Top Banner
Send Orders of Reprints at [email protected] 560 Current Protein and Peptide Science, 2012, 13, 560-569 1389-2037/12 $58.00+.00 © 2012 Bentham Science Publishers Angiotensin II: Role in Skeletal Muscle Atrophy Claudio Cabello-Verrugio 1,* , Gonzalo Córdova 2 and José Diego Salas 1 1 Laboratorio de Biología y Fisiopatología Molecular, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas & Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile; 2 Centro de Regulación Celular y Pa- tología (CRCP), Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, Pontificia Universidad Católica de Chile, Santiago, Chile Abstract: Skeletal muscle, the main protein reservoir in the body, is a tissue that exhibits high plasticity when exposed to changes. Muscle proteins can be mobilized into free amino acids when skeletal muscle wasting occurs, a process called skeletal muscle atrophy. This wasting is an important systemic or local manifestation under disuse conditions (e.g., bed rest or immobilization), in starvation, in older adults, and in several diseases. The molecular mechanisms involved in muscle wasting imply the activation of specific signaling pathways which ultimately manage muscle responses to modu- late biological events such as increases in protein catabolism, oxidative stress, and cell death by apoptosis. Many factors have been involved in the generation and maintenance of atrophy in skeletal muscle, among them angiotensin II (Ang-II), the main peptide of renin-angiotensin system (RAS). Together with Ang-II, the angiotensin–converting enzyme (ACE) and the Ang-II receptor type 1 (AT-1 receptor) are expressed in skeletal muscle, forming an important local axis that can regulate its function. In many of the conditions that lead to muscle wasting, there is an impairment of RAS in a global or local fashion. At this point, there are several pieces of evidence that suggest the participation of Ang-II, ACE, and AT-1 receptor in the generation of skeletal muscle atrophy. Interestingly, the Ang-II participation in muscle atrophy is strongly ligated to the regulation of hypertrophic activity of factors such as insulin-like growth factor 1 (IGF-1). In this article, we reviewed the current state of Ang-II and RAS function on skeletal muscle wasting and its possible use as a therapeutic tar- get to improve skeletal muscle function under atrophic conditions. Keywords: angiotensin II, skeletal muscle atrophy, angiotensin–converting enzyme (ACE), angiotensin II receptor type 1 (AT- 1), AT-1 receptor blocker (ARB), atrogin-1, MuRF-1. INTRODUCTION Skeletal muscle atrophy corresponds to the loss of mus- cle mass produced by muscle wasting concomitantly with a decrease of muscle strength associated with some pathologi- cal conditions. This wasting is an important systemic or local manifestation under acute pathological conditions, such as disuse by immobilization, post-operatory recovering process, microgravity, and starvation, or under chronic conditions, such as aging, a sedentary lifestyle, or several diseases (can- cer, diabetes, AIDS, and cardiac, lung, or renal failure). All of them correspond to an increasing problem in our society [1, 2]. Under these conditions, the catabolism of proteins is exaggerated and the oxidative stress are increased, leading to a decrease in muscle mass [3]. One of the modulators of skeletal muscle function is the renin-angiotensin system (RAS) axis. The main effector of RAS is angiotensin II (Ang-II), a peptide that participates in the control of blood pressure, sodium balance, fluids volume, and some other hemodynamic features. In addition, Ang-II plays a key role in the regulation of skeletal muscle function *Address correspondence to this author at the Laboratorio de Biología y Fisiopatología Molecular, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas & Facultad de Medicina, Universidad Andres Bello, Santiago, Chile. Av. Republica 239, Postal Code 8370146, Santiago, Chile; E-mail: [email protected] by modulating the damage and contractile activity in muscle disease, such as Duchenne muscular dystrophy [4, 5]. How- ever, the main deleterious effect of Ang-II on skeletal muscle is that by inducing changes in enzyme activation, cell oxida- tive state, and gene expression, loss of muscle mass results. All of these changes alter the normal muscle remodeling process, leading to skeletal muscle wasting. Many of the conditions that contribute to skeletal muscle atrophy, such as chronic heart failure (CHF) and chronic kidney disease (CKD), are accompanied by an increase in the activation of the RAS axis, hence promoting an increase in the circulating Ang-II, while the loss of muscle mass worsens [6]. In this review, we address some of the main topics in relation with systemic or local muscle-RAS axis, skeletal muscle atrophy, and the cellular and molecular mechanisms underlying this process. EXPRESSION OF RENIN-ANGIOTENSIN SYSTEM COMPONENTS IN SKELETAL MUSCLE The identification of the local RAS axis has been de- scribed in several tissues [7]. In addition to Ang-II, the local RAS axis is composed of the angiotensin–converting enzyme (ACE) and two membrane receptors for Ang-II called Ang-II receptors type 1 (AT-1) and type 2 (AT-2) [8]. This section reviewed the evidence that support the expression of RAS components in skeletal muscle.
10

Angiotensin II: Role in Skeletal Muscle Atrophy

Apr 28, 2023

Download

Documents

Beatrice Caseau
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Angiotensin II: Role in Skeletal Muscle Atrophy

Send Orders of Reprints at [email protected]

560 Current Protein and Peptide Science, 2012, 13, 560-569

1389-2037/12 $58.00+.00 © 2012 Bentham Science Publishers

Angiotensin II: Role in Skeletal Muscle Atrophy

Claudio Cabello-Verrugio1,*, Gonzalo Córdova2 and José Diego Salas1

1Laboratorio de Biología y Fisiopatología Molecular, Departamento de Ciencias Biológicas, Facultad de Ciencias

Biológicas & Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile; 2Centro de Regulación Celular y Pa-

tología (CRCP), Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular,

Pontificia Universidad Católica de Chile, Santiago, Chile

Abstract: Skeletal muscle, the main protein reservoir in the body, is a tissue that exhibits high plasticity when exposed to changes. Muscle proteins can be mobilized into free amino acids when skeletal muscle wasting occurs, a process called skeletal muscle atrophy. This wasting is an important systemic or local manifestation under disuse conditions (e.g., bed rest or immobilization), in starvation, in older adults, and in several diseases. The molecular mechanisms involved in muscle wasting imply the activation of specific signaling pathways which ultimately manage muscle responses to modu-late biological events such as increases in protein catabolism, oxidative stress, and cell death by apoptosis. Many factors have been involved in the generation and maintenance of atrophy in skeletal muscle, among them angiotensin II (Ang-II), the main peptide of renin-angiotensin system (RAS). Together with Ang-II, the angiotensin–converting enzyme (ACE) and the Ang-II receptor type 1 (AT-1 receptor) are expressed in skeletal muscle, forming an important local axis that can regulate its function. In many of the conditions that lead to muscle wasting, there is an impairment of RAS in a global or local fashion. At this point, there are several pieces of evidence that suggest the participation of Ang-II, ACE, and AT-1 receptor in the generation of skeletal muscle atrophy. Interestingly, the Ang-II participation in muscle atrophy is strongly ligated to the regulation of hypertrophic activity of factors such as insulin-like growth factor 1 (IGF-1). In this article, we reviewed the current state of Ang-II and RAS function on skeletal muscle wasting and its possible use as a therapeutic tar-get to improve skeletal muscle function under atrophic conditions.

Keywords: angiotensin II, skeletal muscle atrophy, angiotensin–converting enzyme (ACE), angiotensin II receptor type 1 (AT-1), AT-1 receptor blocker (ARB), atrogin-1, MuRF-1.

INTRODUCTION

Skeletal muscle atrophy corresponds to the loss of mus-cle mass produced by muscle wasting concomitantly with a decrease of muscle strength associated with some pathologi-cal conditions. This wasting is an important systemic or local manifestation under acute pathological conditions, such as disuse by immobilization, post-operatory recovering process, microgravity, and starvation, or under chronic conditions, such as aging, a sedentary lifestyle, or several diseases (can-cer, diabetes, AIDS, and cardiac, lung, or renal failure). All of them correspond to an increasing problem in our society [1, 2]. Under these conditions, the catabolism of proteins is exaggerated and the oxidative stress are increased, leading to a decrease in muscle mass [3].

One of the modulators of skeletal muscle function is the renin-angiotensin system (RAS) axis. The main effector of RAS is angiotensin II (Ang-II), a peptide that participates in the control of blood pressure, sodium balance, fluids volume, and some other hemodynamic features. In addition, Ang-II plays a key role in the regulation of skeletal muscle function

*Address correspondence to this author at the Laboratorio de Biología y Fisiopatología Molecular, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas & Facultad de Medicina, Universidad Andres Bello, Santiago, Chile. Av. Republica 239, Postal Code 8370146, Santiago, Chile; E-mail: [email protected]

by modulating the damage and contractile activity in muscle disease, such as Duchenne muscular dystrophy [4, 5]. How-ever, the main deleterious effect of Ang-II on skeletal muscle is that by inducing changes in enzyme activation, cell oxida-tive state, and gene expression, loss of muscle mass results. All of these changes alter the normal muscle remodeling process, leading to skeletal muscle wasting. Many of the conditions that contribute to skeletal muscle atrophy, such as chronic heart failure (CHF) and chronic kidney disease (CKD), are accompanied by an increase in the activation of the RAS axis, hence promoting an increase in the circulating Ang-II, while the loss of muscle mass worsens [6]. In this review, we address some of the main topics in relation with systemic or local muscle-RAS axis, skeletal muscle atrophy, and the cellular and molecular mechanisms underlying this process.

EXPRESSION OF RENIN-ANGIOTENSIN SYSTEM

COMPONENTS IN SKELETAL MUSCLE

The identification of the local RAS axis has been de-scribed in several tissues [7]. In addition to Ang-II, the local RAS axis is composed of the angiotensin–converting enzyme (ACE) and two membrane receptors for Ang-II called Ang-II receptors type 1 (AT-1) and type 2 (AT-2) [8]. This section reviewed the evidence that support the expression of RAS components in skeletal muscle.

Page 2: Angiotensin II: Role in Skeletal Muscle Atrophy

Angiotensin II Produces Skeletal Muscle Wasting Current Protein and Peptide Science, 2012, Vol. 13, No. 6 561

ACE

ACE is the enzyme responsible to produce Ang-II from angiotensin I. The levels and activity of ACE are directly related to the circulating Ang-II levels or to the tissues of these levels.

ACE activity has been reported in skeletal muscle and cell lines from skeletal muscle [9-13]. Thus, there is evi-dence that de novo synthesis of angiotensin I (Ang-I) and angiotensin II occurs in skeletal muscle, suggesting the pres-ence of ACE activity in an indirect way [14]. Other in vivo experiments gave similar results, suggesting the presence of functional ACE [15]. The Ang-II production has been asso-ciated with ACE expressed in the capillaries from skeletal muscle, showing a close relationship between capillary den-sity and the number of ACE transcripts [16]. This evidence suggests that the activation of ACE from capillaries has an impact on skeletal muscle physiology [17-20]. This evi-dence, in addition to a short half life of Ang-II (14.8 +/- 2.5 seconds under physiological conditions), suggests that in skeletal muscle could exist a local paracrine/autocrine signal-ing of Ang-II in which the relation muscle - vessels would be is essential [21]. Examples in other tissues in which Ang II can act through local RAS are heart and kidneys [22]. In addition, the existence of local paracrine/autocrine Ang-II signaling in the rat epididymis has been reported [23]. Fur-thermore, in cardiac muscle there is a loop initiated by the heart contraction and induces the secretion of Ang II which led to an increase in force contraction through an autocrine/paracrine signaling [24]. Thus, a possible mecha-nism of Ang-II activity in skeletal muscle is through a paracrine/autocrine action.

Interestingly, there is evidence that ACE is also ex-pressed in the neuromuscular junction [20]. Moreover, ACE is expressed in skeletal muscle fibers, which is supported by studies in vitro, which suggests that ACE is expressed in skeletal muscle cells [12, 13, 25, 26].

Together, these data indicate that ACE is expressed both in muscle fibers and vessels associated with skeletal muscle.

AT Receptors

The main receptors for Ang-II are AT-1 and AT-2 recep-tors [7]. These receptors mediate the majority of Ang-II-dependent effects in vitro and in vivo [27]. However, there is contrasting evidence about the expression of the AT recep-tors and their direct contribution to the Ang-II-dependent effects on skeletal muscle. Some studies have reported that skeletal muscle does not express the Ang-II receptors [28]. However, there is increasing evidence that indicate the ex-pression of AT receptors in skeletal muscle. Thus, it has been reported that Ang-II acts directly on myotubes to in-crease protein degradation or on skeletal muscle cells to pro-duce an increase in extracellular matrix protein levels and reactive oxygen species, which suggests that Ang-II-dependent effects are mediated by receptors located in the muscle surface [29-31]. A more direct evidence has been demonstrated in primary human skeletal myoblasts, which suggests the presence of AT-1 and AT-2 receptors, with AT-1 predominantly expressed, as observed from an analysis of Ang-II binding in the presence of selective inhibitors of AT-

1 and AT-2 receptors and western blot analysis [32]. In addi-tion, studies from whole skeletal muscle, primary and C2C12 myoblasts, and C2C12-derived myotubes showed differen-tially the expression of RAS members, including AT-1 and AT-2 receptors. The AT-2 receptors are distributed with a heterogeneous staining pattern in comparison with AT-1 receptors, which colocalized with the actin cytoskeleton in many areas and were detected in the nucleus [26]. Evidence from our group and others strongly suggest that AT-1 and AT-2 receptors are both expressed in normal and dystrophic skeletal muscle, in myoblasts, and in C2C12-derived myo-tubes [4, 5, 20].

In addition, there are several results that suggest that AT-1 and AT-2 receptors are expressed also in skeletal muscle microvasculature, where one of its possible roles is to regu-late basal muscle microvascular perfusion [33].

To summarize, all the composing elements of the RAS are expressed in skeletal muscle, with differences in their densities and location.

ANGIOTENSIN II IS AN ATROPHIC FACTOR IN SKELETAL MUSCLE

Ang-II levels are increased in diseases such as CHF and CKD, which are characterized by significant skeletal muscle wasting that negatively impacts mortality and morbidity [34, 35]. Thus, several studies in vivo strongly support the par-ticipation of Ang-II in muscle wasting. Delafontaine et al. have described that Ang-II infusion in rats caused a signifi-cant decrease in the animals’ weight by a reduction in food intake through a pressure-independent mechanism [36]. Fur-ther studies based on pair-feeding experiments in rats dem-onstrated that an anorexigenic effect produces the Ang-II-dependent weight loss, which is directly related to the de-crease of skeletal muscle mass [37]. This study indicated that the main mechanism of Ang-II-induced loss of muscle mass involves an increase in protein breakdown and a decrease in the IGF-1 levels and signaling, which is the main anabolic pathway in skeletal muscle [37]. Recently it was demon-strated that Ang-II infusion produces waste in the diaphragm muscle, which could explain respiratory muscle dysfunction in CHF and CKD patients [38]. In addition, there is evidence that suggests the treatment with AT-1 receptor blockers and ACE inhibitors may improve weight loss in CHF, indirectly supporting the participation of Ang-II in this pathological process [39, 40]. Moreover, several studies in vitro indicate the catabolic role of Ang-II on skeletal muscle. In myotubes from C2C12 cells, Ang-II decreases the total amount of muscle proteins [30, 31, 41]. The mechanism in this process involves the increased protein catabolism, although an in-hibitory component on protein synthesis has also been re-ported [42].

Protein Degradation

Ang-II-dependent loss in skeletal muscle mass is caused by enhancing protein degradation through the activation of the ubiquitin-proteasome pathway (UPP) [31, 38].

Briefly, ubiquitin-conjugated proteins are recognized and subsequently degraded by the 26S proteasome, a multicata-lytic enzyme complex. UPP activity depends upon coordi-

Page 3: Angiotensin II: Role in Skeletal Muscle Atrophy

562 Current Protein and Peptide Science, 2012, Vol. 13, No. 6 Cabello-Verrugio et al.

nated interactions among several enzyme families (Fig. 2). First, ubiquitin monomers are activated by the ubiquitin-activating enzyme known as E1. This is an ATP-dependent step in which ubiquitin is linked to E1. Second, activated ubiquitin is transferred to an ubiquitin carrier protein (E2). Third, the E2 interacts with one of several hundred ubiquitin ligases (E3s) to transfer activated ubiquitin to the substrate. This begins an iterative process by which E2/E3 partners attach multiple ubiquitin monomers to form polyubiquitin chains, marking the protein substrate for degradation. Fi-nally, the marked protein is degraded by the 26S proteasome complex [43-45]. The 26S proteasome complex is formed by a 20S core complex, the catalytic subunit, and one or two 19S regulatory complexes in charge of substrate recognition (see Fig. 1) [46, 47]. In skeletal muscle, there are E3 ubiq-uitin ligases called muscle atrophy F-box (MAFbx), or atrogin-1, and muscle RING finger 1 (MuRF-1). These mus-cle-specific E3-ligases are upregulated in skeletal muscle atrophy, essential to produce muscle waste [48, 49]. More interestingly, levels of both proteins increase after Ang-II treatment in skeletal muscle (see Fig. 1) [50].

It has been described that in skeletal muscle atrophy, the levels and activity of the proteasome components are in-creased [51]. Among them is the increase of mRNA levels of several subunits from the 20S core complex (Psma1, Psma5, Psmb3, and Psmb4) and the 19S regulator complex (Psmc4, Psmd8, and Psmd11) of the 26S proteasome in atrophied muscles from different causes including Ang-II treatment (see Fig. 1) [52].

In summary, Ang-II is involved directly in the process of muscle degradation by stimulating expression and activity of several components of the ubiquitin-proteasome pathway.

Protein Synthesis

The catabolic effects of Ang-II to produce muscle waste have been associated with changes in IGF-1 levels and its signaling pathway. IGF-1 has been described as an inductor of protein synthesis and an inhibitor of protein degradation [53]. Briefly, IGF-1 binds to its transducer receptor (IGF-1R) which in turn phosphorylates insulin receptor substrate 1 (IRS-1) and activates PI3kinase and AKT. Subsequently, AKT phosphorylates FOXO3 and mTOR proteins. Phos-phorylated FOXO3 is kept in the cytoplasm, preventing its nuclear translocation to activate the transcription of atro-genes, such as atrogín-1, whereas phosphorylated mTOR favors the signaling involved in the protein synthesis, with the participation of S6 kinase and translational machinery (see Fig. 2).

Delafontaine’s group showed that the infusion of Ang-II in rats decreased the levels of circulating IGF-1 and its bind-ing proteins, IGFBP-2 and IGFBP-3, which regulate PI3K/AKT signaling [36]. Moreover, they further demon-strated that Ang-II infused to rats has an inhibitory effect on the autocrine IGF-1 system in skeletal muscle. This becomes more important than the decrease in circulating IGF-1 [37]. Since IGF-1 levels were strongly diminished in Ang-II-induced wasting, further studies to strengthen the Ang-II/IGF-1 relation showed that the muscle-specific overex-

Fig. (1). The Ubiquitin Proteasome Pathway (UPP) is involved in the Ang-II-induced skeletal muscle atrophy.

The ubiquitination machinery is composed of E1 ubiquitin activation proteins, E2 ubiquitin conjugation enzymes, and E3 ubiquitin ligation proteins, which catalyze the polyubiquitination (U) of substrate proteins (S). The proteasome 26S complex is formed by the 20S catalytic subunit and at least one 19S regulatory complex. Ang-II specifically stimulates the expression of two E3 ubiquitin ligases, atrogin-1, and MuRF-1. In addition, Ang-II that has been implicated in the expression increased in several components of the 20S and 19S subunits of the proteasome.

Page 4: Angiotensin II: Role in Skeletal Muscle Atrophy

Angiotensin II Produces Skeletal Muscle Wasting Current Protein and Peptide Science, 2012, Vol. 13, No. 6 563

pression of IGF-1 blocked the Ang-II-induced deleterious effects, producing a protective effect on skeletal muscle against Ang-II [54].

Ang-II produces muscle atrophy by inhibition of protein synthesis [42]. This effect involves proteins that are part of the translation machinery, such as the alpha subunit of eu-karyotic initiation factor 2 (eIF2 ), the elongation factor 2 (eEF2), and the initiation factor 4E-binding protein (4E-BP1). Eley et al. showed that the treatment of myotubes with Ang-II increases the eIF2 phosphorylation and concomi-tantly the inhibition of protein synthesis. This effect was sensitive to a dsRNA-dependent protein kinase (dsPKR) inhibitor [55]. Moreover, they later showed that Ang-II in-creased the phosphorylation of dsPKR, eEF2, and 4E-BP1. All these effects lead to a decrease in protein synthesis [56].

In summary, one of the mechanisms that participates in the Ang-II-dependent atrophic effect is the impairment of the IGF-1 signaling, which is directly involved in the protein synthesis.

REGULATORY MECHANISMS AND SIGNALING

PATHWAYS INVOLVED IN THE ANGIOTENSIN II–

DEPENDENT SKELETAL MUSCLE ATROPHY

Ang-II levels are essential in the atrophic effect on skele-tal muscle in CHF and CKD [34, 35]. The main factor that modulates the Ang-II levels is the ACE activity. Studies of the ACE gene demonstrated that a polymorphism exists,

which modulates the ACE activity. This polymorphism con-sists of an insertion (I) or a deletion (D) of a 287-base pairs sequence in the 16th intron of ACE gene which is located in the chromosome 17 [57]. One possible explanation about how the polymorphism affects the ACE expression is the proximity of the 16th intron with Alu regions of the gene. Because of this proximity it is possible that the I or D geno-type affects the regulation of the expression in charge of Alu regions [58]. The different possible genotypes (I/I), (I/D), and (D/D) account for around half of the enzyme levels in plasma. The D/D alleles are associated with higher concen-trations of serum ACE, while the I/D and the I/I alleles are ligated to lower ones [59]. This suggests that persons with D/D allele would present a higher circulating Ang-II amount than persons with the other two alleles. The D allele variant correlates with a major risk of CHF. Furthermore, there are contradictory results among ethnic groups in relation with CKD and the different polymorphisms [60]. The D/D allele was correlated with a higher risk of nephropathies and a faster decrease in renal function particularly in Asian indi-viduals with type II diabetes while the data for other ethnic groups was inconsistent [61].

Thus, the ACE polymorphism can be considered as a genetic factor that could modulate the development of Ang-II-dependent skeletal muscle atrophy.

Studies using AT-1 receptor blockers (ARB) and ACE inhibitors are among the first studies performed to identify the role of Ang-II in skeletal muscle atrophy, which resulted

Fig. (2). IGF-1-dependent anabolic pathway is modulated by Ang-II during muscle wasting.

IGF-1 is a factor that stimulates protein synthesis by activating PI3K/AKT/mTOR signaling. In addition, IGF-1 inhibits protein degradation through the PI3K/AKT/FOXO3 pathway in skeletal muscle. Ang-II decreases the levels and/or availability of IGF-1, therefore impairing PI3K/AKT signaling, which leads to the activation of FOXO3 and the expression of E3 ubiquitin ligase atrogin-1, which contributes to pro-tein degradation.

Page 5: Angiotensin II: Role in Skeletal Muscle Atrophy

564 Current Protein and Peptide Science, 2012, Vol. 13, No. 6 Cabello-Verrugio et al.

in a decrease of skeletal muscle wasting, leading to the con-clusion that both Ang-II and the activation of the AT-1 re-ceptor could produce or exacerbate skeletal muscle atrophy by decreasing the normal protein synthesis-degradation rate [39, 62].

On the molecular level, studies from Tisdale’s group suggests that Ang-II induced the autophosphorylation of dsPKR, leading to the phosphorylation of the eukaryotic initiation factor 2 (eIF2) and inducing the decrease in protein synthesis [41, 55]. This Ang-II-dependent mechanism in-volves the transient increase of intracellular Ca(2+), which participates in the caspases-3- and caspases-8-dependent dsPKR activation, producing an increase in protein degrada-tion and a decrease in protein synthesis [63]. Moreover, the activation of caspases-3 and caspases-8 induced by Ang-II leads to the activation of p38 MAPK and to the increase in ROS formation, which is known to promote protein degrada-tion through the ubiquitin-proteasome pathway [56, 64].

In addition to dsPKR-caspase pathway, Ang-II has been described to induce the activation of nuclear factor kappa B (NF- B), increasing protein degradation by the ubiquitin-proteasome pathway [55]. This activation of NF- B requires the activity of protein kinase C (PKC) and the further inacti-vation of the NF- B inhibitor, I- B, through phosphorylation and degradation induced by the ubiquitin-proteasome path-way [30].

The ability of Ang-II to induce muscle atrophy through inhibition of protein synthesis is attenuated by insulin-like growth factor-1 (IGF-1) [50, 54]. In addition, studies show that IGF-1 prevents the catabolic effects of Ang-II, and this was mediated by AKT and FOXO3 signaling cascades, lead-ing to the inhibition of atrogin-1 and suggesting a critical role for this ubiquitin ligase in the Ang-II-induced catabolic effects [41, 50, 54]. Moreover, Ang-II was able to reduce the IGF-1/AKT/mTOR signaling cascade, preventing the normal maintenance of muscle mass [65]. Evidence shows that Ang-II can induce the expression of the ubiquitin ligases, atrogin-1 and MuRF-1, previous to the reduction in the expression of IGF-1 in the muscle, thus promoting skeletal muscle wasting [50].

The atrophic effects of Ang-II require the reactive oxy-gen species (ROS) that depends on the production of super-oxide by NADPH oxidase [66]. Interestingly, there is an increase in ROS levels and in the expression of the NADPH oxidase subunits in rats treated with an infusion of Ang-II [67]. In addition to its catabolic effect, Ang-II strongly en-hanced NADPH oxidase activity and ROS generation in myotubes from L6 cells. The incubation with ARB losartan and also the NADPH oxidase inhibitor, apocynin, blocked these effects, suggesting the participation of AT-1 receptor and ROS derived from NADPH oxidase in the Ang-II-induced catabolic effects [6, 68]. Moreover, Sukhanov et al. demonstrated an increase of mitochondria-derived superox-ides in skeletal muscle of Ang-II-infused animals, which suggests the involvement of NADPH oxidase and the mito-chondrial system in Ang-II-induced muscle atrophy [6, 66]. The treatment of murine myotubes with antioxidants, which prevented the Ang-II-induced activation of NF- B, corrobo-rated other evidence for the role of ROS in Ang-II-induced skeletal muscle atrophy [69].

The other mechanism involved in the Ang-II-induced skeletal muscle wasting is mediated by the pro-inflammatory cytokine interleukin type 6 (IL-6). This cytokine has been demonstrated to be produced in the liver, together with se-rum amyloid A (SAA), in response to high levels of circulat-ing Ang-II, and then to act synergistically with SAA to im-pair IGF-1 signaling [28]. IL-6 and SAA are able to activate the suppressor of cytokine signaling 3 (SOCS-3), and as a consequence, the (IRS-1) protein levels are strongly de-creased, leading to the perturbation in the insulin/IGF-1 pathway, thus worsening the protein synthesis in skeletal muscle [28]. Interestingly, these effects are totally inhibited in IL-6-deficient mice or potentiated by an SAA-overexpressing adenovirus, suggesting that Ang-II-mediated catabolic effects on skeletal muscle can be mediated by IL-6-dependent signaling pathways [28].

In conclusion, Ang-II induces its catabolic effects through the activation of several signaling pathways, includ-ing dsPKR and NF- B, and also through the participation of ROS and the increases of expression and activity of the ubiquitin-proteasome pathway to promote the degradation of muscle proteins. On the other hand, Ang-II impairs the IGF-1/AKT/mTOR signaling cascade, decreasing the synthesis and normal maintenance of muscle proteins and contributing to skeletal muscle waste (see Fig. 3).

RENIN-ANGIOTENSIN SYSTEM AS A POSSIBLE

THERAPEUTICAL TARGET TO PREVENT SKELE-TAL MUSCLE ATROPHY

As we have characterized in this review, Ang-II is an atrophic factor that participates in the genesis of muscle wasting in several pathological conditions [34, 35]. This fact indicates that the regulation of levels or the signaling of Ang-II can be an attractive therapeutic target to decrease or prevent muscle wasting. Since ACE is the enzyme that con-verts Ang-I into Ang-II, the ACE inhibitors have been used in the modulation of muscle mass. Several clinical studies showed that patients treated with ACE inhibitors as anti-hypertensive therapy have a lower decrease in skeletal mus-cle mass and functionality as compared with the group that used different antihypertensive drugs [70-72]. Moreover, patients with chronic heart failure, which have high Ang-II levels and muscle wasting, showed that long-term therapy with the ACE inhibitors improved their respiratory muscle strength [73]. Interestingly, the treatment of older adults with ACE inhibitors produced an increase in the IGF-1 levels and IGFBP-3 [74, 75]. Since IGF-1 attenuates the Ang-II-induced skeletal muscle waste, a possible mechanism through ACE inhibitors that prevent muscle mass loss in-volves the increases in the IGF-1 levels and signaling (Table 1). Patients with congestive heart failure showed the other possible mechanism of the action of ACE inhibitors on skeletal muscle atrophy. ACE inhibition prevented the fail-ure in the sarcoplasmic reticulum Ca2+ pump and also the early fatigue and exercise intolerance, suggesting that Ang-II should have detrimental effects in skeletal muscle through the alteration of the function of the sarcoplasmic reticulum Ca2+ pump [76]. In addition to direct effects on skeletal mus-cle, patients with heart failure treated with chronic therapy involving ACE inhibitors showed an improvement in endo-thelial function and increases in the proportion of slow

Page 6: Angiotensin II: Role in Skeletal Muscle Atrophy

Angiotensin II Produces Skeletal Muscle Wasting Current Protein and Peptide Science, 2012, Vol. 13, No. 6 565

Fig. (3). Molecular mechanisms involved in Ang-II-induced skeletal muscle atrophy.

Some of the mechanisms involved in Ang-II-mediated skeletal muscle atrophy: 1. Decrease in protein synthesis is due to the inhibition of both the initiation and elongation components of translational machinery. 2. Increase in protein degradation is the effect of the increment in the levels and activation of proteasome components, such as atrogin-1 and MuRF-1.

Table 1. Strategies for Therapeutic Treatment of the Skeletal Muscle Atrophy

Target Agonist / Antagonist Effects

Renin angiotensin system

ACE ACE inhibitors

AT-1 receptor ARB

Decrease the Ang-II levels and Ang-II dependent signaling, de-

crease of apoptosis, endothelial dysfunction, ROS production andprotein degradation, increase the IGF-1 signaling

Cytokines and soluble factors

TNF-a Anti-TNF-a antibodies

Myostatin Anti-myostatin antibodies

IL-6 Anti-IL-6antibodies

Decrease the signaling pathways of these cytokines involved atro-

phy

Oxidate stress Trolox

Apocynin

Vitamin E

Attenuate the oxidative stress-induced calpain and/or caspase-

mediated cleavage of structural sarcomeric proteins

NF-kB activity Resveratrol

Thalidomide

Ibuprofen

EPA

HMB

Decrease the protein degradation by inhibiting the NF-kB activity

Page 7: Angiotensin II: Role in Skeletal Muscle Atrophy

566 Current Protein and Peptide Science, 2012, Vol. 13, No. 6 Cabello-Verrugio et al.

(Table 1) contd….

Target Agonist / Antagonist Effects

Ubiquitin-Proteasome Bortezomib

Adenosyl-phospho-ubiquitinol

Phenylarsenoxides

MG-132

Attenuate the protein degradation by direct inhibition of ubiquitinconjugation or proteasome activity

Glucocorticoides Antagonist of receptor (RU-486) Decrease the protein degradation

Beta2 adrenergic receptor Antagonist of receptor (Clenbuterol) Decrease the protein degradation and glucocorticoid effect on mass

muscle

Deacetylase histone (HDAC) HDAC inhibitors Increase mass muscle modulating myostatin, FOXO and IGF-1

signaling

Different targets for therapeutic intervention of skeletal muscle atrophy are shown together with their antagonist or agonist and the effect observed when they are used (TNF- , tumor necrotic factor type alpha; EPA, eicosapentaenoic acid; HMB, -hydroxi- -metylbutyrate).

twitch fibers, in the oxidative capacity, and in the capillary density, thereby enhancing aerobic capacity and skeletal muscle perfusion [15, 77]. Thus, the beneficial effects of ACE inhibition on skeletal muscle atrophy can be explained by multiple mechanisms.

The other strategy to decrease the Ang-II-dependent atrophic effects is the blockade of AT-1 receptor with ARB. Early studies on rats with congestive heart failure indicate that the blockade of the AT-1 receptor reduced the apoptotic dependent atrophy and prevented changes in the myosin heavy chains (Table 1) [39]. Recently, Burks et al. demon-strated that in immobilized hind limbs of mice, the ARB losartan prevented skeletal muscle mass loss [65]. The mechanism in this antiatrophic effect of losartan involves the increase of IGF-1/AKT/mTOR signaling cascade [65].

In addition to renin angiotensin system inhibition by ACE inhibitors and ARB, there are several target to decrease skeletal muscle atrophy (See Table 1). Briefly, the use of blocking antibodies for myostatin, TNF- or IL-6 strongly decrease the protein degradation during process atrophic in the muscle [78-80]. Interestingly, Ang-II has been involved in the skeletal muscle atrophy induced by TNF- and IL-6, and myostatin has been demonstrated to increase the levels of Ang-II [28, 81-83]. Moreover, the use of antioxidant compounds such as trolox, apocynin and vitamin E indicate that oxidative stress play a key role in the skeletal muscle atrophy [84-87]. Ang-II induce ROS in vitro and in vivo and the use of ACE inhibitors or ARB can decrease the ROS-dependent effects of Ang-II. Other agents that could thera-peutically be used are inhibitors of NF- B signaling, gluco-corticoids, proteasome activity and histone deacetylase en-zymes or agonists for beta 2 adrenergic receptor (See Table 1) [88-100].

In summary, the decrease of Ang-II levels and/or signal-ing by ACE inhibitors or ARB improves the muscle function and muscle mass loss in a disease wherein high Ang-II levels have been described. Thus, the use of these antihypertensive drugs as antiatrophic treatment could be useful in the treat-ments against skeletal muscle atrophy.

CONCLUSIONS

Skeletal muscle atrophy is a muscle-wasting syndrome associated with aging, disuse, immobilization, and several pathological conditions such as AIDS, cancer, diabetes, CHF, and CKD. In this article, we reviewed the participation of one of the atrophic factors: Ang-II. We showed a vast, complex, and interlaced scenario that leads to muscle atro-phy, where Ang-II appears to be an interesting and active participant in the events that will lead to protein degradation, inhibition of protein synthesis, and muscle waste. The mechanism by which Ang-II exerts these effects includes the following: increase in the levels and activity of proteasome components, inhibition of proteins from the translational machinery, and increase of ROS through different signaling pathways. Although further information is necessary to fully elucidate the mechanisms involved in these events and the participation of RAS axis in muscle atrophy, ACE and AT-1 receptor are revealed as promising targets for muscle atrophy therapy development.

CONFLICT OF INTEREST

The author(s) confirm that this article content has no con-flicts of interest.

ACKNOWLEDGEMENT

This publication was supported by research grants from FONDECYT # 1120380, CARE PFB12/2007.

REFERENCES

[1] Booth, F. W.; Gollnick, P. D. Effects of disuse on the structure and function of skeletal muscle. Med. Sci. Sports. Exerc., 1983, 15 (5), 415-20.

[2] Jagoe, R. T.; Goldberg, A. L. What do we really know about the ubiquitin-proteasome pathway in muscle atrophy? Curr. Opin.

Clin. Nutr. Metab. Care, 2001, 4 (3), 183-90. [3] Kandarian, S. C.; Stevenson, E. J., Molecular events in skeletal

muscle during disuse atrophy. Exerc. Sport. Sci. Rev., 2002, 30 (3), 111-6.

[4] Cabello-Verrugio, C.; Morales, M. G.; Cabrera, D.; Vio, C. P.; Brandan, E., Angiotensin II receptor type 1 blockade decreases

Page 8: Angiotensin II: Role in Skeletal Muscle Atrophy

Angiotensin II Produces Skeletal Muscle Wasting Current Protein and Peptide Science, 2012, Vol. 13, No. 6 567

CTGF/CCN2-mediated damage and fibrosis in normal and dys-trophic skeletal muscles. J. Cell Mol. Med., 2012, 16 (4), 752-64.

[5] Cabello-Verrugio, C.; Acuna, M. J.; Morales, M. G.; Becerra, A.; Simon, F.; Brandan, E., Fibrotic response induced by angiotensin-II requires NAD(P)H oxidase-induced reactive oxygen species (ROS) in skeletal muscle cells. Biochem. Biophys. Res. Commun., 2011, 410 (3), 665-70.

[6] Sukhanov, S.; Semprun-Prieto, L.; Yoshida, T.; Michael Tabony, A.; Higashi, Y.; Galvez, S.; Delafontaine, P., Angiotensin II, oxida-tive stress and skeletal muscle wasting. Am. J. Med. Sci., 2011, 342 (2), 143-7.

[7] de Gasparo, M.; Catt, K. J.; Inagami, T.; Wright, J. W.; Unger, T., International union of pharmacology. XXIII. The angiotensin II re-ceptors. Pharmacol. Rev., 2000, 52 (3), 415-72.

[8] Matsubara, H.; Inada, M., Molecular insights into angiotensin II type 1 and type 2 receptors: expression, signaling and physiological function and clinical application of its antagonists. Endocr. J.,

1998, 45 (2), 137-50. [9] Reneland, R.; Lithell, H., Angiotensin-converting enzyme in hu-

man skeletal muscle. A simple in vitro assay of activity in needle biopsy specimens. Scand. J. Clin. Lab. Invest., 1994, 54 (2), 105-11.

[10] Ward, P. E.; Russell, J. S.; Vaghy, P. L., Angiotensin and bradyki-nin metabolism by peptidases identified in skeletal muscle. Pep-

tides, 1995, 16 (6), 1073-8. [11] Reneland, R.; Haenni, A.; Andersson, P. E.; Andren, B.; Lithell, H.,

Skeletal muscle angiotensin-converting enzyme and its relationship to blood pressure in primary hypertension and healthy elderly men. Blood Press, 1999, 8 (1), 16-22.

[12] Dragovic, T.; Minshall, R.; Jackman, H. L.; Wang, L. X.; Erdos, E. G., Kininase II-type enzymes. Their putative role in muscle energy metabolism. Diabetes 1996, 45 Suppl 1, S34-7.

[13] Mori, S.; Tokuyama, K., Variation in ACE activity affects myo-genic differentiation in C2C12 cells. Biochem. Biophys. Res. Com-

mun., 2007, 353 (2), 369-75. [14] Danser, A. H.; Koning, M. M.; Admiraal, P. J.; Sassen, L. M.;

Derkx, F. H.; Verdouw, P. D.; Schalekamp, M. A., Production of angiotensins I and II at tissue sites in intact pigs. Am. J. Physiol.,

1992, 263 (2 Pt 2), H429-37. [15] Jones, A.; Woods, D. R., Skeletal muscle RAS and exercise per-

formance. Int. J. Biochem. Cell Biol., 2003, 35 (6), 855-66. [16] Ohishi, M.; Ueda, M.; Rakugi, H.; Okamura, A.; Naruko, T.;

Becker, A. E.; Hiwada, K.; Kamitani, A.; Kamide, K.; Higaki, J.; Ogihara, T., Upregulation of angiotensin-converting enzyme during the healing process after injury at the site of percutaneous translu-minal coronary angioplasty in humans. Circulation, 1997, 96 (10), 3328-37.

[17] Schaufelberger, M.; Drexler, H.; Schieffer, E.; Swedberg, K., An-giotensin-converting enzyme gene expression in skeletal muscle in patients with chronic heart failure. J. Card. Fail, 1998, 4 (3), 185-91.

[18] Linderman, J. R.; Greene, A. S., Distribution of angiotensin II receptor expression in the microcirculation of striated muscle. Mi-

crocirculation, 2001, 8 (4), 275-81. [19] Weber, D. S.; Lombard, J. H., Angiotensin II AT1 receptors pre-

serve vasodilator reactivity in skeletal muscle resistance arteries. Am. J. Physiol. Heart Circ. Physiol., 2001, 280 (5), H2196-202.

[20] Sun, G.; Haginoya, K.; Dai, H.; Chiba, Y.; Uematsu, M.; Hino-Fukuyo, N.; Onuma, A.; Iinuma, K.; Tsuchiya, S., Intramuscular renin-angiotensin system is activated in human muscular dystro-phy. J. Neurol. Sci., 2009, 280 (1-2), 40-8.

[21] Chapman, B. J.; Brooks, D. P.; Munday, K. A., Half-life of angio-tensin II in the conscious and barbiturate-anaesthetized rat. Br. J.

Anaesth., 1980, 52 (4), 389-93. [22] van Kats, J. P.; de Lannoy, L. M.; Jan Danser, A. H.; van Meegen,

J. R.; Verdouw, P. D.; Schalekamp, M. A., Angiotensin II type 1 (AT1) receptor-mediated accumulation of angiotensin II in tissues and its intracellular half-life in vivo. Hypertension, 1997, 30 (1 Pt 1), 42-9.

[23] Chan, H. C.; Wong, P. Y., Paracrine/autocrine regulation of anion secretion in the epididymis: role of angiotensin II. Biol. Signals,

1996, 5 (6), 309-16.

[24] Cingolani, H. E.; Perez, N. G.; Aiello, E. A.; de Hurtado, M. C., Intracellular signaling following myocardial stretch: an autocrine/paracrine loop. Regul. Pept., 2005, 128 (3), 211-20.

[25] Fernandes, T.; Hashimoto, N. Y.; Oliveira, E. M., Characterization of angiotensin-converting enzymes 1 and 2 in the soleus and plan-taris muscles of rats. Braz. J. Med. Biol. Res., 2010, 43 (9), 837-42.

[26] Johnston, A. P.; Baker, J.; De Lisio, M.; Parise, G., Skeletal muscle myoblasts possess a stretch-responsive local angiotensin signalling system. J. Renin. Angiotensin. Aldosterone Syst., 2011, 12 (2), 75-84.

[27] Stegbauer, J.; Coffman, T. M., New insights into angiotensin recep-tor actions: from blood pressure to aging. Curr. Opin. Nephrol. Hy-

pertens., 2011, 20 (1), 84-8. [28] Zhang, L.; Du, J.; Hu, Z.; Han, G.; Delafontaine, P.; Garcia, G.;

Mitch, W. E., IL-6 and serum amyloid A synergy mediates angio-tensin II-induced muscle wasting. J. Am. Soc. Nephrol., 2009, 20 (3), 604-12.

[29] Morales, M., Vazquez, Y., Acuña, MJ., Rivera, JC., Simon, F., Salas, JD., Álvarez-Ruf, J., Brandan, E., Cabello-Verrugio, C., Angiotensin II- induced pro-fibrotic effects require p38MAPK ac-tivity and Transforming Growth Factor beta 1 expression in skele-tal muscle cells. Int. J. Biochem. Cell Biol., 2012, 44, 1993-2002.

[30] Russell, S. T.; Wyke, S. M.; Tisdale, M. J., Mechanism of induc-tion of muscle protein degradation by angiotensin II. Cell Signal,

2006, 18 (7), 1087-96. [31] Sanders, P. M.; Russell, S. T.; Tisdale, M. J., Angiotensin II di-

rectly induces muscle protein catabolism through the ubiquitin-proteasome proteolytic pathway and may play a role in cancer cachexia. Br. J. Cancer, 2005, 93 (4), 425-34.

[32] Qi, J. S.; Minor, L. K.; Smith, C.; Hu, B.; Yang, J.; Andrade-Gordon, P.; Damiano, B., Characterization of functional urotensin II receptors in human skeletal muscle myoblasts: comparison with angiotensin II receptors. Peptides, 2005, 26 (4), 683-90.

[33] Chai, W.; Wang, W.; Liu, J.; Barrett, E. J.; Carey, R. M.; Cao, W.; Liu, Z., Angiotensin II type 1 and type 2 receptors regulate basal skeletal muscle microvascular volume and glucose use. Hyperten-

sion, 2010, 55 (2), 523-30. [34] Masson, S.; Latini, R.; Bevilacqua, M.; Vago, T.; Sessa, F.; Torri,

M.; Anesini, A.; Salio, M.; Pasotti, E.; Agnello, D.; Santoro, L.; Catania, A.; Ghezzi, P.; Moccetti, T.; Maggioni, A. P., Within-patient variability of hormone and cytokine concentrations in heart failure. Pharmacol. Res., 1998, 37 (3), 213-7.

[35] Roig, E.; Perez-Villa, F.; Morales, M.; Jimenez, W.; Orus, J.; Heras, M.; Sanz, G., Clinical implications of increased plasma an-giotensin II despite ACE inhibitor therapy in patients with conges-tive heart failure. Eur. Heart J., 2000, 21 (1), 53-7.

[36] Brink, M.; Wellen, J.; Delafontaine, P., Angiotensin II causes weight loss and decreases circulating insulin-like growth factor I in rats through a pressor-independent mechanism. J. Clin. Invest.,

1996, 97 (11), 2509-16. [37] Brink, M.; Price, S. R.; Chrast, J.; Bailey, J. L.; Anwar, A.; Mitch,

W. E.; Delafontaine, P., Angiotensin II induces skeletal muscle wasting through enhanced protein degradation and down-regulates autocrine insulin-like growth factor I. Endocrinology, 2001, 142 (4), 1489-96.

[38] Rezk, B. M.; Yoshida, T.; Semprun-Prieto, L.; Higashi, Y.; Suk-hanov, S.; Delafontaine, P., Angiotensin II infusion induces marked diaphragmatic skeletal muscle atrophy. PLoS One, 2012, 7 (1), e30276.

[39] Dalla Libera, L.; Ravara, B.; Angelini, A.; Rossini, K.; Sandri, M.; Thiene, G.; Battista Ambrosio, G.; Vescovo, G., Beneficial effects on skeletal muscle of the angiotensin II type 1 receptor blocker ir-besartan in experimental heart failure. Circulation, 2001, 103 (17), 2195-200.

[40] Anker, S. D.; Negassa, A.; Coats, A. J.; Afzal, R.; Poole-Wilson, P. A.; Cohn, J. N.; Yusuf, S., Prognostic importance of weight loss in chronic heart failure and the effect of treatment with angiotensin-converting-enzyme inhibitors: an observational study. Lancet 2003, 361 (9363), 1077-83.

[41] Russell, S. T.; Eley, H.; Tisdale, M. J., Mechanism of attenuation of angiotensin-II-induced protein degradation by insulin-like growth factor-I (IGF-I). Cell Signal, 2007, 19 (7), 1583-95.

Page 9: Angiotensin II: Role in Skeletal Muscle Atrophy

568 Current Protein and Peptide Science, 2012, Vol. 13, No. 6 Cabello-Verrugio et al.

[42] Russell, S. T.; Sanders, P. M.; Tisdale, M. J., Angiotensin II di-rectly inhibits protein synthesis in murine myotubes. Cancer Lett.,

2006, 231 (2), 290-4. [43] Welchman, R. L.; Gordon, C.; Mayer, R. J., Ubiquitin and ubiq-

uitin-like proteins as multifunctional signals. Nat. Rev. Mol. Cell

Biol., 2005, 6 (8), 599-609. [44] Hershko, A.; Heller, H.; Elias, S.; Ciechanover, A., Components of

ubiquitin-protein ligase system. Resolution, affinity purification, and role in protein breakdown. J. Biol. Chem., 1983, 258 (13), 8206-14.

[45] Reid, M. B., Response of the ubiquitin-proteasome pathway to changes in muscle activity. Am. J. Physiol. Regul. Integr. Comp.

Physiol., 2005, 288 (6), R1423-31. [46] Sorokin, A. V.; Kim, E. R.; Ovchinnikov, L. P., Proteasome system

of protein degradation and processing. Biochemistry (Mosc), 2009, 74 (13), 1411-42.

[47] Tanaka, K., The proteasome: overview of structure and functions. Proc. Jpn. Acad. Ser. B. Phys. Biol. Sci., 2009, 85 (1), 12-36.

[48] Gomes, M. D.; Lecker, S. H.; Jagoe, R. T.; Navon, A.; Goldberg, A. L., Atrogin-1, a muscle-specific F-box protein highly expressed during muscle atrophy. Proc. Natl. Acad. Sci. USA., 2001, 98 (25), 14440-5.

[49] Bodine, S. C.; Latres, E.; Baumhueter, S.; Lai, V. K.; Nunez, L.; Clarke, B. A.; Poueymirou, W. T.; Panaro, F. J.; Na, E.; Dharmara-jan, K.; Pan, Z. Q.; Valenzuela, D. M.; DeChiara, T. M.; Stitt, T. N.; Yancopoulos, G. D.; Glass, D. J., Identification of ubiquitin li-gases required for skeletal muscle atrophy. Science, 2001, 294 (5547), 1704-8.

[50] Yoshida, T.; Semprun-Prieto, L.; Sukhanov, S.; Delafontaine, P., IGF-1 prevents ANG II-induced skeletal muscle atrophy via Akt- and Foxo-dependent inhibition of the ubiquitin ligase atrogin-1 ex-pression. Am. J. Physiol. Heart Circ. Physiol., 2010, 298 (5), H1565-70.

[51] Price, S. R., Increased transcription of ubiquitin-proteasome system components: molecular responses associated with muscle atrophy. Int. J. Biochem. Cell Biol., 2003, 35 (5), 617-28.

[52] Lecker, S. H.; Jagoe, R. T.; Gilbert, A.; Gomes, M.; Baracos, V.; Bailey, J.; Price, S. R.; Mitch, W. E.; Goldberg, A. L., Multiple types of skeletal muscle atrophy involve a common program of changes in gene expression. FASEB J., 2004, 18 (1), 39-51.

[53] Gluckman, P. D.; Douglas, R. G.; Ambler, G. R.; Breier, B. H.; Hodgkinson, S. C.; Koea, J. B.; Shaw, J. H., The endocrine role of insulin-like growth factor I. Acta. Paediatr. Scand. Suppl., 1991, 372, 97-105; discussion 106.

[54] Song, Y. H.; Li, Y.; Du, J.; Mitch, W. E.; Rosenthal, N.; Delafon-taine, P., Muscle-specific expression of IGF-1 blocks angiotensin II-induced skeletal muscle wasting. J. Clin. Invest., 2005, 115 (2), 451-8.

[55] Eley, H. L.; Tisdale, M. J., Skeletal muscle atrophy, a link between depression of protein synthesis and increase in degradation. J. Biol.

Chem., 2007, 282 (10), 7087-97. [56] Eley, H. L.; Russell, S. T.; Tisdale, M. J., Attenuation of depression

of muscle protein synthesis induced by lipopolysaccharide, tumor necrosis factor, and angiotensin II by beta-hydroxy-beta-methylbutyrate. Am. J. Physiol. Endocrinol. Metab., 2008, 295 (6), E1409-16.

[57] Rieder, M. J.; Taylor, S. L.; Clark, A. G.; Nickerson, D. A., Se-quence variation in the human angiotensin converting enzyme. Nat.

Genet., 1999, 22 (1), 59-62. [58] Castellon, R.; Hamdi, H. K., Demystifying the ACE polymor-

phism: from genetics to biology. Curr. Pharm. Des., 2007, 13 (12), 1191-8.

[59] Rigat, B.; Hubert, C.; Alhenc-Gelas, F.; Cambien, F.; Corvol, P.; Soubrier, F., An insertion/deletion polymorphism in the angiotensin I-converting enzyme gene accounting for half the variance of se-rum enzyme levels. J. Clin. Invest., 1990, 86 (4), 1343-6.

[60] Gard, P. R., Implications of the angiotensin converting enzyme gene insertion/deletion polymorphism in health and disease: a snapshot review. Int. J. Mol. Epidemiol. Genet., 2010, 1 (2), 145-57.

[61] Rudnicki, M.; Mayer, G., Significance of genetic polymorphisms of the renin-angiotensin-aldosterone system in cardiovascular and renal disease. Pharmacogenomics, 2009, 10 (3), 463-76.

[62] Vescovo, G.; Ambrosio, G. B.; Dalla Libera, L., Apoptosis and changes in contractile protein pattern in the skeletal muscle in heart failure. Acta. Physiol. Scand., 2001, 171 (3), 305-10.

[63] Eley, H. L.; Russell, S. T.; Tisdale, M. J., Mechanism of activation of dsRNA-dependent protein kinase (PKR) in muscle atrophy. Cell

Signal, 2010, 22 (5), 783-90. [64] Eley, H. L.; Russell, S. T.; Tisdale, M. J., Mechanism of attenua-

tion of muscle protein degradation induced by tumor necrosis fac-tor-alpha and angiotensin II by beta-hydroxy-beta-methylbutyrate. Am. J. Physiol. Endocrinol. Metab., 2008, 295 (6), E1417-26.

[65] Burks, T. N.; Andres-Mateos, E.; Marx, R.; Mejias, R.; Van Erp, C.; Simmers, J. L.; Walston, J. D.; Ward, C. W.; Cohn, R. D., Losartan restores skeletal muscle remodeling and protects against disuse atrophy in sarcopenia. Sci. Transl. Med., 2011, 3 (82), 82ra37.

[66] Semprun-Prieto, L. C.; Sukhanov, S.; Yoshida, T.; Rezk, B. M.; Gonzalez-Villalobos, R. A.; Vaughn, C.; Michael Tabony, A.; De-lafontaine, P., Angiotensin II induced catabolic effect and muscle atrophy are redox dependent. Biochem. Biophys. Res. Commun.,

2011, 409 (2), 217-21. [67] Zhao, W.; Swanson, S. A.; Ye, J.; Li, X.; Shelton, J. M.; Zhang,

W.; Thomas, G. D., Reactive oxygen species impair sympathetic vasoregulation in skeletal muscle in angiotensin II-dependent hy-pertension. Hypertension, 2006, 48 (4), 637-43.

[68] Wei, Y.; Sowers, J. R.; Nistala, R.; Gong, H.; Uptergrove, G. M.; Clark, S. E.; Morris, E. M.; Szary, N.; Manrique, C.; Stump, C. S., Angiotensin II-induced NADPH oxidase activation impairs insulin signaling in skeletal muscle cells. J. Biol. Chem., 2006, 281 (46), 35137-46.

[69] Russell, S. T.; Eley, H.; Tisdale, M. J., Role of reactive oxygen species in protein degradation in murine myotubes induced by pro-teolysis-inducing factor and angiotensin II. Cell Signal, 2007, 19 (8), 1797-806.

[70] Cicoira, M., Inhibitors of angiotensin-converting enzyme and physical function in older women. Lancet, 2002, 360 (9339), 1099; author reply 1099-100.

[71] Di Bari, M.; van de Poll-Franse, L. V.; Onder, G.; Kritchevsky, S. B.; Newman, A.; Harris, T. B.; Williamson, J. D.; Marchionni, N.; Pahor, M., Antihypertensive medications and differences in muscle mass in older persons: the Health, Aging and Body Composition Study. J. Am. Geriatr. Soc., 2004, 52 (6), 961-6.

[72] Carter, C. S.; Cesari, M.; Ambrosius, W. T.; Hu, N.; Diz, D.; Oden, S.; Sonntag, W. E.; Pahor, M., Angiotensin-converting enzyme in-hibition, body composition, and physical performance in aged rats. J. Gerontol. A. Biol. Sci. Med. Sci., 2004, 59 (5), 416-23.

[73] Coirault, C.; Hagege, A.; Chemla, D.; Fratacci, M. D.; Guerot, C.; Lecarpentier, Y., Angiotensin-converting enzyme inhibitor therapy improves respiratory muscle strength in patients with heart failure. Chest, 2001, 119 (6), 1755-60.

[74] Giovannini, S.; Cesari, M.; Marzetti, E.; Leeuwenburgh, C.; Mag-gio, M.; Pahor, M., Effects of ACE-inhibition on IGF-1 and IGFBP-3 concentrations in older adults with high cardiovascular risk profile. J. Nutr. Health Aging, 2010, 14 (6), 457-60.

[75] Onder, G.; Liperoti, R.; Russo, A.; Capoluongo, E.; Minucci, A.; Lulli, P.; Cesari, M.; Maggio, M.; Bernabei, R.; Landi, F., Use of ACE inhibitors is associated with elevated levels of IGFBP-3 among hypertensive older adults: results from the IlSIRENTE study. Eur. J. Clin. Pharmacol., 2007, 63 (4), 389-95.

[76] Shah, K. R.; Ganguly, P. K.; Netticadan, T.; Arneja, A. S.; Dhalla, N. S., Changes in skeletal muscle SR Ca2+ pump in congestive heart failure due to myocardial infarction are prevented by angio-tensin II blockade. Can. J. Physiol. Pharmacol., 2004, 82 (7), 438-47.

[77] Zoll, J.; Monassier, L.; Garnier, A.; N'Guessan, B.; Mettauer, B.; Veksler, V.; Piquard, F.; Ventura-Clapier, R.; Geny, B., ACE inhi-bition prevents myocardial infarction-induced skeletal muscle mi-tochondrial dysfunction. J. Appl. Physiol., 2006, 101 (2), 385-91.

[78] Murphy, K. T.; Chee, A.; Gleeson, B. G.; Naim, T.; Swiderski, K.; Koopman, R.; Lynch, G. S., Antibody-directed myostatin inhibition enhances muscle mass and function in tumor-bearing mice. Am. J.

Physiol. Regul. Integr. Comp. Physiol., 2011, 301 (3), R716-26. [79] Llovera, M.; Carbo, N.; Garcia-Martinez, C.; Costelli, P.; Tessitore,

L.; Baccino, F. M.; Agell, N.; Bagby, G. J.; Lopez-Soriano, F. J.; Argiles, J. M., Anti-TNF treatment reverts increased muscle ubiq-

Page 10: Angiotensin II: Role in Skeletal Muscle Atrophy

Angiotensin II Produces Skeletal Muscle Wasting Current Protein and Peptide Science, 2012, Vol. 13, No. 6 569

uitin gene expression in tumour-bearing rats. Biochem. Biophys.

Res. Commun., 1996, 221 (3), 653-5. [80] Fujita, J.; Tsujinaka, T.; Yano, M.; Ebisui, C.; Saito, H.; Katsume,

A.; Akamatsu, K.; Ohsugi, Y.; Shiozaki, H.; Monden, M., Anti-interleukin-6 receptor antibody prevents muscle atrophy in colon-26 adenocarcinoma-bearing mice with modulation of lysosomal and ATP-ubiquitin-dependent proteolytic pathways. Int. J. Cancer,

1996, 68 (5), 637-43. [81] De Larichaudy, J.; Zufferli, A.; Serra, F.; Isidori, A. M.; Naro, F.;

Dessalle, K.; Desgeorges, M.; Piraud, M.; Cheillan, D.; Vidal, H.; Lefai, E.; Nemoz, G., TNF-alpha- and tumor-induced skeletal mus-cle atrophy involves sphingolipid metabolism. Skelet. Muscle,

2012, 2 (1), 2. [82] Sishi, B. J.; Engelbrecht, A. M., Tumor necrosis factor alpha (TNF-

alpha) inactivates the PI3-kinase/PKB pathway and induces atro-phy and apoptosis in L6 myotubes. Cytokine, 2011, 54 (2), 173-84.

[83] Wang, B. W.; Chang, H.; Kuan, P.; Shyu, K. G., Angiotensin II activates myostatin expression in cultured rat neonatal cardiomyo-cytes via p38 MAP kinase and myocyte enhance factor 2 pathway. J. Endocrinol., 2008, 197 (1), 85-93.

[84] McClung, J. M.; Kavazis, A. N.; Whidden, M. A.; DeRuisseau, K. C.; Falk, D. J.; Criswell, D. S.; Powers, S. K., Antioxidant admini-stration attenuates mechanical ventilation-induced rat diaphragm muscle atrophy independent of protein kinase B (PKB Akt) signal-ling. J. Physiol., 2007, 585 (Pt 1), 203-15.

[85] McClung, J. M.; Van Gammeren, D.; Whidden, M. A.; Falk, D. J.; Kavazis, A. N.; Hudson, M. B.; Gayan-Ramirez, G.; Decramer, M.; DeRuisseau, K. C.; Powers, S. K., Apocynin attenuates diaphragm oxidative stress and protease activation during prolonged mechani-cal ventilation. Crit. Care Med., 2009, 37 (4), 1373-9.

[86] Appell, H. J.; Duarte, J. A.; Soares, J. M., Supplementation of vitamin E may attenuate skeletal muscle immobilization atrophy. Int. J. Sports Med., 1997, 18 (3), 157-60.

[87] Demiryurek, S.; Babul, A., Effects of vitamin E and electrical stimulation on the denervated rat gastrocnemius muscle malondial-dehyde and glutathione levels. Int. J. Neurosci., 2004, 114 (1), 45-54.

[88] Shadfar, S.; Couch, M. E.; McKinney, K. A.; Weinstein, L. J.; Yin, X.; Rodriguez, J. E.; Guttridge, D. C.; Willis, M., Oral resveratrol therapy inhibits cancer-induced skeletal muscle and cardiac atrophy in vivo. Nutr. Cancer, 63 (5), 749-62.

[89] Smuder, A. J.; Hudson, M. B.; Nelson, W. B.; Kavazis, A. N.; Powers, S. K., Nuclear factor-kappaB signaling contributes to me-chanical ventilation-induced diaphragm weakness*. Crit. Care

Med., 40 (3), 927-34.

[90] Khan, Z. H.; Simpson, E. J.; Cole, A. T.; Holt, M.; MacDonald, I.; Pye, D.; Austin, A.; Freeman, J. G., Oesophageal cancer and cachexia: the effect of short-term treatment with thalidomide on weight loss and lean body mass. Aliment. Pharmacol. Ther., 2003, 17 (5), 677-82.

[91] McMillan, D. C.; Wigmore, S. J.; Fearon, K. C.; O'Gorman, P.; Wright, C. E.; McArdle, C. S., A prospective randomized study of megestrol acetate and ibuprofen in gastrointestinal cancer patients with weight loss. Br. J. Cancer, 1999, 79 (3-4), 495-500.

[92] Barber, M. D.; Ross, J. A.; Voss, A. C.; Tisdale, M. J.; Fearon, K. C., The effect of an oral nutritional supplement enriched with fish oil on weight-loss in patients with pancreatic cancer. Br. J. Cancer,

1999, 81 (1), 80-6. [93] Smith, H. J.; Wyke, S. M.; Tisdale, M. J., Mechanism of the at-

tenuation of proteolysis-inducing factor stimulated protein degrada-tion in muscle by beta-hydroxy-beta-methylbutyrate. Cancer Res.,

2004, 64 (23), 8731-5. [94] Beehler, B. C.; Sleph, P. G.; Benmassaoud, L.; Grover, G. J., Re-

duction of skeletal muscle atrophy by a proteasome inhibitor in a rat model of denervation. Exp. Biol. Med. (Maywood), 2006, 231 (3), 335-41.

[95] Wilkinson, K. D.; Smith, S. E.; O'Connor, L.; Sternberg, E.; Tag-gart, J. J.; Berges, D. A.; Butt, T., A specific inhibitor of the ubiq-uitin activating enzyme: synthesis and characterization of adenosyl-phospho-ubiquitinol, a nonhydrolyzable ubiquitin adenylate ana-logue. Biochemistry, 1990, 29 (32), 7373-80.

[96] Berleth, E. S.; Kasperek, E. M.; Grill, S. P.; Braunscheidel, J. A.; Graziani, L. A.; Pickart, C. M., Inhibition of ubiquitin-protein li-gase (E3) by mono- and bifunctional phenylarsenoxides. Evidence for essential vicinal thiols and a proximal nucleophile. J. Biol.

Chem., 1992, 267 (23), 16403-11. [97] Aboudrar, S.; Sempore, B.; Koubi, H.; Dechaud, H.; Desplanches,

D., Effects of adrenalectomy or RU-486 on rat muscle fibers during hindlimb suspension. J. Appl. Physiol., 1993, 75 (6), 2767-73.

[98] Tischler, M. E., Effect of the antiglucocorticoid RU38486 on pro-tein metabolism in unweighted soleus muscle. Metabolism, 1994, 43 (11), 1451-5.

[99] Sneddon, A. A.; Delday, M. I.; Maltin, C. A., Amelioration of denervation-induced atrophy by clenbuterol is associated with in-creased PKC-alpha activity. Am. J. Physiol. Endocrinol. Metab.,

2000, 279 (1), E188-95. [100] Guasconi, V.; Puri, P. L., Epigenetic drugs in the treatment of

skeletal muscle atrophy. Curr. Opin. Clin. Nutr. Metab. Care,

2008, 11 (3), 233-41.

Received: April 29, 2012 Revised: August 17, 2012 Accepted: September 07, 2012