Kidney Res Clin Pract > Epub ahead of print
Price and Wang: Protein-energy wasting in chronic kidney disease: mechanisms responsible for loss of muscle mass and function

Abstract

The worldwide prevalence of chronic kidney disease (CKD) is high and growing, making CKD a leading cause of mortality. Skeletal muscle wasting, sometimes called sarcopenia or protein-energy wasting, is a frequent, serious consequence of CKD that reduces muscle strength and function, diminishes the quality of life of patients, and raises their risk of comorbidities and death. Muscle atrophy results from a disturbance in muscle protein balance that results from some combination of an increased rate of protein degradation, a decreased rate of protein synthesis, and dysfunctional muscle regeneration. Development of therapeutic strategies to ameliorate muscle loss, or maintain muscle mass, is challenging because of the multifactorial nature of the signals that alter protein homeostasis. This review discusses the cellular signals and mechanisms that negatively alter protein turnover in skeletal muscle during CKD.

Introduction

The collective mass of skeletal muscle constitutes 40% to 45% of a normal healthy adult’s body weight [1]. In addition to movement and postural support functions, skeletal muscle serves as a major reservoir of proteins which can be degraded to produce amino acids for energy production and for remodeling responses to other physiological challenges. A broad spectrum of physiologic signals regulates the protein turnover process which consists of protein synthesis, protein degradation, and branched-chain amino acid degradation. According to estimates of protein turnover in healthy adults, approximately ~280 g of protein is synthesized and degraded each day, with the bulk of this activity occurring in skeletal muscle [2]. In chronic diseases such as chronic kidney disease (CKD), small catabolic shifts in protein turnover and dysfunction in muscle’s ability to repair itself lead to protein wasting that impacts the quality of life and increases risks of mortality and morbidity of affected patients.
Reduced muscle mass and function are frequent consequences of CKD and other chronic conditions such as cancer, diabetes mellitus, heart failure, and aging. In 2013, the International Society of Renal Nutrition and Metabolism proposed a unique term, protein-energy wasting or PEW, to denote the chronic loss of protein and energy sources that are hallmarks of CKD [3]. The goal of defining PEW was to accurately identify a physiological context of the disease to enable clinicians and investigators to better understand the disorders and mechanisms for muscle protein and energy dysfunction in CKD. Over the years, other terms such as muscle atrophy, cachexia, and sarcopenia have also been used in the context of CKD, however, the diagnostic criteria applied to these terms are sometimes different [4].
The prevalence of PEW in kidney disease is high. A recent meta-analysis reported that 11% to 54% of stage 3–5 CKD patients in five studies (1,776 subjects) had signs of PEW and the middle two quartiles range of PEW was 28% to 54% in 90 studies (16,434 subjects) of maintenance dialysis patients [5]. Others have reported that loss of lean body mass or PEW is strongly associated with comorbidities, increased hospitalizations, and higher risks of mortality [6,7]. Consequently, there is much interest in understanding the pathophysiologic mechanisms that cause PEW so that improvements can be made in treatment strategies to preserve muscle mass and strength.
In this review, we present evidence for how some major signals and mechanisms contribute to muscle atrophy in CKD. Many of these mechanisms are active in other catabolic disease states as well. Several areas of discovery that will hopefully lead to increased success in maintaining lean body mass in CKD and other patients with kidney disease are also discussed.

Skeletal muscle: a dynamic organ

Skeletal muscles are important for mobility and daily activities. The skeletal muscle contraction process injures myofibers and damages contractile and other proteins. Intracellular and structural proteins are also in a constant state of flux as normal metabolic functions damage or modify them, requiring they be degraded. Rates of protein synthesis and degradation fluctuate throughout the day due to changes in physiological conditions. A typical healthy individual turns over approximately 280 g of protein a day and approximately 75% of this turnover protein occurs in skeletal muscle [2]. Despite the sizeable quantity of daily protein turnover, there is little fluctuation in the mass of skeletal muscle. This lack of change is because skeletal muscle protein synthesis and degradation are balanced in healthy individuals (Fig. 1). In conditions that are associated with muscle atrophy, protein synthesis and degradation in skeletal muscle become imbalanced, leading to a reduction in overall protein synthesis, an acceleration of overall protein degradation, or a combination of the two responses. Studies of experimental animals or patients with CKD have consistently shown that protein degradation in skeletal muscle is increased [811]. In studies of animals with CKD, protein synthesis is typically decreased, compared to animals without CKD. The outcomes of studies with patients with CKD or on dialysis have been inconsistent with some results indicating a decrease in protein synthesis, whereas other studies found no change or an increase in protein synthesis [1215]. The reasons for the inconsistencies in determined protein synthesis rates in humans with CKD are unclear.
Protein synthesis and degradation are nonoverlapping cellular processes that are separately regulated by various internal and external cell signals. Changes in anabolic or catabolic signals cause shifts in protein turnover. For example, after a meal, the increase in serum insulin and amino acids leads to an increase in protein synthesis and a reduction in protein degradation in skeletal muscle. In pathophysiologic conditions like CKD, catabolic signals are ever present and consequently, there is a chronic acceleration in protein degradation while protein synthesis is frequently suppressed. In patients with CKD, these shifts in skeletal muscle protein turnover lead to frequently observed muscle loss and weakness [16].

What changes in muscle physiology and metabolism lead to atrophy in chronic kidney disease?

Protein synthesis

Proteins are synthesized from messenger RNA (mRNA) which carries the nucleic acid code for the protein’s amino acid sequence, a process called protein translation. Each protein is encoded by a unique mRNA that is transcribed from the cell’s genomic DNA. Translation of a protein begins with the formation of the ribosomal complex on the mRNA. This ribosomal complex is composed of ribosomal RNAs (rRNAs), ribosome proteins, initiation factors, and transfer RNAs and it binds to a methionine codon at the mRNA’s initiation site. Additional amino acids are added to the initiator methionine to produce a growing peptide. The processive translation process continues down the mRNA until the ribosomal complex encounters a termination codon on the mRNA and the fully formed nascent peptide and the ribosomal complex dissociate.
A cell’s capacity to synthesize proteins is determined by multiple factors. Formation of the ribosomal complex is a tightly regulated process. Epigenetic and transcriptional mechanisms modulate the levels of ribosomal proteins and rRNAs. Signals internal and external to the cell also influence the rate of protein translation. The mechanistic target of rapamycin protein, mTORC1, is a key integrator of these signals. mTORC1 is a kinase that phosphorylates two key translational proteins—ribosomal protein S6 kinase-1 and eukaryotic initiation factor 4E binding protein (eIF4E-BP). eIF4E is normally inhibited by eIF4E-BP. Phosphorylation of eIF4E-BP allows eIF4E to join the ribosome-mRNA preinitiation complex. S6 facilitates the initiation and elongation steps of protein translation and phosphorylation enhances its activities.
Leucine and insulin/insulin-like growth factor 1 (IGF-1) are key modulators that positively influence protein synthesis through mTORC1, albeit by different signaling pathways (Fig. 1). Leucine activates mTORC1 via Rheb signaling whereas insulin and IGF-1 activate mTORC1 via Akt-induced phosphorylation. In CKD, the circulating and intracellular levels of leucine in skeletal muscle are reduced and likely contribute to a lower rate of translation [11,17]. Insulin and IGF-1 signaling through Akt in muscle is also impaired [18]. In theory, the resulting anabolic resistant state should reduce mTORC1 signaling and protein synthesis. However, measuring the activation state of mTORC1 in skeletal muscles is difficult and few, if any, attempts have been made to do so in patients or animals with CKD.
A recent 2020 paper by Zhang et al. [19] suggests the assumption that anabolic resistance is a major cause of reduced protein synthesis in muscle during CKD may be an oversimplification and that multidimensional causes are responsible. Zhang et al. [19] found that nucleolar protein 66 (NO66) was increased in muscle biopsies from dialysis patients and in muscles of mice with CKD. This was a notable finding because NO66 is a chromatin-modifying demethylase that modulates histone methylation, a modification associated with gene expression. Knockout of NO66 in the skeletal muscles of mice with CKD resulted in a higher rate of protein synthesis than in normal mice with CKD. NO66 was shown to suppress ribosomal biogenesis by a mechanism that requires demethylation activity. Inflammatory cytokines like tumor necrosis factor (TNF) and interleukin 6 (IL-6) were responsible for increased expression of NO66 in skeletal muscle via activation of nuclear factor kappa B (NF-κB). Thus, protein synthesis is likely to be negatively impacted by both a reduction in ribosomal function as well as impaired anabolic signaling in skeletal muscle during CKD (Fig. 1).

Protein degradation

Cellular and contractile proteins in skeletal muscles have different turnover rates, depending on function and structure. Myofibrillar proteins have long half-lives whereas signaling and other metabolic enzymes turn over much more rapidly. A variety of proteases and several proteolytic systems are responsible for degrading cellular proteins in skeletal muscles. For this review, only the proteolytic systems that are known to be upregulated during CKD will be discussed (Fig. 2).

Ubiquitin-proteasome system

The ubiquitin-proteasome system targets proteins for degradation by covalently linking a polyubiquitin chain to the protein (Fig. 2). These “ubiquitinated” proteins are then unfolded and degraded by a large proteasome complex that contains over 40 regulatory, structural, and proteolytic subunits. The multistep ubiquitination process is specific and exhibits substrate selectivity. First, a ubiquitin moiety is activated by an E1 activating enzyme and an E2 conjugating enzyme. This activated ubiquitin moiety is then transferred to the protein to be degraded via one of a family of 500 mammalian E3 ubiquitin ligase enzymes that recognize target proteins. Some E3 ligases are single proteins while others are multimeric complexes. They are very selective and recognize one or just a few target proteins and facilitate their ubiquitination. Proteins must be polyubiquitinated with four or more moieties in a chain to be recognized by the proteasome. Proteasomal “cap” subunits recognize and remove the polyubiquitin chains, unfold the protein, and insert it into the barrel-shaped proteasome where catalytic subunits cleave the target protein in multiple sites. This process produces small peptides and single amino acids plus recycled ubiquitin protein moieties.
During CKD and other conditions associated with muscle atrophy, the expression of ubiquitin, key proteasome subunits, and a small subset of E3 ubiquitin ligases—atrogin1, MuRF1, and FBXO40—are upregulated [20]. These E3 enzymes target a variety of myofibrillar and other key enzymes in myofibers. Key transcription factors that are responsible for these events include the FOXOs, glucocorticoid receptors (GR), and NF-κB. The anabolic resistance state in CKD reduces insulin/IGF-1/Akt signaling which decreases the inhibitory phosphorylation of the FOXOs. The increased presence of inflammatory cytokines and increased glucocorticoid production during CKD activate NF-κB and GRs, respectively. Altogether, these transcriptional responses lead to increased degradation of muscle proteins. Inhibition of these transcriptional or proteolytic responses, either genetically or pharmacologically, reduces the loss of muscle protein and helps maintain muscle function [2123].

Autophagy

In contrast to the processive degradation of select proteins by the ubiquitin-proteasome system, autophagy is a nonselective proteolytic process (Fig. 2). It degrades organelles and cytosolic proteins that are not typically targeted for degradation by the ubiquitin-proteasome system. The autophagic process starts with the formation of an autophagosome vesicle. Initiation and formation of autophagosomes involves key proteins such as beclin, BNIP3, class III phosphatidylinositol-3 kinase, the autophagy-related gene (ATG) proteins, SQSTM1/p62, and LC3. The process starts with formation of a double membrane structure called a phagophore which begins to surround portions of cytoplasm. The phagophore continues to elongate and seals to form the autophagosome. This process is highly regulated. The initial steps are suppressed by mTORC1 when the cell state favors anabolism, whereas AMPK, SIRT1, and FOXO signaling proteins upregulate autophagy when the intracellular environment favors catabolism. Once formed, autophagosomes fuse with lysosomes which contain acidic proteases that digest engulfed proteins to amino acids. In the specific case of autophagic destruction of mitochondria, the process is known as mitophagy. Destruction of damaged mitochondria via mitophagy is regulated by and depends on mTORC1 [24].
Both direct and indirect evidence supports increased autophagic activity in muscles during CKD. The FOXO transcription factors and AMPK are more active in CKD and have been linked to increased expression of several autophagic proteins (REF) [25,26]. Any suppression of autophagy by mTORC1 would seem to be minimal during CKD. Proteins and mRNAs related to autophagy have been detected in serum and in muscle samples of mice with CKD and patients with CKD [26,27]. When the muscles of CKD mice underwent acupuncture with electrical stimulation to simulate exercise, a CKD-related increase in autophagy markers was reversed [26]. The procedure also reduced the overall rate of protein degradation in muscle. Others have reported elevated markers of autophagy and mitochondrial dysfunction in the muscles of mice with CKD induced by partial nephrectomy [28]. The addition of uremic serum to cultured C2C12 myotubes replicated the mitochondrial abnormalities seen in muscle samples whereas co-incubation with n3-polyunsaturated fatty acids (n3-PUFA) reversed the actions of uremic serum [28]. Curiously, the improvement in mitochondrial function in uremic serum-treated myotubes following n3-PUFA was associated with an increase in mitophagy and the effect was blocked by siRNA inhibitors of autophagy.

Caspase-3

The third proteolytic system that becomes more active during CKD is the caspase-3 system (Fig. 2). In mononucleated cells, caspase-3 serves as a key proteolytic enzyme in the cell death or apoptosis process. However, in multinucleated skeletal muscle fibers (i.e., myofibers), caspase-3 seems to play a somewhat different role since myofibers do not undergo apoptosis per se. Signals such as inflammatory cytokines or damaged mitochondria (i.e., increased BNIP3) still activate the signaling cascade that ultimately results in cleavage of inactive pro-caspase-3 to active caspase-3. One reported action of active caspase-3 is to cleave Rpt2 and Rpt6 subunits of the proteasome [29]. This process produces a feed-forward action that increases proteasomal activity in skeletal muscle.
In the mid-1990s, actin was identified as a protein substrate of the apoptosis-related IL-1 beta-converting enzyme (ICE-like) family of cysteine proteases which have subsequently renamed caspases [30]. A 14-kDa actin fragment was a product of this cleavage. In 2004, Du et al. [31] reported that caspase-3 activity was higher in the muscles of experimental animals with CKD or other muscle-wasting conditions such as diabetes mellitus. Pharmacological inhibition of caspase-3 in incubated muscles from rats with diabetes mellitus resulted in a greater decrease in the rate of proteolysis and accumulation of the 14-kDa actin fragment than in similarly treated muscles from control rats. Importantly, the actin fragment was degraded by the proteasome. The 14-kDa actin fragment was subsequently shown to be an accurate biomarker for muscle wasting in patients undergoing dialysis and in patients following hip replacement or severe burns [32,33]. This collective evidence indicates that caspase-3 activation and actin cleavage is a critical process that contributes to muscle atrophy during CKD, diabetes mellitus, and other wasting conditions (Fig. 2).

Defective muscle regeneration

Muscles are constantly in use throughout the day. This leads to damage to the myofibers that in healthy individuals get repaired through regeneration (Fig. 1). This repair process involves macrophage-dependent removal of the damaged portions of the myofibrils and repair of the site [34]. The regenerative process is sensitive to IGF-1 and occurs in sequential steps. First, localized muscle stem cells (MSCs), which are also called satellite cells and located under the basal lamina of the muscles, become activated. The activation signals are mediators produced by local macrophages and endothelial and immune cells within the muscle fibers at the site. The activated MSCs then migrate to the repair site in the myofiber where they proliferate, differentiate into myotubes, and fuse with other myotubes and myofibers. In addition, multinucleated myotubes can fuse together to produce new fibers. In mice with CKD, satellite cell function and muscle regeneration are defective [19,35]. Markers of satellite cell activation (i.e., MyoD, myogenin), proliferation (i.e., bromodeoxyuridine incorporation), and differentiation (i.e., embryonic myosin heavy chain) were reduced. At least one signal for this regenerative dysfunction would seem to be impaired IGF-1 signaling. Deletion of the IGF-1 receptor from satellite cells recapitulated the CKD-related defects in their proliferation, differentiation, and fusion [35].
Satellite cell senescence was recently identified as a contributory process to CKD-induced dysfunctional muscle regeneration, function, and muscle atrophy [36]. Senescence refers to as an irreversible state in which cells no longer divide and proliferate. When satellite cells become senescent, they can no longer participate in the muscle regeneration process. Despite their inability to divide and proliferate, senescent cells retain a capacity to produce and secrete various growth factors, proinflammatory cytokines, and chemokines which can negatively impact other nonsenescent cells. This cellular capacity is referred to as a senescence-associated secretory phenotype or SASP. Senescent cells are frequently also resistant to apoptosis. Senescence can be induced by one of two conditions; telomere erosion such as is associated with aging or chronic cellular stresses such as DNA damage or oxidative stress. Stress-induced premature senescence can result from activation of either the p21 or the p16INK4a signaling pathways (Fig. 3). Both pathways inhibit the cyclin-dependent kinase which regulates cell cycle progression.
Muscle content of three senescence markers—senescence-associated β-galactosidase (SA-β-gal), p21, and p16INK4a—was increased in muscles of mice with CKD and was negatively correlated with weights of the gastrocnemius muscles [36]. Administration of a senolytics cocktail of dasatinib plus quercetin, compounds that induce apoptosis in senescent cells but not in proliferating or quiescent cells, led to a decline in the amounts of SA-β-gal, p21, and p16INK4a. The levels of proinflammatory cytokines, such as TNF-α and IL-6, in the muscle of CKD mice were reduced by the treatment. Markers of protein degradation (i.e., MuRF1 and atrogin-1) also declined whereas muscle function, as measured as grip strength, and average muscle fiber size increased. Notably, addition of uremic mouse serum to cultured satellite cells increased the number of p21+ cells. Co-addition of the senolytics cocktail plus uremic serum to satellite cells decreased the number of p21+ cells by increasing their apoptosis. Overexpression of constitutively activated FOXO1 in satellite cells also increased the number of p21+ cells but the action persisted despite addition of senolytics. These findings with p21 indicate that it is activated in satellite cells when FOXO activity is increased.
In summary, CKD impairs the ability of muscle to repair itself. Multiple factors, including an inefficient regenerative process and an increase in satellite cell senescence, contribute to the dysfunction. Regenerative medicine and therapies with senolytics are important continuing areas of investigation in muscle biology and CKD.

Signals responsible for muscle atrophy in chronic kidney disease

Dysfunctional insulin/insulin-like growth factor 1 signaling

Insulin and IGF-1 are critical anabolic hormones that support the health of skeletal muscle. Activation of their receptors triggers cell signaling through insulin receptor substrate-1 (IRS-1) which docks with the receptors. IRS-1 also interacts and activates phosphatidyl-3 phosphate kinase (PI3K), which in turn, phosphorylates and activates Akt (Fig. 4). Akt serves as a signaling nexus. It activates mTOR which positively regulates protein synthesis as described earlier. Akt also acts as a suppressor of protein degradation by negatively regulating the FOXO transcription factors that control atrogene expression.
Studies involving patients and animals with CKD have repeatedly found that insulin/IGF-1 signaling is impaired. The conditions underlying these defects are numerous and include dialysis, inflammation, metabolic acidosis, increased glucocorticoid production, and others [37]. Bailey et al. [18] found that IRS-1-associated PI3K kinase activity in muscle was decreased by CKD. Later studies by Thomas et al. [38] identified a membrane-associated glycoprotein, signal regulatory protein-α (SIRP-α), that is upregulated in muscle during CKD. SIRP-α interacts with the insulin/IGF-1 receptors and IRS-1. Overexpression of SIRP-α in cultured muscle cells suppressed insulin signaling whereas knockdown of SIRP-α in muscle cells overcame a cytokine-induced suppression of insulin signaling and increase in protein degradation. Other investigators have reported that phosphatase and tensin homolog (PTEN), a phosphatase that inactivates Akt, was increased in the muscles of mice with CKD [39]. When PTEN was knocked out specifically in skeletal muscle, the CKD-induced activation of protein degradation was dampened [40]. To summarize, CKD impairs insulin/IGF-1 anabolic signaling in muscle. Evidence indicates that multiple mechanisms are responsible for the aberrant insulin/IGF-1 signaling that increases FOXO-mediated atrogene responses such as MuRF1, atrogin-1, and ATG proteins and ultimately, accelerates protein degradation.

Metabolic acidosis and glucocorticoids

Normal daily metabolism generates acids that are excreted by healthy kidneys in the urine. In CKD, this urinary excretion of acids is reduced, resulting in an accumulation of acids in the blood, a condition called metabolic acidosis. Studies involving patients or animals with CKD have consistently shown how metabolic acidosis is linked to the loss of muscle mass and how correction of CKD-related metabolic acidosis improves muscle protein turnover as well as other measures of nutritional status [8,11,15,4143]. In rats with ammonium chloride-induced acidosis, physiological levels of glucocorticoids were necessary for accelerating protein degradation; however, the same levels of steroids were insufficient to elevate protein degradation in adrenalectomized nonacidotic rats [43]. Similarly, in a study of patients with CKD, arterial [HCO3] was inversely correlated with the rate of forearm protein degradation [41]. Interestingly, the subjects’ plasma cortisol levels were in the normal range; however, the cortisol levels were also inversely correlated with their arterial [HCO3]. These correlations are reminiscent of the synergistic relationship between acidosis and glucocorticoids in animal studies.
How do acidosis and glucocorticoids increase protein degradation in muscle? One obvious way for glucocorticoids to influence protein degradation is through their transcription factor functionality. Indeed, pharmacological doses of glucocorticoids increase the expression of several atrogenes including MuRF1 and myostatin [44,45]. Interestingly, these genes have binding sites for both the GR and the FOXO transcription factors suggesting the transcription factors might act synergistically. Inhibition of either the FOXO proteins or GR is sufficient to attenuate muscle loss in rats and cultured myotubes. Hu et al. [23] also reported that both glucocorticoids and insulin/IGF-1 resistance are necessary to activate protein degradation in skeletal muscles of rodents. They found that GR directly interacts with PI3K to reduce signaling via IRS-1, which results in greater FOXO activity. This nongenomic mechanism of GR is an important aspect of the atrophy process as muscle loss does not occur in adrenalectomized rodents with either metabolic acidosis or insulinopenia [46,47]. Acidosis also seems to exert actions on insulin signaling that are independent of the GR. In dogs, ammonium chloride-induced acidosis caused insulin resistance [48]. In a cultured myotube model, incubation in acidified media (pH 7.1) produced defective IRS-1-associated PI3K and Akt activities [49]. Media acidification also increased the expression of mRNAs several components of the ubiquitin-proteasome system [50]. In summary, considerable evidence indicates that acidosis and glucocorticoids have a synergistic relationship that negatively impacts insulin/IGF-1 signaling and influences atrogene expression via multiple mechanisms.

Inflammatory cytokines

Inflammation is a frequent phenotypic aspect of CKD that is strongly correlated with increased patient mortality and progression of CKD [51,52]. Acidosis, infections and sepsis, oxidative stress and mitochondrial dysfunction, obesity, premature aging and senescence, and end-stage dialysis are among the many conditions that contribute to a chronic inflammatory state in CKD.
Some inflammation is necessary for healthy muscle repair and regeneration; however, muscle atrophy occurs when levels of inflammatory cytokines are elevated above physiologic levels. Release of low levels of IL-6 and other cytokines from the muscle initiates the recruitment of M1 macrophages to the injury site where they help to clear the damaged tissue [53]. These cells are replaced by M2 macrophages and, when combined with an increase in muscle IGF-1 production, facilitate myogenesis and repair. Mice with CKD that were treated with a single session of acupuncture with electrical stimulation exhibited a post-session increase in IL-6 from muscle, relative to mice with CKD alone. Markers of myogenesis and the rate of protein degradation were also improved significantly after the treatment [54].
In other studies, elevated levels of inflammatory cytokines, especially IL-6 and TNFα, were found to induce insulin resistance, increase atrogene expression, and accelerate the rate of muscle protein degradation in rodents [5558]. Pharmacologic antagonism of these cytokines reduced both atrogene expression and protein breakdown. In muscle biopsies from late-stage nondiabetic CKD patients, TNF-α was elevated and phosphorylated Akt was decreased, relative to control patients [59]. Similarly, forearm release of IL-6 and plasma IL-6 were elevated in ESRD patients on hemodialysis and their forearm net protein balance was correlated with the release of IL-6 [60]. In summary, evidence from experimental animals as well as CKD and ESRD patients strongly suggest that elevated levels of inflammatory cytokines in the muscle milieu are strongly linked to negative protein turnover.

Myostatin and activin A

Myostatin, which is also known as growth/differentiation factor-8 or GDF-8, was discovered in 1997 as a cytokine produced by muscle fibers (i.e., a myokine), that suppresses muscle growth [61,62]. It belongs to the transforming growth factor-β (TGF-β) protein superfamily. Humans and animals with naturally occurring loss of function mutations in the myostatin gene have extensive muscle development. The myokine is produced by myofibers as an inactive proprotein that becomes active when proteolytic cleaved by bone morphogenetic protein-1. When released into the bloodstream, active myostatin can bind to the activin receptors IIa and IIb (ActRIIs) on various cell types.
In patients and animals with CKD, reduced renal clearance and increased myostatin production in muscles result in an elevated circulating level of myostatin [63,64]. When the myokine binds to ActRIIs, the receptors activate downstream cachexia-inducing signaling pathways in skeletal muscle and adipose tissue. In muscle, myostatin exerts at least three seemingly distinct actions. First, activation of the ActRIIs triggers STAT3 signaling which upregulates atrogenes expression, especially atrogin-1 and MuRF1 [64]. Second, myostatin is linked to insulin resistance in muscle and other tissues, although the responsible mechanism(s) in CKD remain unclear [65]. Third, the myokine retards myogenesis via a mechanism involving Pax-7 [66]. Satellite cell activation in adult muscle is also reduced by myostatin via a p21-dependent mechanism [67]. As noted earlier, dysfunctional myogenesis and satellite cell activation (i.e., quiescence) have been noted in CKD, suggesting that myostatin may be one of the mediators responsible for these effects [35,36].
Activin A is another member of the TGF-β protein superfamily that is closely related to myostatin. It is produced by many cells throughout the body, including some kidney cell types. It also binds to ActRIIa and ActIIb receptors and activates the same pathways as myostatin. Recently, increased production of activin A in kidney fibroblast and cells of the juxtaglomerular apparatus in mice with CKD was linked to the development of muscle loss [68]. In the same report, CKD-induced muscle atrophy in multiple mouse models was effectively prevented by antagonizing the actions of activin A by two methods—systemic blockade of ActRIIs via a receptor-ligand trap or downregulation of muscle-specific ActRII signaling using an adeno-associated virus. This study firmly established the existence of organ-to-organ crosstalk between the failing kidneys and skeletal muscle.
Given the strong links between both activin A and myostatin and muscle atrophy during CKD and other wasting conditions, myostatin/activin A signaling is an attractive therapeutic target to prevent muscle loss. In one preclinical study, the administration of an inhibitory myostatin peptibody to mice with CKD improved muscle mass and muscle protein synthesis while reducing muscle protein degradation, atrophy-related signaling, and muscle inflammation [69]. The treatment also attenuated the muscle fibrosis that developed during CKD [70]. Unfortunately, the outcomes of randomized clinical trials testing anti-myostatin therapies have been inconsistent. In the only study involving kidney diseases, an anti-myostatin peptibody (PINTA 745) treatment was stopped early due to a lack of efficacy. In two phase 2 trials of LY2495655, a myostatin-specific monoclonal antibody, inconsistent outcomes were reported. When LY2495655 was given to elderly patients (≥75 years old) after falls, the treatment group (24 weeks) experienced a small significant increase in muscle mass with some limited benefits in gait and stair climbing speeds [71]. In a different trial with arthroplasty patients given LY2495655, small increases in lean body mass were noted during the 12 weeks of treatment; however, the treatment effect was not sustained 12 weeks after treatments ended [72]. In a trial of domagrozumab, a humanized monoclonal antibody to myostatin, ambulatory boys with Duchenne muscular dystrophy were treated for 48 weeks [73]. Small but nonsignificant increases in muscle volume without functional benefit were reported. In a trial of diabetic patients given bimagrumab, a humanized monoclonal antibody that targets the activin type II receptors for 48 weeks, lean mass increased significantly although the study did not include any functional evaluation of muscle mass or strength [74]. In a 24-week trial of bimagrumab in arthroplasty patients, lean body mass increased significantly but no functional benefit was noted in the treated patients [75]. The reasons for the seemingly different outcomes between preclinical studies and patient clinical trials are unclear. There has been speculation that anti-myostatin therapies alone may not be effective since activins would likely escape the inhibition. It is also unclear whether the anti-activin receptor antibodies are equally effective against type IIA and IIB receptors. Nonetheless, this remains a promising area of investigation as the tools to target the myostatin/activin A pathway evolve.

Uremic toxins

Declining kidney function in CKD results in elevated levels of “toxic” molecules, including urea, creatinine, phosphorous, and organic acids such as indoxyl sulfate (IS) and p-cresyl sulfate. Recent evidence underscores how IS may serve as a signal for muscle wasting. In the intestines, bacteria degrade dietary tryptophan to indole which enters the circulation. In the liver, microsomal enzymes convert indole to IS which accumulates in the body during CKD due to a reduction in its clearance by the kidneys [76]. In several studies of patients with CKD or ESRD, handgrip strength was inversely related to the serum concentrations of IS [77,78].
IS has been linked to the production of reactive oxygen species, oxidative stress, and inflammation in vascular and other tissues [76]. These signals are also linked to muscle atrophy. In studies of mice with adenine-induced CKD, IS accumulated in their skeletal muscles [79]. This accumulation was accompanied by structural abnormalities in the arrangement of skeletal muscle fibers, reduction in muscle fiber size and shifts in metabolites associated with glycolysis, energy production, and antioxidants. Administration of IS to cultured myotubes produced an increase in the production of inflammatory cytokines (i.e., TNF and IL-6) as well as myostatin and atrogin-1 [80]. When IS was added to cultured myoblasts, differentiation was blocked due to suppression of key myogenic regulatory factors such as myogenic regulatory factor 4 (MRF4/Myf6) that regulate key steps in myogenic differentiation [81]. The same study confirmed that MRF4/Myf6 was suppressed in the muscles of mice with CKD. In in vitro studies, adding IS to mitochondria isolated from skeletal muscle resulted in impaired electron transport, reduced energetics, and increased hydrogen peroxide production [82]. Together, these preclinical studies support the hypothesis that IS exerts wasting effects on skeletal muscle by negatively impacting both protein turnover in myofibers and by suppressing satellite cells involved in muscle repair and maintenance.
Efforts to develop compounds that slow progression of CKD by reducing the accumulation of IS and other uremic toxins date back to the previous century. In the late 20th century, an oral activated carbon compound, AST-120, was developed and an abundance of preclinical studies and clinical trials with the compound have been conducted with the compound. Administration of AST-120 to mice with CKD for 20 weeks resulted in reductions in the levels of IS, myostatin, and atrogin-1 in hindlimb skeletal muscles [83]. A corresponding increase in phosphorylated Akt was also noted. Levels of inflammatory cytokines in muscle were unchanged by the treatment, however, muscle weights were improved following AST-120 therapy. In a similar study involving the administration of AST-120 to CKD mice for 20 weeks, the compound reversed the reduction of exercise capacity, as measured by runtime and distance [84]. Notably, several measures of mitochondrial function and biogenesis in muscle were suppressed by CKD and were normalized to the control levels by treatment with AST-120.
A considerable number of clinical trials of AST-120 have been conducted in patients with CKD over the past 2–3 decades with variable outcomes. We will summarize the findings of two meta-analyses that have been conducted within the last 5 years; both reported limited positive overall actions of AST-120 in terms of slowing the progression of CKD and lowering the level of IS. A 2019 review analyzed data from five trials from 1982 to 2013 [85]. In the second review conducted in 2021, the authors analyzed data from 15 randomized clinical trials (3,763 subjects) with a duration of treatment consisting of between 7 days and 37 months [86]. In both reviews, the authors concluded that AST-120 may slow the decline in renal function and delay the onset of dialysis. No assessment of muscle function was included in either study. According to PubMed, only one clinical trial has been conducted that investigated the effects of AST-120 on skeletal muscle health [87]. The 2022 study involved 124 CKD patients who completed the 48-week AST-120 intervention; measures of muscle health included gait speed, hand grip strength, and body composition. No significant improvements were noted in any of the muscle-related outcomes although a trend towards improving sarcopenia was noted. Given the limited positive outcomes that have been reported for AST-120, the overall benefits of the therapy remain controversial [86]. The overall importance of uremic toxins in the pathophysiology of CKD, especially with regard to muscle wasting, remains an unanswered question.

Concluding thoughts

CKD is a devastating and complex condition that impacts many organ systems of the body, including skeletal muscle. Loss of skeletal muscle mass is a frequent pathologic consequence of CKD that is referred to in the literature by various terms, depending on whether and what criteria are applied—muscle atrophy, wasting, sarcopenia, or PEW. The causes of reduced muscle mass and muscle function are multifactorial and stem from many catabolic signals present during CKD, including acidosis, inflammation, and uremic toxins. These extracellular signals alter intracellular signaling pathways that control muscle protein turnover and regeneration. The consequences of these phenotypic and metabolic changes are a state of protein and energy wasting. Therapies developed so far typically address one aspect of this disorder, leaving in place other aspects of the catabolic state. This perhaps explains why treatments have largely been unsuccessful. The most promising approaches to minimize muscle loss will likely need to address multiple aspects of the condition, much as modern multipronged cancer therapies frequently combat multiple critical cell targets.

Notes

Conflicts of interest

All authors have no conflicts of interest to declare.

Data sharing statement

The data presented in this study are available from the corresponding author upon reasonable request.

Authors’ contributions

Conceptualization, Supervision: SRP

Investigation: SRP, XHW

Writing–original draft: SRP, XHW

Writing–review & editing: SRP, XHW

All authors read and approved the final manuscript.

Figure 1.

Key processes for healthy muscle maintenance and muscle loss in CKD.

(A) In healthy muscle, several physiological processes support the growth and maintenance of skeletal muscle. Clockwise, starting in the upper left, muscle satellite cells (also called muscle precursor or stem cells) contribute to muscle repair and regeneration; following activation, they undergo self-renewal or form myoblasts which proliferate, differentiate, and fuse into myotubes. Anabolic signaling through mechanistic target of rapamycin (mTOR) supports ribosomal biogenesis and the translation of messenger RNA (mRNA) produces new muscle proteins. Protein degradation is necessary for muscle growth and maintenance. (B) In CKD, these processes are disrupted, which leads to muscle protein loss. Clockwise, starting in the upper left, reduced insulin/insulin-like growth factor 1 (Ins/IGF-1) signaling and satellite cell senescence impair muscle repair and regeneration. Diminished anabolic signaling and increased activation of nucleolar protein 66 (NO66) lead to a reduction in ribosomal biogenesis and protein synthesis. Accelerated degradation of muscle proteins produces atrophy and muscle weakness.
CKD, chronic kidney disease.
j-krcp-24-214f1.jpg
Figure 2.

Mechanisms of muscle protein loss.

Intracellular muscle proteins are degraded by different proteolytic systems. Some proteins undergo degradation by the proteasome. This process starts with a polyubiquitinylation process that involves an E1 ubiquitin-activating enzyme, an E2 ubiquitin-conjugating enzyme, and an E3 ubiquitin ligase; the E3 ligases are a family of proteins that provide specificity to the process by recognizing target proteins. Once polyubiquitinylated, the target proteins are recognized and degraded by the 26S proteasome complex. Cellular conditions like anabolic resistance are associated with mitochondrial dysfunction which leads to the release of cytochrome C and activation of caspase-3. In muscle, activated caspase-3 cleaves several regulatory subunits in the proteasome, a process that increases the activity of the protease complex. It also cleaves actin in actomyosin contractile protein complexes to produce a 14-kDa peptide which can serve as a biomarker for muscle wasting in catabolic conditions, including chronic kidney disease. Actin cleavage leads to disruption of myofibrillar proteins which can then be degraded by the ubiquitin-proteasome system. Autophagy is a proteolytic process that nonselectively degrades intracellular proteins and damaged organelles. Anabolic resistance and damaged organelles trigger a process whereby a vesicle engulfs cytoplasmic cargo, including proteins and organelles, and forms an autophagosome. Attachment of LC3-II to the vesicle designates a mature autophagosome which fuses with lysosomes that contain acidic proteases. The engulfed cargo is degraded to amino acids and other constituent components.
j-krcp-24-214f2.jpg
Figure 3.

Satellite cell senescence contributes to impaired muscle repair and regeneration during CKD.

CKD is associated with catabolic signals such as anabolic resistance and uremic toxins that activate the FOXOs and induce DNA damage. These initiator signals activate downstream signaling pathways (i.e., p21, p16) which inhibit the cell cycle, a state called senescence. In this state, cells cannot proliferate to maintain a pool of progenitor cells. They also exhibit the senescence-associated secretory phenotype (SASP) which is characterized by increased production of inflammatory cytokines. These cellular responses impair the repair and regeneration mechanisms of muscle fibers during CKD.
CKD, chronic kidney disease.
j-krcp-24-214f3.jpg
Figure 4.

Pathophysiological signals that alter proteostasis and induce muscle protein loss.

Chronic kidney disease is associated with a variety of catabolic signals such as metabolic acidosis, glucocorticoids, inflammation, myostatin, and uremic toxins. These signals impair anabolic signaling through the insulin/insulin-like growth factor 1 (IGF-1) receptors in skeletal muscle; this signaling pathway is a key regulator of protein homeostasis (i.e., proteostasis) and muscle mass. Pathway activation leads to activation of Akt which promotes protein synthesis via mechanistic target of rapamycin complex 1 (mTORC1) and suppresses the FOXO transcription factors which regulate the expression of atrophy-inducing proteins. When insulin/IGF-1 signaling is low, proteostasis becomes unbalanced; protein degradation is activated while protein synthesis and muscle regeneration are suppressed. These changes lead to muscle loss.
j-krcp-24-214f4.jpg

References

1. Janssen I, Heymsfield SB, Wang ZM, Ross R. Skeletal muscle mass and distribution in 468 men and women aged 18-88 yr. J Appl Physiol (1985) 2000;89:81–88.
crossref pmid
2. Mitch WE, Goldberg AL. Mechanisms of muscle wasting: the role of the ubiquitin-proteasome pathway. N Engl J Med 1996;335:1897–1905.
crossref pmid
3. Carrero JJ, Stenvinkel P, Cuppari L, et al. Etiology of the protein-energy wasting syndrome in chronic kidney disease: a consensus statement from the International Society of Renal Nutrition and Metabolism (ISRNM). J Ren Nutr 2013;23:77–90.
crossref pmid
4. Koppe L, Fouque D, Kalantar-Zadeh K. Kidney cachexia or protein-energy wasting in chronic kidney disease: facts and numbers. J Cachexia Sarcopenia Muscle 2019;10:479–484.
crossref pmid pmc pdf
5. Carrero JJ, Thomas F, Nagy K, et al. Global Prevalence of protein-energy wasting in kidney disease: a meta-analysis of contemporary observational studies from the International Society of Renal Nutrition and Metabolism. J Ren Nutr 2018;28:380–392.
crossref pmid
6. Lowrie EG, Lew NL. Death risk in hemodialysis patients: the predictive value of commonly measured variables and an evaluation of death rate differences between facilities. Am J Kidney Dis 1990;15:458–482.
crossref pmid
7. Sabatino A, Cuppari L, Stenvinkel P, Lindholm B, Avesani CM. Sarcopenia in chronic kidney disease: what have we learned so far? J Nephrol 2021;34:1347–1372.
crossref pmid pdf
8. Bailey JL, Wang X, England BK, Price SR, Ding X, Mitch WE. The acidosis of chronic renal failure activates muscle proteolysis in rats by augmenting transcription of genes encoding proteins of the ATP-dependent ubiquitin-proteasome pathway. J Clin Invest 1996;97:1447–1453.
crossref pmid pmc
9. Deger SM, Hung AM, Gamboa JL, et al. Systemic inflammation is associated with exaggerated skeletal muscle protein catabolism in maintenance hemodialysis patients. JCI Insight 2017;2:e95185.
crossref pmid pmc
10. Garibotto G, Sofia A, Russo R, et al. Insulin sensitivity of muscle protein metabolism is altered in patients with chronic kidney disease and metabolic acidosis. Kidney Int 2015;88:1419–1426.
crossref pmid pmc
11. Reaich D, Channon SM, Scrimgeour CM, Daley SE, Wilkinson R, Goodship TH. Correction of acidosis in humans with CRF decreases protein degradation and amino acid oxidation. Am J Physiol 1993;265:E230–E235.
crossref pmid
12. Adey D, Kumar R, McCarthy JT, Nair KS. Reduced synthesis of muscle proteins in chronic renal failure. Am J Physiol Endocrinol Metab 2000;278:E219–E225.
crossref pmid
13. Raj DS, Zager P, Shah VO, et al. Protein turnover and amino acid transport kinetics in end-stage renal disease. Am J Physiol Endocrinol Metab 2004;286:E136–E143.
crossref pmid
14. van Vliet S, Skinner SK, Beals JW, et al. Dysregulated handling of dietary protein and muscle protein synthesis after mixed-meal ingestion in maintenance hemodialysis patients. Kidney Int Rep 2018;3:1403–1415.
crossref pmid pmc
15. Garibotto G, Russo R, Sofia A, et al. Muscle protein turnover in chronic renal failure patients with metabolic acidosis or normal acid-base balance. Miner Electrolyte Metab 1996;22:58–61.
pmid
16. Wang XH, Mitch WE, Price SR. Pathophysiological mechanisms leading to muscle loss in chronic kidney disease. Nat Rev Nephrol 2022;18:138–152.
crossref pmid pdf
17. Hara Y, May RC, Kelly RA, Mitch WE. Acidosis, not azotemia, stimulates branched-chain, amino acid catabolism in uremic rats. Kidney Int 1987;32:808–814.
crossref pmid
18. Bailey JL, Zheng B, Hu Z, Price SR, Mitch WE. Chronic kidney disease causes defects in signaling through the insulin receptor substrate/phosphatidylinositol 3-kinase/Akt pathway: implications for muscle atrophy. J Am Soc Nephrol 2006;17:1388–1394.
crossref pmid
19. Zhang L, Chen Q, Chen Z, et al. Mechanisms regulating muscle protein synthesis in CKD. J Am Soc Nephrol 2020;31:2573–2587.
crossref pmid pmc
20. Wang XH, Price SR. Organ crosstalk contributes to muscle wasting in chronic kidney disease. Semin Nephrol 2023;43:151409.
crossref pmid
21. Peris-Moreno D, Taillandier D, Polge C. MuRF1/TRIM63, master regulator of muscle mass. Int J Mol Sci 2020;21:6663.
crossref pmid pmc
22. Bodine SC, Latres E, Baumhueter S, et al. Identification of ubiquitin ligases required for skeletal muscle atrophy. Science 2001;294:1704–1708.
crossref pmid
23. Hu Z, Wang H, Lee IH, Du J, Mitch WE. Endogenous glucocorticoids and impaired insulin signaling are both required to stimulate muscle wasting under pathophysiological conditions in mice. J Clin Invest 2009;119:3059–3069.
crossref pmid pmc
24. Bartolomé A, García-Aguilar A, Asahara SI, et al. MTORC1 Regulates both general autophagy and mitophagy induction after oxidative phosphorylation uncoupling. Mol Cell Biol 2017;37:e00441.
crossref pmid pmc
25. Hu R, Wang MQ, Liu LY, et al. Calycosin inhibited autophagy and oxidative stress in chronic kidney disease skeletal muscle atrophy by regulating AMPK/SKP2/CARM1 signalling pathway. J Cell Mol Med 2020;24:11084–11099.
crossref pmid pmc pdf
26. Su Z, Klein JD, Du J, et al. Chronic kidney disease induces autophagy leading to dysfunction of mitochondria in skeletal muscle. Am J Physiol Renal Physiol 2017;312:F1128–F1140.
crossref pmid pmc
27. Zhang YY, Gu LJ, Huang J, et al. CKD autophagy activation and skeletal muscle atrophy: a preliminary study of mitophagy and inflammation. Eur J Clin Nutr 2019;73:950–960.
crossref pmid pdf
28. Gortan Cappellari G, Semolic A, Ruozi G, et al. n-3 PUFA dietary lipid replacement normalizes muscle mitochondrial function and oxidative stress through enhanced tissue mitophagy and protects from muscle wasting in experimental kidney disease. Metabolism 2022;133:155242.
crossref pmid
29. Wang XH, Zhang L, Mitch WE, LeDoux JM, Hu J, Du J. Caspase-3 cleaves specific 19 S proteasome subunits in skeletal muscle stimulating proteasome activity. J Biol Chem 2010;285:21249–21257.
crossref pmid pmc
30. Mashima T, Naito M, Noguchi K, Miller DK, Nicholson DW, Tsuruo T. Actin cleavage by CPP-32/apopain during the development of apoptosis. Oncogene 1997;14:1007–1012.
crossref pmid pdf
31. Du J, Wang X, Miereles C, et al. Activation of caspase-3 is an initial step triggering accelerated muscle proteolysis in catabolic conditions. J Clin Invest 2004;113:115–123.
crossref pmid pmc
32. Workeneh BT, Rondon-Berrios H, Zhang L, et al. Development of a diagnostic method for detecting increased muscle protein degradation in patients with catabolic conditions. J Am Soc Nephrol 2006;17:3233–3239.
crossref pmid
33. Boivin MA, Battah SI, Dominic EA, et al. Activation of caspase-3 in the skeletal muscle during haemodialysis. Eur J Clin Invest 2010;40:903–910.
crossref pmid pmc
34. Yin H, Price F, Rudnicki MA. Satellite cells and the muscle stem cell niche. Physiol Rev 2013;93:23–67.
crossref pmid pmc
35. Zhang L, Wang XH, Wang H, Du J, Mitch WE. Satellite cell dysfunction and impaired IGF-1 signaling cause CKD-induced muscle atrophy. J Am Soc Nephrol 2010;21:419–427.
crossref pmid pmc
36. Huang Y, Wang B, Hassounah F, et al. The impact of senescence on muscle wasting in chronic kidney disease. J Cachexia Sarcopenia Muscle 2023;14:126–141.
crossref pmid pdf
37. Garibotto G, Saio M, Aimasso F, et al. How to overcome anabolic resistance in dialysis-treated patients? Front Nutr 2021;8:701386.
crossref pmid pmc
38. Thomas SS, Dong Y, Zhang L, Mitch WE. Signal regulatory protein-α interacts with the insulin receptor contributing to muscle wasting in chronic kidney disease. Kidney Int 2013;84:308–316.
crossref pmid pmc
39. Wang B, Zhang C, Zhang A, Cai H, Price SR, Wang XH. MicroRNA-23a and MicroRNA-27a mimic exercise by ameliorating CKD-induced muscle atrophy. J Am Soc Nephrol 2017;28:2631–2640.
crossref pmid pmc
40. Peng H, Cao J, Yu R, et al. CKD stimulates muscle protein loss via Rho-associated protein kinase 1 activation. J Am Soc Nephrol 2016;27:509–519.
crossref pmid
41. Garibotto G, Russo R, Sofia A, et al. Skeletal muscle protein synthesis and degradation in patients with chronic renal failure. Kidney Int 1994;45:1432–1439.
crossref pmid
42. Graham KA, Reaich D, Channon SM, et al. Correction of acidosis in CAPD decreases whole body protein degradation. Kidney Int 1996;49:1396–1400.
crossref pmid
43. May RC, Kelly RA, Mitch WE. Mechanisms for defects in muscle protein metabolism in rats with chronic uremia: influence of metabolic acidosis. J Clin Invest 1987;79:1099–1103.
crossref pmid pmc
44. Waddell DS, Baehr LM, van den Brandt J, et al. The glucocorticoid receptor and FOXO1 synergistically activate the skeletal muscle atrophy-associated MuRF1 gene. Am J Physiol Endocrinol Metab 2008;295:E785–E797.
crossref pmid pmc
45. Allen DL, Unterman TG. Regulation of myostatin expression and myoblast differentiation by FoxO and SMAD transcription factors. Am J Physiol Cell Physiol 2007;292:C188–C199.
crossref pmid
46. Mitch WE, Bailey JL, Wang X, Jurkovitz C, Newby D, Price SR. Evaluation of signals activating ubiquitin-proteasome proteolysis in a model of muscle wasting. Am J Physiol 1999;276:C1132–C1138.
crossref pmid
47. May RC, Kelly RA, Mitch WE. Metabolic acidosis stimulates protein degradation in rat muscle by a glucocorticoid-dependent mechanism. J Clin Invest 1986;77:614–621.
crossref pmid pmc
48. Mackler B, Lichtenstein H, Guest GM. Effects of ammonium chloride acidosis on the action of insulin in dogs. Am J Physiol 1951;166:191–198.
crossref pmid
49. Franch HA, Raissi S, Wang X, Zheng B, Bailey JL, Price SR. Acidosis impairs insulin receptor substrate-1-associated phosphoinositide 3-kinase signaling in muscle cells: consequences on proteolysis. Am J Physiol Renal Physiol 2004;287:F700–F706.
crossref pmid
50. Isozaki U, Mitch WE, England BK, Price SR. Protein degradation and increased mRNAs encoding proteins of the ubiquitin-proteasome proteolytic pathway in BC3H1 myocytes require an interaction between glucocorticoids and acidification. Proc Natl Acad Sci U S A 1996;93:1967–1971.
crossref pmid pmc
51. Ruiz-Ortega M, Rayego-Mateos S, Lamas S, Ortiz A, Rodrigues-Diez RR. Targeting the progression of chronic kidney disease. Nat Rev Nephrol 2020;16:269–288.
crossref pmid pdf
52. Ebert T, Pawelzik SC, Witasp A, et al. Inflammation and premature ageing in chronic kidney disease. Toxins (Basel) 2020;12:227.
crossref pmid pmc
53. Tidball JG, Villalta SA. Regulatory interactions between muscle and the immune system during muscle regeneration. Am J Physiol Regul Integr Comp Physiol 2010;298:R1173–R1187.
crossref pmid pmc
54. Hu L, Klein JD, Hassounah F, et al. Low-frequency electrical stimulation attenuates muscle atrophy in CKD: a potential treatment strategy. J Am Soc Nephrol 2015;26:626–635.
crossref pmid
55. Zhang L, Du J, Hu Z, et al. IL-6 and serum amyloid A synergy mediates angiotensin II-induced muscle wasting. J Am Soc Nephrol 2009;20:604–612.
crossref pmid pmc
56. Carson JA, Baltgalvis KA. Interleukin 6 as a key regulator of muscle mass during cachexia. Exerc Sport Sci Rev 2010;38:168–176.
crossref pmid pmc
57. Fischer DR, Sun X, Williams AB, et al. Dantrolene reduces serum TNFalpha and corticosterone levels and muscle calcium, calpain gene expression, and protein breakdown in septic rats. Shock 2001;15:200–207.
crossref pmid
58. Combaret L, Tilignac T, Claustre A, et al. Torbafylline (HWA 448) inhibits enhanced skeletal muscle ubiquitin-proteasome-dependent proteolysis in cancer and septic rats. Biochem J 2002;361:185–192.
crossref pmid pmc
59. Verzola D, Bonanni A, Sofia A, et al. Toll-like receptor 4 signalling mediates inflammation in skeletal muscle of patients with chronic kidney disease. J Cachexia Sarcopenia Muscle 2017;8:131–144.
crossref pmid pdf
60. Garibotto G, Sofia A, Procopio V, et al. Peripheral tissue release of interleukin-6 in patients with chronic kidney diseases: effects of end-stage renal disease and microinflammatory state. Kidney Int 2006;70:384–390.
crossref pmid
61. McPherron AC, Lawler AM, Lee SJ. Regulation of skeletal muscle mass in mice by a new TGF-beta superfamily member. Nature 1997;387:83–90.
crossref pmid pdf
62. Lee SJ, McPherron AC. Regulation of myostatin activity and muscle growth. Proc Natl Acad Sci U S A 2001;98:9306–9311.
crossref pmid pmc
63. Verzola D, Procopio V, Sofia A, et al. Apoptosis and myostatin mRNA are upregulated in the skeletal muscle of patients with chronic kidney disease. Kidney Int 2011;79:773–782.
crossref pmid
64. Zhang L, Pan J, Dong Y, et al. Stat3 activation links a C/EBPδ to myostatin pathway to stimulate loss of muscle mass. Cell Metab 2013;18:368–379.
crossref pmid pmc
65. Guo T, Jou W, Chanturiya T, Portas J, Gavrilova O, McPherron AC. Myostatin inhibition in muscle, but not adipose tissue, decreases fat mass and improves insulin sensitivity. PLoS One 2009;4:e4937.
crossref pmid pmc
66. McFarlane C, Hennebry A, Thomas M, et al. Myostatin signals through Pax7 to regulate satellite cell self-renewal. Exp Cell Res 2008;314:317–329.
crossref pmid
67. McCroskery S, Thomas M, Maxwell L, Sharma M, Kambadur R. Myostatin negatively regulates satellite cell activation and self-renewal. J Cell Biol 2003;162:1135–1147.
crossref pmid pmc pdf
68. Solagna F, Tezze C, Lindenmeyer MT, et al. Pro-cachectic factors link experimental and human chronic kidney disease to skeletal muscle wasting programs. J Clin Invest 2021;131:e135821.
crossref pmid pmc
69. Zhang L, Rajan V, Lin E, et al. Pharmacological inhibition of myostatin suppresses systemic inflammation and muscle atrophy in mice with chronic kidney disease. FASEB J 2011;25:1653–1663.
crossref pdf
70. Dong J, Dong Y, Chen Z, Mitch WE, Zhang L. The pathway to muscle fibrosis depends on myostatin stimulating the differentiation of fibro/adipogenic progenitor cells in chronic kidney disease. Kidney Int 2017;91:119–128.
crossref pmid
71. Becker C, Lord SR, Studenski SA, et al. Myostatin antibody (LY2495655) in older weak fallers: a proof-of-concept, randomised, phase 2 trial. Lancet Diabetes Endocrinol 2015;3:948–957.
crossref pmid
72. Woodhouse L, Gandhi R, Warden SJ, et al. A phase 2 randomized study investigating the efficacy and safety of myostatin antibody LY2495655 versus placebo in patients undergoing elective total hip arthroplasty. J Frailty Aging 2016;5:62–70.
crossref pmid
73. Wagner KR, Abdel-Hamid HZ, Mah JK, et al. Randomized phase 2 trial and open-label extension of domagrozumab in Duchenne muscular dystrophy. Neuromuscul Disord 2020;30:492–502.
crossref
74. Heymsfield SB, Coleman LA, Miller R, et al. Effect of bimagrumab vs placebo on body fat mass among adults with type 2 diabetes and obesity: a phase 2 randomized clinical trial. JAMA Netw Open 2021;4:e2033457.
crossref pmid pmc
75. Hofbauer LC, Witvrouw R, Varga Z, et al. Bimagrumab to improve recovery after hip fracture in older adults: a multicentre, double-blind, randomised, parallel-group, placebo-controlled, phase 2a/b trial. Lancet Healthy Longev 2021;2:e263–e274.
crossref pmid
76. Lu CL, Zheng CM, Lu KC, Liao MT, Wu KL, Ma MC. Indoxyl-sulfate-induced redox imbalance in chronic kidney disease. Antioxidants (Basel) 2021;10:936.
crossref pmid pmc
77. Lee SM, Han MY, Kim SH, et al. Indoxyl sulfate might play a role in sarcopenia, while myostatin is an indicator of muscle mass in patients with chronic kidney disease: analysis from the recovery study. Toxins (Basel) 2022;14:660.
crossref pmid pmc
78. Hou YC, Liu YM, Liao MT, et al. Indoxyl sulfate mediates low handgrip strength and is predictive of high hospitalization rates in patients with end-stage renal disease. Front Med (Lausanne) 2023;10:1023383.
crossref pmid pmc
79. Sato E, Mori T, Mishima E, et al. Metabolic alterations by indoxyl sulfate in skeletal muscle induce uremic sarcopenia in chronic kidney disease. Sci Rep 2016;6:36618.
crossref pmid pmc pdf
80. Enoki Y, Watanabe H, Arake R, et al. Indoxyl sulfate potentiates skeletal muscle atrophy by inducing the oxidative stress-mediated expression of myostatin and atrogin-1. Sci Rep 2016;6:32084.
crossref pmid pmc pdf
81. Bataille S, McKay N, Koppe L, et al. Indoxyl sulfate inhibits muscle cell differentiation via Myf6/MRF4 and MYH2 downregulation. Nephrol Dial Transplant 2023;39:103–113.
crossref pmid pdf
82. Thome T, Salyers ZR, Kumar RA, et al. Uremic metabolites impair skeletal muscle mitochondrial energetics through disruption of the electron transport system and matrix dehydrogenase activity. Am J Physiol Cell Physiol 2019;317:C701–C713.
crossref pmc
83. Enoki Y, Watanabe H, Arake R, et al. Potential therapeutic interventions for chronic kidney disease-associated sarcopenia via indoxyl sulfate-induced mitochondrial dysfunction. J Cachexia Sarcopenia Muscle 2017;8:735–747.
crossref pmid pmc pdf
84. Nishikawa M, Ishimori N, Takada S, et al. AST-120 ameliorates lowered exercise capacity and mitochondrial biogenesis in the skeletal muscle from mice with chronic kidney disease via reducing oxidative stress. Nephrol Dial Transplant 2015;30:934–942.
crossref pmid
85. Asai M, Kumakura S, Kikuchi M. Review of the efficacy of AST-120 (KREMEZIN®) on renal function in chronic kidney disease patients. Ren Fail 2019;41:47–56.
crossref pmid pmc pdf
86. Su PY, Lee YH, Kuo LN, et al. Efficacy of AST-120 for patients with chronic kidney disease: a network meta-analysis of randomized controlled trials. Front Pharmacol 2021;12:676345.
crossref pmid pmc
87. Cha RH, Kang SH, Han MY, An WS, Kim SH, Kim JC. Effects of AST-120 on muscle health and quality of life in chronic kidney disease patients: results of RECOVERY study. J Cachexia Sarcopenia Muscle 2022;13:397–408.
crossref pmid pdf


ABOUT
BROWSE ARTICLES
EDITORIAL POLICY
FOR CONTRIBUTORS
Editorial Office
#301, (Miseung Bldg.) 23, Apgujenog-ro 30-gil, Gangnam-gu, Seoul 06022, Korea
Tel: +82-2-3486-8736    Fax: +82-2-3486-8737    E-mail: registry@ksn.or.kr                

Copyright © 2025 by The Korean Society of Nephrology.

Developed in M2PI

Close layer