myofiber and the number of myonuclei present in a given myofiber (5, 6, 23, 34, 43, 59, 63, 99, 102, 104). However, mammalian myofibers become permanently differentiated shortly after birth and cannot undergo mitotic division or directly increase their myonuclear number (i.e., myonuclear division) (22). The requirement for additional nuclei to support hypertrophy appears to be met via the proliferation, differentiation, and finally the fusion of muscle satellite cells or their progeny with the enlarging myofibers, providing the new myonuclei needed to support the hypertrophy process (5, 63, 72, 81, 82, 91) (Fig.3). Among the well-characterized growth factors, IGF-I is the only one that has been consistently reported to facilitate each of these processes.

Fig. 3.

Fig. 3.IGF-I and “myogenesis” during compensatory hypertrophy. Increased loading leads to satellite cell proliferation, differentiation, and fusion. IGF-I has been shown to stimulate these myogeninc processes in skeletal muscles. It is postulated that IGF-I, and/or the loading-sensitive IGF-I isoform mechanogrowth factor (MGF), is produced and released by myofibers in response to increased loading or stretch. The increased local concentration of IGF-I (MGF) would then stimulate the myogenic processes needed to drive the hypertrophy response.


Interestingly, relatively acute overexpression of IGF-I has been shown to increase the number of times that satellite cells can replicate, possibly explaining some of the palliative effects of this treatment on age-related changes in skeletal muscle cited above (19-21). However, chronic overexpression of IGF-I appears to exhaust the replicative capacity of satellite cell in vivo and thus does not prevent age-related declines in proliferation (21).

IGF-I AND EXCITATION-CONTRACTION COUPLING

In addition to the anabolic and myogenic effects attributed to IGF-I, this growth factor also appears to have the ability to modulate components of the excitation-contraction coupling mechanism in vivo. Skeletal muscle dihydropyridine receptors (DHPR) are L-type calcium channels that act as the voltage sensor in the transverse tubular system. The primary function of these L-type channels appears to be the detection of depolarization and the direct activation of the calcium release channels in the sarcoplasmic reticulum (49). Unlike the cardiac version of DHPR, the inward current carried by these channels in skeletal muscle does not appear to be important in the acute regulation of excitation contraction coupling (49). However, it is possible that the slow inward conductance of Ca2+ may have some function with regard to long-term intracellular calcium signaling. In cell culture, IGF-I induces an increase in DHPR that results in a significant increase in charge movement (112). In vivo, overexpression of IGF-I in muscle results in a significant increase in DHPR receptor concentration in both fast-twitch and slow-twitch skeletal muscles (74). In addition, IGF-I-induced increases in DHPR appear to ameliorate age-related effects on contractile function in mice (73). To date, the potential impacts of IGF-I on excitation-contraction coupling have received relatively less attention than the myogenic and anabolic aspects detailed above.

IGF-I AND PROINFLAMMATORY CYTOKINES

There have been reports that prolonged and/or intense exercise may result in significant increases in circulating levels of proinflammatory cytokines such as interleukin (IL)-6 and/or IL-1β (33, 64, 69, 100, 113). Interestingly, there are reports that exercise can increase proinflammatory cytokines and concurrently depress circulating IGF-I in children (35, 90). In general, the cellular and molecular effects of exercise-induced cytokine responses on the IGF-I system have not been extensively evaluated in the context of exercise. However, it is known that in disease states such as sepsis the elevated proinflammatory cytokine levels can either directly or indirectly mediate catabolic effects on skeletal muscle (36, 37, 39, 50, 51, 54, 55). In direct relation to the IGF-I system, Fan et al. (36) found that systemic injection of IL-1β or tumor necrosis factor-α (TNF-α) resulted in a reduction in muscle IGF-I in rats. In a similar finding, Lang et al. (50) reported that systemic sepsis is associated with a decline in skeletal muscle and plasma IGF-I. These authors found that blocking the IL-1 receptor prevented the decline of IGF-I in skeletal muscle and reduced the degree of IGF-I decrease seen in the plasma. The IL-1 blockade also prevented the 43% decrease in skeletal muscle protein synthesis-induced by the septic state. Thus it appears that the anti-anabolic and/or catabolic effects of cytokines such as IL-1β may be mediated at least in part via the IGF-I axis.

In addition to indirect effects such as a decrease in circulating IGF-I, there is evidence that some cytokines may interact with intracellular IGF-I receptor signaling. One area of intersection involves intracellular signaling via the Janus-activated kinases (JAK) and signal transducers and activators of transcription (STAT), which participate in cytokine signaling (47). Recent evidence suggests that the IGF-I receptor may also activate JAK/STAT signaling (41, 56, 103, 120). Among the targets for STATs are a family of suppressors of cytokine signaling, which act as part of a negative feedback loop to the cytokine receptors. This raises the possibility that elevated cytokine signaling could also feedback to and possibly inhibit the IGF-I receptor as well (31, 32, 103). This would provide an indirect mechanism for the inhibition of IGF-I signaling whereby increased proinflammatory cytokine levels might stimulate the production of suppressors of cytokine signaling, which would then feedback to both the cytokine and IGF-I receptors. It is also possible that proinflammatory cytokines modulate IGF-I less directly via increasing circulating corticosteroid levels (51). Both endogenous and exogenous glucocorticoids are known to modulate both IGF-I abundance and IGF-I effects in muscle (28, 36, 51, 95, 96).

In a seemingly paradoxical set of findings, it has been shown that the proinflammatory cytokines IL-6 and TNF-α stimulate the proliferation of satellite cells or myoblasts in vitro (7, 18, 101,111). This suggests that there may be a role for components of the inflammatory response in muscle adaptation (111). It is tempting to speculate that a potential role of inflammatory responses in muscle adaptation may be a function of the degree of response and as such that this may be one of the mechanisms that separates training from overtraining (97).

SUMMARY

The continued interest in the role of IGF-I in skeletal muscle adaptation is founded on the extensive body of evidence indicating that1) IGF-I is both anabolic and mitogenic for skeletal muscle or muscle lineage cells, 2) IGF-I operates in an autocrine/paracrine mode in skeletal muscle, and 3) muscle IGF-I and MGF production are sensitive to increases in loading state. In addition to the effects of IGF-I in promoting skeletal muscle hypertrophy or regeneration, there are a number of other systems that may impact or be impacted by IGF-I signaling that should be considered by muscle researchers. Future challenges in this area include the identification of the cellular level mechanisms that transduce mechanical signals leading to changes in IGF-I signaling and elucidation of the relationships between the various intracellular signaling pathways that allow IGF-I signaling to stimulate the competing processes of cellular differentiation and cellular proliferation.

FOOTNOTES

  • Address for reprint requests and other correspondence: G. R. Adams, Univ. of California Irvine, Dept. of Physiology & Biophysics, 346-D Medical Sciences 1, Irvine, CA 92697-4560 (E-mail:).

  • 1There is evidence that multiple muscle stem cell populations may be contributing to processes traditionally ascribed to satellite cells (e.g., Refs. 26, 118).

  • 2A number of growth factors, such as FGF-6 and hepatocyte growth factor, are being actively investigated in this respect, but to date the results have not proved conclusive in in vivo models.

  • The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • 10.1152/japplphysiol.01264.2001

REFERENCES

  • 1 Adamo ML, Neuenschwander S, LeRoith D, Roberts CT.Structure, expression, and regulation of the IGF-I gene.Adv Exp Med Biol3431993111
    PubMed | Google Scholar
  • 2 Adams GR, Haddad F.The relationships between IGF-1, DNA content, and protein accumulation during skeletal muscle hypertrophy.J Appl Physiol81199625092516
    Link | ISI | Google Scholar
  • 3 Adams GR, Haddad F, Baldwin KM.Time course of changes in markers of myogenesis in overloaded rat skeletal muscles.J Appl Physiol87199917051712
    Link | ISI | Google Scholar
  • 4 Adams GR, McCue SA.Localized infusion of IGF-I results in skeletal muscle hypertrophy in rats.J Appl Physiol84199817161722
    Link | ISI | Google Scholar
  • 5 Allen DL, Monke SR, Talmadge RJ, Roy RR, Edgerton VR.Plasticity of myonuclear number in hypertrophied and atrophied mammalian skeletal muscle fibers.J Appl Physiol78199519691976
    Link | ISI | Google Scholar
  • 6 Allen DL, Yasui W, Tanaka T, Ohira Y, Nagaoka S, Sekiguchi C, Hinds WE, Roy RR, Edgerton VR.Myonuclear number and myosin heavy chain expression in rat soleus single muscle fibers after spaceflight.J Appl Physiol811996145151
    Link | ISI | Google Scholar
  • 7 Austin L, Bower J, Kurek J, Vakakis N.Effects of leukaemia inhibitory factor and other cytokines on murine and human myoblast proliferation.J Neurol Sci1121992185191
    Crossref | PubMed | ISI | Google Scholar
  • 8 Awede B, Thissen J, Gailly P, Lebacq J.Regulation of IGF-I, IGFBP-4 and IGFBP-5 gene expression by loading in mouse skeletal muscle.FEBS Lett4611999263267
    Crossref | PubMed | ISI | Google Scholar
  • 9 Bamman MM, Shipp JR, Jiang J, Gower BA, Hunter GR, Goodman A, McLafferty CL, Urban RJ.Mechanical load increases muscle IGF-I and androgen receptor mRNA concentrations in humans.Am J Physiol Endocrinol Metab2802001E383E390
    Link | ISI | Google Scholar
  • 10 Barton-Davis ER, Shoturma DI, Musaro A, Rosenthal N, Sweeney HL.Viral mediated expression of insulin-like growth factor I blocks the aging-related loss of skeletal muscle function.Proc Natl Acad Sci USA9519981560315607
    Crossref | PubMed | ISI | Google Scholar
  • 11 Baxter RC.Insulin-like growth factor (IGF)-binding proteins: interactions with IGFs and intrinsic bioactivities.Am J Physiol Endocrinol Metab2782000E967E976
    Link | ISI | Google Scholar
  • 12 Bayol S, Loughna PT, Brownson C.Phenotypic expression of IGF binding protein transcripts in muscle, in vitro and in vivo.Biochem Biophys Res Commun2732000282286
    Crossref | PubMed | ISI | Google Scholar
  • 13 Benito M, Valverde AM, Lorenzo M.IGF-1: a mitogen also involved in differentiation processes in mammalian cells.Int J Biochem Cell Biol281996499510
    Crossref | PubMed | ISI | Google Scholar
  • 14 Bodine SC, Stitt TN, Gonzalez M, Kline WO, Stover GL, Bauerlein R, Zlotchenko E, Scrimgeour A, Lawrence JC, Glass DJ, Yancopoulos GD.Akt/mTOR pathway is a crucial regulator of skeletal muscle hypertrophy and can prevent muscle atrophy in vivo.Nat Cell Biol3200110141019
    Crossref | PubMed | ISI | Google Scholar
  • 15 Butler AA, LeRoith D.Control of growth by the somatropic axis: growth hormone and the insulin-like growth factors have related and independent roles.Annu Rev Physiol632001161164
    Crossref | ISI | Google Scholar
  • 16 Calera MR, Pilch PF.Induction of Akt-2 correlates with differentiation in Sol8 muscle cells.Biochem Biophys Res Commun2511998835841
    Crossref | PubMed | ISI | Google Scholar
  • 17 Canicio J, Gallardo E, Illa I, Testar X, Palacin M, Zorzano A, Kaliman P.p70 S6 kinase activation is not required for insulin-like growth factor-induced differentiation of rat, mouse or human skeletal muscle cells.Endocrinology139199850425049
    Crossref | PubMed | ISI | Google Scholar
  • 18 Cantini M, Massimino ML, Rapizzi E, Rossini K, Catani C, Libera LD, Carraro U.Human satellite cell proliferation in vitro is regulated by autocrine secretion of IL-6 stimulated by a soluble factor(s) released by activated monocytes.Biochem Biophys Res Commun21619954953
    Crossref | PubMed | ISI | Google Scholar
  • 19 Chakravarthy MV, Abraha TW, Schwartz RJ, Fiorotto ML, Booth FW.Insulin-like growth factor-I extends in vitro replicative life span of skeletal muscle satellite cells by enhancing G1/S cell cycle progression via the activation of phosphatidylinositol 3′-kinase/Akt signaling pathway.J Biol Chem27520003594235952
    Crossref | PubMed | ISI | Google Scholar
  • 20 Chakravarthy MV, Davis BS, Booth FW.IGF-I restores satellite cell proliferative potential in immobilized old skeletal muscle.J Appl Physiol89200013651379
    Link | ISI | Google Scholar
  • 21 Chakravarthy MV, Fiorotto ML, Schwartz RJ, Booth FW.Long-term insulin-like growth factor-I expression in skeletal muscles attenuates the enhanced in vitro proliferation ability of the resident satellite cells in transgenic mice.Mech Ageing Dev122200113031320
    Crossref | ISI | Google Scholar
  • 22 Chambers RL, McDermott JC.Molecular basis of skeletal muscle regeneration.Can J Appl Physiol211996155184
    Crossref | PubMed | Google Scholar
  • 23 Cheek DB, Holt AB, Hill DE, Talbert JL.Skeletal muscle mass and growth: the concept of the deoxyribonucleic acid unit.Pediatr Res51971312328
    Crossref | ISI | Google Scholar
  • 24 Coleman ME, DeMayo F, Yin KC, Lee HM, Geske R, Montgomery C, Schwartz RJ.Myogenic vector expression of insulin-like growth factor I stimulates muscle cell differentiation and myofiber hypertrophy in transgenic mice.J Biol Chem27019951210912116
    Crossref | PubMed | ISI | Google Scholar
  • 25 Coolican SA, Samuel DS, Ewton DZ, McWade FJ, Florini JR.The mitogenic and myogenic actions of insulin-like growth factors utilize distinct signaling pathways.J Biol Chem272199766536662
    Crossref | PubMed | ISI | Google Scholar
  • 26 Creuzet S, Lescaudron L, Li Z, Fontaine-Perus J.MyoD, myogenin, and desmin-nls-lacZ transgene emphasize the distinct patterns of satellite cell activation in growth and regeneration.Exp Cell Res2431998241253
    Crossref | PubMed | ISI | Google Scholar
  • 27 Criswell DS, Booth FW, DeMayo F, Schwartz RJ, Gordon SE, Fiorotto ML.Overexpression of IGF-I in skeletal muscle of transgenic mice does not prevent unloading-induced atrophy.Am J Physiol Endocrinol Metab2751998E373E379
    Link | ISI | Google Scholar
  • 28 Dardevet D, Sornet C, Savary I, Debras E, Patureau-Mirand P, Grizard J.Glucocorticoid effects on insulin- and IGF-I-regulated muscle protein metabolism during aging.J Endocrinol15619988389
    Crossref | PubMed | ISI | Google Scholar
  • 29 Delling U, Tureckova J, Lim HW, De Windt LJ, Rotwein P, Molkentin JD.A calcineurin-NFATc3-dependent pathway regulates skeletal muscle differentiation and slow myosin heavy-chain expression.Mol Cell Biol20200066006611
    Crossref | PubMed | ISI | Google Scholar
  • 30 DeVol DL, Rotwein P, Sadow JL, Novakofski J, Bechtel PJ.Activation of insulin like growth factor gene expression during work induced skeletal muscle growth.Am J Physiol Endocrinol Metab2591990E89E95
    Link | ISI | Google Scholar
  • 31 Dey BR, Furlanetto RW, Nissley P.Suppressor of cytokine signaling (SOCS)-3 protein interacts with the insulin-like growth factor-I receptor.Biochem Biophys Res Commun27820003843
    Crossref | ISI | Google Scholar
  • 32 Dey BR, Spence SL, Nissley P, Furlanetto RW.Interaction of human suppressor of cytokine signaling (SOCS)-2 with the insulin-like growth factor-I receptor.J Biol Chem27319982409524101
    Crossref | ISI | Google Scholar
  • 33 Drenth JPH, Van Uum SHM, Van Deuren M, Pesman GJ, Van Der Ven-Jongekrijg J, Van Der Meer JWMEndurance run increases circulating IL-6 and IL-1re but downregulates ex vivo TNF-α and IL-1β production.J Appl Physiol79199514971503
    Link | ISI | Google Scholar
  • 34 Edgerton VR, Roy RR.Regulation of skeletal muscle fiber size, shape and function.J Biomech241991123133
    Crossref | ISI | Google Scholar
  • 35 Eliakim A, Scheett TP, Newcomb R, Mohan S, Cooper DM.Fitness, training, and the growth hormone insulin-like growth factor I axis in prepubertal girls.J Clin Endocrinol Metab86200127972802
    ISI | Google Scholar
  • 36 Fan J, Wojnar MM, Theodorakis M, Lang CH.Regulation of insulin like growth factor (IGF)-1 mRNA and peptide and IGF-binding proteins by interleukin-1.Am J Physiol Regul Integr Comp Physiol2701996R621R629
    Link | ISI | Google Scholar
  • 37 Fang CH, Li BG, James H, Fischer JE, Hasselgren PO.Cytokines block the effects of insulin-like growth factor-1 on glucose uptake and lactate production in skeletal muscle but do not influence IGF-I-induced changes in protein turnover.Shock81997362367
    Crossref | PubMed | ISI | Google Scholar
  • 38 Florini JR, Ewton DZ, Coolican SA.Growth hormone and insulin like growth factor system in myogenesis.Endocr Rev171996481517
    PubMed | ISI | Google Scholar
  • 39 Garcia-Martinez C, Llovera M, Agell N, Lopez-Soriano FJ, Argiles JM.Ubiquitin gene expression in skeletal muscle is increased during sepsis: Involvement of TNF-α but not IL-1.Biochem Biophys Res Commun2171995839844
    Crossref | PubMed | ISI | Google Scholar
  • 40 Goldberg AL.Work induced growth of skeletal muscle in normal and hypophysectomized rats.Am J Physiol213196711931198
    Link | ISI | Google Scholar
  • 41 Gual P, Baron V, Lequoy V, Van Obberghen E.Interaction of janus kinases JAK-1 and JAK-2 with the insulin receptor and the insulin-like growth factor-1 receptor.Endocrinology1391998884893
    Crossref | ISI | Google Scholar
  • 42 Gulati AK.The effect of X-irradiation on skeletal muscle regeneration in the adult rat.J Neurol Sci781987111120
    Crossref | PubMed | ISI | Google Scholar
  • 43 Hikida RS, Van Nostran S, Murray JD, Staron RS, Gordon SE, Kraemer WJ.Myonuclear loss in atrophied soleus muscle fibers.Anat Rec2471997350354
    Crossref | PubMed | Google Scholar
  • 44 Jennische E, Hansson HA.Regenerating skeletal muscle cells express insulin-like growth factor 1.Acta Physiol Scand1301987327332
    Crossref | PubMed | Google Scholar
  • 45 Jennische E, Skottner A, Hansson HA.Satellite cells express the trophic factor IGF-1 in regenerating skeletal muscle.Acta Physiol Scand1291987915
    Crossref | PubMed | Google Scholar
  • 46 Keller HL, St B. Pierre Schneider Eppihimer LA, Cannon JG.Association of IGF-I and IGF-II with myofiber regeneration in vivo.Muscle Nerve221999347354
    Crossref | PubMed | ISI | Google Scholar
  • 47 Kelso A.Cytokines: principles and prospects.Immunol Cell Biol761998300317
    Crossref | ISI | Google Scholar
  • 48 Kim S, Jee K, Kim D, Koh H, Chung J.Cyclic AMP inhibits Akt activity by blocking the membrane localization of PDK1.J Biol Chem27620011286412870
    Crossref | PubMed | ISI | Google Scholar
  • 49 Lamb GD.Excitation-contraction coupling in skeletal muscle: comparisons with cardiac muscle.Clin Exp Pharmacol Physiol272000216224
    Crossref | PubMed | ISI | Google Scholar
  • 50 Lang CH, Fan J, Coney R, Vary TC.IL-1 receptor antagonist attenuates sepsis-induced alterations in the IGF system and protein synthesis.Am J Physiol Endocrinol Metab2701996E340E437
    Link | ISI | Google Scholar
  • 51 Lang CH, Fan J, Wojnar MM, Vary TC, Cooney R.Role of central IL-1 in regulating peripheral IGF-I during endotoxemia and sepsis.Am J Physiol Regul Integr Comp Physiol2741998R956R962
    Link | ISI | Google Scholar
  • 52 Lawlor MA, Rotwein P.Insulin-like growth factor-mediated muscle cell survival: central roles for Akt and cyclin-dependent kinase inhibitor p21.Mol Cell Biol20200089838995
    Crossref | PubMed | ISI | Google Scholar
  • 53 LeFaucheur JP, Sebille A.Muscle regeneration following injury can be modified in vivo by immune neutralization of fibroblast growth factor, transforming growth factor β1 or insulin-like growth factor I.J Neuroimmunol5719958591
    Crossref | PubMed | ISI | Google Scholar
  • 54 Ling PR, Schwartz JH, Bistrian BR.Mechanisms of host wasting induced by administration of cytokines in rats.Am J Physiol Endocrinol Metab2721997E333E339
    Link | ISI | Google Scholar
  • 55 Llovera M, Garcia-Martinez C, Agell N, Lopez-Soriano F, Argiles JM.TNF can directly induce the expression of ubiquitin-dependent proteolytic system in rat soleus muscles.Biochem Biophys Res Commun2301997238241
    Crossref | PubMed | ISI | Google Scholar
  • 56 Lopaczynski W.Differential regulation of signaling pathways for insulin and insulin-like growth factor I.Acta Biochim Pol4619995161
    ISI | Google Scholar
  • 57 Luque E, Pena J, Salas P, Martin JD.Changes in satellite cell population associated with regenerating muscle fibers in rats.J Submicrosc Cytol Pathol281996305311
    Google Scholar
  • 58 Malbon CC, Karoor V.G-protein-linked receptors as tyrosine kinase substrates: New paradigms in signal integration.Cell Signal101998523527
    Crossref | PubMed | ISI | Google Scholar
  • 59 McCall GE, Allen DL, Linderman JK, Grindeland RE, Roy RR, Mukku VR, Edgerton VR.Maintenance of myonuclear domain size in rat soleus after overload and growth hormone/IGF-I treatment.J Appl Physiol84199814071412
    Link | ISI | Google Scholar
  • 60 McCusker RH, Clemmons DR.Effects of cytokines on insulin-like growth factor binding protein secretion by muscle cells in vitro.Endocrinology134199420952102
    Crossref | ISI | Google Scholar
  • 61 McKoy G, Ashley W, Mander J, Yang SY, Williams N, Russell B, Goldspink G.Expression of insulin growth factor-1 splice variants and structural genes in rabbit skeletal muscle induced by stretch and stimulation.J Physiol5161999583592
    Crossref | PubMed | ISI | Google Scholar
  • 62 Mehrhof FB, Muller FU, Bergmann MW, Li P, Wang Y, Schmitz W, Dietz R, von Harsdorf R.In cardiomyocyte hypoxia, insulin-like growth factor-I-induced antiapoptotic signaling requires phosphatidylinositol-3-OH-kinase-dependent and mitogen-activated protein kinase-dependent activation of the transcription factor cAMP response element-binding.Circulation104200120882094
    Crossref | PubMed | ISI | Google Scholar
  • 63 Mitchell PO, Pavlath GK.A muscle precursor cell-dependent pathway contributes to muscle growth after atrophy.Am J Physiol Cell Physiol2812001C1706C1715
    Link | ISI | Google Scholar
  • 64 Moldoveanu AI, Shephard RJ, Shek PN.Exercise elevates plasma levels but not gene expression of IL-1β, IL-6 and TNF-α in blood mononuclear cells.J Appl Physiol89200014991504
    Link | ISI | Google Scholar
  • 65 Molnar G, Ho ML, Schroedl NA.Evidence for multiple satellite cell populations and a non-myogenic cell type that is regulated differently in regenerating and growing skeletal muscle.Tissue Cell281996547556
    Crossref | PubMed | ISI | Google Scholar
  • 66 Moss FP, Leblond CP.Satellite cells as the source of nuclei in muscles of growing rats.Anat Record1701971421436
    Crossref | PubMed | Google Scholar
  • 67 Musarò A, McCullagh KJ, Naya FJ, Olson EN, Rosenthal N.IGF-1 induces skeletal myocyte hypertrophy through calcineurin in association with GATA-2 and NF-ATc1.Nature4001999581584
    Crossref | PubMed | ISI | Google Scholar
  • 68 Musarò A, McCullagh K, Paul A, Houghton L, Dobrowolny G, Molinaro M, Barton ER, Sweeney HL, Rosenthal N.Localized Igf-1 transgene expression sustains hypertrophy and regeneration in senescent skeletal muscle.Nat Genet272001195200
    Crossref | PubMed | ISI | Google Scholar
  • 69 Nieman DC.Immune response to heavy exertion.J Appl Physiol82199713851394
    Link | ISI | Google Scholar
  • 70 Owino V, Yang SY, Goldspink G.Age-related loss of skeletal muscle function and the inability to express the autocrine form of insulin-like growth factor-1 (MGF) in response to mechanical overload.FEBS Lett5052001259263
    Crossref | PubMed | ISI | Google Scholar
  • 71 Petley T, Graff K, Jiang W, Florini J.Variation among cell types in the signaling pathways by which IGF-I stimulates specific cellular responses.Horm Metab Res3119997076
    Crossref | PubMed | ISI | Google Scholar
  • 72 Phelan JN, Gonyea WJ.Effect of radiation on satellite cell activity and protein expresion in overloaded mammalian skeletal muscle.Anat Rec2471997179188
    Crossref | PubMed | Google Scholar
  • 73 Renganathan M, Messi ML, Delbono O.Over expression of IGF-1 exclusively in skeletal muscle prevents age-related decline in the number of dihydropyridine receptors.J Biol Chem27319982884528851
    Crossref | PubMed | ISI | Google Scholar
  • 74 Renganathan M, Messi ML, Schwartz R, Delbono O.Over expression of IGF-1 exclusively in skeletal muscle increases the number of dihydropyridine receptors in adult transgenic mice.FEBS Lett41719971316
    Crossref | PubMed | ISI | Google Scholar
  • 75 Robertson TA, Grounds MD, Papadimitriou JM.Elucidation of aspects of murine skeletal muscle regeneration using local and whole body irradiation.J Anat1811992265276
    PubMed | ISI | Google Scholar
  • 77 Robertson TA, Papadimitriou JM, Grounds MD.Fusion of myogenic cells to the newly sealed region of damaged myofibers in skeletal muscle regeneration.Neuropathol Appl Neurobiol191993350358
    Crossref | PubMed | ISI | Google Scholar
  • 78 Robertson TA, Papadimitriou JM, Grounds MD.Fusion between a myogenic cell in the satellite cell position and undamaged adult myofiber segments.Experientia481992394395
    Crossref | Google Scholar
  • 79 Rommel C, Bodine SC, Clarke BA, Rossman R, Nunez L, Stitt TN, Yancopoulos GD, Glass DJ.Mediation of IGF-1-induced skeletal myotube hypertrophy by PI(3)K/Akt/mTOR and PI(3)K/Akt/GSK3 pathways.Nat Cell Biol3200110091013
    Crossref | PubMed | ISI | Google Scholar
  • 80 Rommel C, Clarke BA, Zimmermann S, Nuñez L, Rossman R, Reid K, Moelling K, Yancopoulos GD, Glass DJ.Differentiation stage-specific inhibition of the Raf-MEK-ERK pathway by Akt.Science286199917381741
    Crossref | PubMed | ISI | Google Scholar
  • 81 Rosenblatt JD, Parry DJ.Adaptation of rat extensor digitorum longus muscle to gamma irradiation and overload.Pflügers Arch4231993255264
    Crossref | PubMed | ISI | Google Scholar
  • 82 Rosenblatt JD, Yong D, Parry DJ.Satellite cell activity is required for hypertrophy of overloaded adult rat skeletal muscle.Muscle Nerve171994608613
    Crossref | PubMed | ISI | Google Scholar
  • 83 Rosenthal SM, Brown EJ, Brunetti A, Goldfine ID.Fibroblast growth factor inhibits insulin like growth factor-II (IGF-II) gene expression and increases IGF-I receptor abundance.Mol Endocrinol51991678684
    Crossref | Google Scholar
  • 84 Rosenthal SM, Cheng ZQ.Opposing early and late effects of insulin-like growth factor I on differentiation and the cell cycle regulatory retinoblastoma protein in skeletal myoblasts.Proc Natl Acad Sci USA9219951030710311
    Crossref | PubMed | ISI | Google Scholar
  • 85 Russell-Jones DL, Umpleby M.Protein anabolic action of insulin, growth hormone and insulin-like growth factor-1.Eur J Endocrinol1351996631642
    Crossref | PubMed | ISI | Google Scholar
  • 86 Sakuma K, Watanabe K, Totsuka T, Uramoto I, Sano M, Sakamoto K.Differential adaptations of insulin-like growth factor-I, basic fibroblast growth factor, and leukemia inhibitory factor in the plantaris muscle of rats by mechanical overloading: an immunohistochemical study.Acta Neuropathol (Berl)951998123130
    Crossref | PubMed | ISI | Google Scholar
  • 87 Salleo A, LaSpada G, Falzea G, Denaro MG, Cicciarello R.Response of satellite cells and muscle fibers to long-term compensatory hypertrophy.J Submicrosc Cytol Pathol151983929940
    Google Scholar
  • 88 Samuel DS, Ewton DZ, Coolican SA, Petley TD, McWade FJ, Florini JR.Raf-1 activation stimulates proliferation and inhibits IGF-stimulated differentiation in L6A1 myoblasts.Horm Metab Res3119995564
    Crossref | ISI | Google Scholar
  • 89 Sarbassov DD, Peterson CA.Insulin receptor substrate-1 and phosphatidylinositol 3-kinase regulate extracellular signal-regulated kinase-dependent and -independent signaling pathways during myogenic differentiation.Mol Endocrinol12199818701878
    Crossref | Google Scholar
  • 90 Scheett TP, Mills PJ, Ziegler MG, Stoppani J, Cooper DM.Effect Of exercise on cytokines and growth mediators in prepubertal children.Pediatr Res461999429434
    Crossref | ISI | Google Scholar
  • 91 Schiaffino S, Pierobon Bormioli S, Aloisi M.The fate of newly formed satellite cells during compensatory muscle hypertrophy.Virchows ArchB211976113118
    Google Scholar
  • 92 Schlechter NL, Russell SM, Spencer EM, Nicoll CS.Evidence suggesting that the growth promoting effect of GH on cartilage in vivo is mediated by local production of somatomedia.Proc Natl Acad Sci USA83198679237934
    Crossref | ISI | Google Scholar
  • 93 Schultz E.Satellite cell behavior during skeletal muscle growth and regeneration.Med Sci Sports Exerc211989S181S186
    Crossref | PubMed | ISI | Google Scholar
  • 94 Schultz E, McCormick KM.Skeletal muscle satellite cells.Rev Physiol Biochem Pharmacol1231994213257
    Crossref | PubMed | ISI | Google Scholar
  • 95 Singleton JR, Baker BL, Thorburn A.Dexamethasone inhibits insulin-like growth factor signaling and potentiates myoblast apoptosis.Endocrinology141200029452950
    Crossref | PubMed | ISI | Google Scholar
  • 96 Singleton JR, Feldman EL.Insulin-like growth factor-I in muscle metabolism and myotherapies.Neurobiol Dis82001541554
    Crossref | PubMed | ISI | Google Scholar
  • 97 Smith LL.Cytokine hypothesis of overtraining: a physiological adaptation to excessive stress?Med Sci Sports Exerc322000317331
    Crossref | PubMed | ISI | Google Scholar
  • 98 Snow MH.Satellite cell response in rat soleus muscle undergoing hypertrophy due to surgical ablation of synergists.Anat Rec2271990437446
    Crossref | PubMed | Google Scholar
  • 99 Snow MH.Myogenic cell formation in regenerating rat skeletal muscle injured by mincing.Anat Rec1881976181200
    Crossref | Google Scholar
  • 100 Sprenger H, Jacobs C, Nain M, Gressner AM, Prinz H, Wesemann W, Gemsa D.Enhanced release of cytokines, interleukin-2 receptors, and neopterin after long-distance running.Clin Immunol Immunopathol631992188195
    Crossref | PubMed | Google Scholar
  • 101 Szalay K, Razga Z, Duda E.TNF inhibits myogenesis and downregulates the expression of myogenic regulatory factors myoD and myogenin.Eur J Cell Biol741997391398
    PubMed | ISI | Google Scholar
  • 102 Taipale J, Keski-Oja J.Growth factors in the extracellular matrix.FASEB J1119975159
    Crossref | PubMed | ISI | Google Scholar
  • 103 Takahashi T, Fukuda K, Pan J, Kodama H, Sano M, Makino S, Kato T, Manabe T, Ogawa S.Characterization of insulin-like growth factor-I-induced activation of the JAK/STAT pathway in rat cardiomyocytes.Circ Res851999884891
    Crossref | ISI | Google Scholar
  • 104 Talmadge RJ, Roy RR, Chalmers GR, Edgerton VR.MHC and sarcoplasmic reticulum protein isoforms in functionally overloaded cat plantaris muscle fibers.J Appl Physiol80199612961303
    Link | ISI | Google Scholar
  • 105 Tamir Y, Bengal E.Phosphoinositide 3-kinase induces the transcriptional activity of MEF2 proteins during muscle differentiation.J Biol Chem27520003442434432
    Crossref | PubMed | ISI | Google Scholar
  • 106 Thomas G, Hall MN.TOR signaling and control of cell growth.Curr Opin Cell Biol91997782787
    Crossref | PubMed | ISI | Google Scholar
  • 107 Tureckova J, Wilson EM, Cappalonga JL, Rotwein P.Insulin-like growth factor-mediated muscle differentiation. Collaboration between phosphatidylinositol 3-kinase-akt-signaling pathways and myogenin.J Biol Chem27620013926439270
    Crossref | PubMed | ISI | Google Scholar
  • 108 Valentinis B, Baserga R.IGF-I receptor signalling in transformation and differentiation.Mol Pathol542001133137
    Crossref | Google Scholar
  • 109 Vandenburgh HH, Karlisch P, Shansky J, Feldstein R.Insulin and IGF-I induce pronounced hypertrophy of skeletal myofibers in tissue culture.Am J Physiol Cell Physiol2601991C475C484
    Link | ISI | Google Scholar
  • 110 Vandromme M, Rochat A, Meier R, Carnac G, Besser D, Hemmings BA, Fernandez A, Lamb NJ.Protein kinase B beta/Akt2 plays a specific role in muscle differentiation.J Biol Chem276200181738179
    Crossref | PubMed | ISI | Google Scholar
  • 111 Vierck J, O'Reilly B, Hossner K, Antonio J, Byrne K, Bucci L, Dodson M.Satellite cell regulation following myotrauma caused by resistance exercise.Cell Biol Int242000263272
    Crossref | PubMed | ISI | Google Scholar
  • 112 Wang ZM, Messi ML, Renganathan M, Delbono O.Insulin-like growth factor-1 enhances rat skeletal muscle charge movement and L-type Ca2+ channel gene expression.J Physiol5161999331341
    Crossref | PubMed | ISI | Google Scholar
  • 113 Weinstock C, Konig D, Harnschmacher R, Keul J, Berg A, Northoff H.Effect of exhaustive exercise stress on the cytokine response.Med Sci Sports Exerc291997345354
    Crossref | PubMed | ISI | Google Scholar
  • 114 Yan Z, Biggs RB, Booth FW.Insulin-like growth factor immunoreactivity increases in muscle after acute eccentric contractions.J Appl Physiol741993410414
    Link | ISI | Google Scholar
  • 115 Yang S, Alnaqeeb M, Simpson H, Goldspink G.Cloning and characterization of an IGF-1 isoform expressed in skeletal muscle subjected to stretch.J Muscle Res Cell Motil171996487495
    Crossref | PubMed | ISI | Google Scholar
  • 116 Yarasheski KE.Growth hormone effects on metabolism, body composition, muscle mass, and strength.Exerc Sport Sci Rev221994285312
    Crossref | PubMed | Google Scholar
  • 117 Yu CF, Roshan B, Liu ZX, Cantley LG.ERK regulates the hepatocyte growth factor-mediated interaction of Gab1 and the phosphatidylinositol 3-kinase.J Biol Chem27620013225232258
    Crossref | ISI | Google Scholar
  • 118 Zammit P, Beauchamp J.The skeletal muscle satellite cell: stem cell or son of stem cell?Differentiation682001193204
    Crossref | PubMed | ISI | Google Scholar
  • 119 Zimmermann S, Moelling K.Phosphorylation and regulation of Raf by Akt (protein kinase B).Science286199917411744
    Crossref | PubMed | ISI | Google Scholar
  • 120 Zong C, Chan J, Levy DE, Horvath C, Sadowski HB, Wang L.Mechanism of STAT3 activation by insulin-like growth factor I receptor.J Biol Chem27520001509915105
    Crossref | PubMed | ISI | Google Scholar