Int J Sports Med 2022; 43(13): 1084-1096
DOI: 10.1055/a-1797-7622
Review

Current Insights into Cellular Senescence and Myotoxicity Induced by Doxorubicin: The Role of Exercise and Growth Factors

Charalampos Andreou
1   Molecular Physiology Laboratory, Centre for Atherothrombosis & Metabolic Disease, Hull York Medical School, University of Hull
,
Antonios Matsakas
1   Molecular Physiology Laboratory, Centre for Atherothrombosis & Metabolic Disease, Hull York Medical School, University of Hull
› Author Affiliations
 

Abstract

Doxorubicin is an anti-neoplasmic drug that prevents DNA replication but induces senescence and cellular toxicity. Intensive research has focused on strategies to alleviate the doxorubicin-induced skeletal myotoxicity. The aim of the present review is to critically discuss the relevant scientific evidence about the role of exercise and growth factor administration and offer novel insights about newly developed-tools to combat the adverse drug reactions of doxorubicin treatment on skeletal muscle. In the first part, we discuss current data and mechanistic details on the impact of doxorubicin on skeletal myotoxicity. We next review key aspects about the role of regular exercise and the impact of growth factors, administered either pharmacologically or via genetic interventions. Future strategies such as combination of exercise and growth factor administration remain to be established to combat the pharmacologically-induced myotoxicity.


#

Introduction

Doxorubicin is a chemotherapeutic drug widely used against a variety of malignancies such as acute leukemia, non-Hodgkin lymphomas, Hodgkin’s disease, breast cancer, lung cancer, childhood solid tumors and sarcomas [1] [2]. Doxorubicin is an antibiotic (i. e. diminishes bacterial cell growth by inhibiting DnaG primase) of the anthracyclines group used since the 1960s, and it was the first approved liposomal injection in 1995 in the form of a hydrochloride salt of doxorubicin [3] [4] [5]. Doxorubicin exhibits antineoplastic effects by inhibiting DNA replication to cause tumor cell death [6]. In brief, intercalation of doxorubicin with the DNA splits the double-strand and induces apoptosis by inhibiting macromolecular biosynthesis [7]. Inhibition of the topoisomerase II enzyme by doxorubicin, prevents DNA replication by DNA chain-breakage and prevention of DNA double-helix resealing [8] [9]. In addition, doxorubicin is oxidized to doxorubicin semiquinone (an unstable intermediate) and returns back to doxorubicin by producing mitochondrial reactive oxygen species (ROS). The increased ROS production causes oxidative stress that leads to cell death and apoptosis, and damages the cell membrane by lipid peroxidation [10]. Increased ROS enhances p53-DNA binding to activate the DNA cross-linking and caspase signaling and results in DNA damage and apoptosis [1]. However, the doxorubicin non-specific mechanism of action has deleterious effects on healthy cells and tissues that restrict its clinical use [2]. As a matter of fact, several studies have shown that doxorubicin induces cellular senescence in various cell types [11] [12] [13] [14]. Moreover, doxorubicin contributes to cachexia (i. e. a complicated metabolic syndrome related to underlying illness including malignancy and is presented by induced inflammatory process, insulin resistance, and increased protein turnover) in cancer patients due to increased chemotherapeutic toxicity on skeletal and cardiac muscles [1] [9] [15] [16] [17]. Cachexia is a significant death contributor in 20–30% of patients and 50% of patients suffer from it [18] [19]. In addition to cachexia, sarcopenia (i. e. loss of skeletal muscle mass and strength) represents a comorbidity during cancer that affects the quality of life and increases the mortality rates of cancer patients [1] [9]. Among the main side effects of doxorubicin are nausea, hair and weight loss, fatigue, vasculature and liver toxicity, cardiotoxicity and, last but not least, skeletal muscle atrophy [1] [2] [6] [15].

The aim of this review is to assimilate how doxorubicin causes muscle atrophy, senescence and toxicity, and to discuss the current insights on the role of exercise and growth factors against doxorubicin-induced myotoxicity. The beneficial effects of exercise training on the skeletal muscle are well-established [20] [21]. Regular exercise promotes skeletal muscle functional adaptations including mitochondrial biogenesis, followed by increased antioxidant capacity [22] [23]. The alterations in muscle phenotype induced by exercise are responsible for muscle protection against stress and more specifically, against doxorubicin-induced atrophy [23] [24]. The potential mechanisms of the protective effect of exercise against the doxorubicin-induced toxicity on skeletal muscle have been reviewed previously [23] [24] [25]. Furthermore, many studies focused on the regenerative effect of individual growth factors, such as platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), epidermal growth factor (EGF), vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) on various tissues and species [26] [27] [28] [29]. These growth factors were used mainly as therapeutic strategies against various disorders such as senescence, wound healing, injury, muscular dystrophy, disease and chemotherapy. Finally, we suggest future directions for reversing the myotoxic effect of doxorubicin such as the combination of exercise and autologous biomaterials containing growth factors since their synergistic effects remain unexplored. For the present review, relevant studies were identified on PubMed by using combinations of the following keywords: doxorubicin, myotoxicity, growth factors, exercise, cachexia and cancer, in accordance with the ethical standards of the journal [30].


#

Materials and Methods

Section I: The impact of doxorubicin on skeletal myotoxicity

Doxorubicin induces skeletal muscle atrophy

Doxorubicin treatment has numerous detrimental effects on skeletal muscle biology. It has been shown to affect muscle mass and size, ROS production, proteolysis via multiple pathways, autophagy, protein synthesis and disrupt the insulin pathway [6] [23]. Doxorubicin affects skeletal muscle directly but also indirectly by being toxic for the heart, which subsequently impacts on skeletal muscle [31]. For example, a common phenomenon in congestive heart failure is the reduced cardiac function, which results in lower levels of blood flow to skeletal muscle and, as a result, muscle dysfunction. Reduced cardiac function causes skeletal muscle dysfunction, which in turns leads to muscle wasting and weakness [31] [32]. Doxorubicin causes strength reduction, maximal twitch force reduction and impaired resistance to fatigue [5] [31] [33] [34]. Experimental evidence suggests that doxorubicin induces a profound mass loss in the extensor digitorum longus (EDL) muscle [5] [6]. In addition to skeletal muscle mass loss, muscle fiber cross sectional area (CSA), capillary density and the number of muscle satellite cells are significantly reduced [35] [36]. This effect is evident in type I, type IIa and type IIx/b muscle fiber CSA in the diaphragm, plantaris and soleus muscles [6]. The various pathways involved in doxorubicin-induced atrophy in skeletal muscle are illustrated in [Fig. 1] and are discussed in the following sections. Key evidence from studies on doxorubicin administration impacting on skeletal muscle is presented in [Table 1].

Zoom Image
Fig. 1 Doxorubicin affects multiple signaling pathways that induce muscle atrophy and senescence. Doxorubicin interferes with the DNA by intercalation and inhibits the topoisomerase II enzyme, impairing the repair of DNA chain breaks, DNA replication and transcription. Oxidation of doxorubicin to doxorubicin semiquinone and back, produces mitochondrial reactive oxygen species (ROS). The increased ROS production causes cell death via oxidative stress and DNA damage by p53-DNA binding. Increased ROS damages the cell membrane by lipid peroxidation. In the red pathway: mitochondrial degradation is increased due to augmented reactive oxygen species (ROS) production. Increased ROS activates calpain-1 and caspase-3 which results in proteolysis and eventually muscle atrophy (green pathway). In addition, increased ROS induces mitochondrial degradation and subsequent muscle atrophy. In the green pathway: protein degradation is increased as a result of calpain-caspase and ubiquitin-proteasome proteolysis and induced autophagy. As myostatin (Mstn) is increased, forkhead (FOXO) family transcription factors are activated and in turn upregulate atrogin-1 and MuRF-1. In the blue pathway: protein synthesis is reduced as a result of disrupted insulin pathway. Insulin-like growth factor 1 (IGF-1) enhances protein synthesis via Akt and mTOR however doxorubicin disrupts this pathway as well as glucose transporter type 4 (GLUT4) and AMP-activated protein kinase (AMPK) involved in glucose uptake, which ends in decreased protein synthesis. Finally, in the yellow pathway: protein synthesis is decreased due to increased REDD1 through activation of p53/p21 pathway by doxorubicin. Data derived from [6] [10].

Table 1 Characteristics and outcomes from doxorubicin studies against skeletal muscle.

Reference

Species

Origin skeletal muscle sample

Dose (mg/kg)

Number of doses

Doxorubicin effect on skeletal muscle atrophy

de Lima Junior et al. 2016[1]

Wistar rats

EDL

15

1

↓: Muscle Weight & CSA; testosterone levels; AMPK; glucose uptake; IL-6/TNF-α

↑: Corticosterone levels; systemic insulin resistance

Yu et al. 2014[5]

C57BL/6 J mice

Gastrocnemius

15

1

↑: Myofibers with centralized nuclei; TUNEL apoptotic index; cell death; Bax & Bcl-2 proteins; LC3II-to-LC3I ratio

↓: p-Akt/total Akt & p-ERK/total ERK proteins

Hulmi et al. 2018[15]

C57BL/6 J mice

TA, gastrocnemius, soleus

Long- term exp: 6

Long-term: 4

↑: p21/Cdkn1a; Atrogin1 mRNA; p53 protein; Myod1 mRNA; Redd1/Ddit4

Acute exp: 15

Acute: 1

↓: Tfrc mRNA; Pgc-1α exon 1a; Pgc-1α exon 1c; Pgc-1β; Activin A; Gdf11 mRNA

Nissinen et al. 2016[18]

C57BL/6 J mice

TA, gastrocnemius, soleus

Long term exp. 1–3:6

Exp. 1–3:4

↓: Muscle weight & CSA; lean mass & fat mass; bone mineral density & bone mineral content; maximal running performance; Ulk1 & Becn1 genes; apoptosis; protein synthesis; phosphorylation of ERK ½; blood hemoglobin & hematocrit

Acute exp. 4:15

Exp.4: 1

↑: FOXO1; REDD1

Long term exp. 5–6:12

Exp. 5:2

Gilliam et al. 2016[36]

C57BL/6 N mice

Soleus

20

1

↓: Muscle weight & CSA; body weight; lean mass & fat mass; ability to scavenge H2O2; complex I- and complex II-supported respiration; maximal isometric tetanic force

↑: Mitochondrial H2O2-emitting potential: global protein carbonylation; oxidative modifications of myofibrillar proteins

Gilliam et al. 2013[38]

Sprague Dawley

rats

Gastrocnemius

20

1

↓: Body weight; lean mass & fat mass; body oxygen consumption; ambulatory activity; total energy expenditure; respiratory exchange ratios; NADH-supported respiration; FADH2-supported respiration; complex I- and complex II-supported respiration

↑: Mitochondrial H2O2-emitting potential; potential for electron leak; potential for ROS production

Hydock et al. 2011[32]

Sprague Dawley rats

Soleus & EDL

dose 1: 10

1

↓: Body mass in dose 1 & 2; maximal twitch force; maximal rate of force production; rate of force decline

dose 2: 12.5

↑: Muscle fatigue

dose 3: 15

Kavazis et al. 2014[41]

Sprague Dawley rats

Soleus

20

1

↑: FoxO1; FoxO3; Atrogin-1/MaFbx; MuRF-1 & BNIP3 mRNA in sedentary; myostatin mRNA in sedentary

↓: pAMPK/AMPK

Smuder et al. 2011[44]

Sprague Dawley rats

Soleus

20

1

↑: Carbonyl derivatives in myofibrillar protein; 4-HNE protein conjugates; CuZn-SOD; Mn-SOD; catalase; calpain; caspase-3; α-II spectrin calpain-specific cleavage; α-II spectrin caspase3-specific cleavage; easily releasable myofilaments

↓: GPX1; HSP72; actin

Smuder et al. 2011[42]

Sprague Dawley rats

Soleus

20

1

↑: Muscle Damage; TUNEL-positive nuclei; Beclin-1; Atg12 mRNA & protein; Atg12-Atg5 complex; Atg7 proteins; LC3 mRNA; LC3II to LC3I ratio; cathepsin L mRNA & protein

Bredahl et al. 2017[31]

Sprague-Dawley rats

Soleus & EDL

Incubated in 24 μM

2 incubations

↓: Rate of force production; rate of force decline

↑: Muscle fatigue

Gibson et al. 2014[2]

Sprague Dawley rats

Soleus & EDL

15

1

↑: Muscle fatigue as animal aged from 4 to 24 weeks; DOX accumulation in EDL as animal aged from 4 to 24 weeks

↓: MRP-2 & MRP-7 in EDL as animal aged from 4 to 24 weeks

Hayward et al. 2013[34]

Sprague Dawley rats

Soleus & EDL

15

1

↓: Maximal twitch force; rate of force development; rate of force decline

Sin et al. 2016[45]

SAMP8 mice

Gastrocnemius

18

1

↓: Body Mass & Muscle Mass; SIRT1 in old, deacetylase activity of SIRT1; PDK1 protein; phosphorylation of mTORSer2481; phospho-AktSer473 in young

↑: PDK4 in young; Bax protein expression; caspase 3 activity & apoptotic DNA fragmentation; MuRF-1; ubiquitinated proteins; proteasomal activity

Gouspillou et al. 2015[37]

C57BL/6 mice

Gastrocnemius, plantaris & quadricep

10

Early group: 2

↓: Body Mass & Muscle Mass; fiber size; complex I- and complex II-supported respiration; parkin protein; parkin/VDAC

Late group: 4

↑: Mitochondrial ROS production

D’Lugos et al. 2019[35]

Sprague Dawley rats

Soleus & EDL

4

3

↓: Muscle fiber size; Pax7-positive satellite cells; capillary content in soleus; MGF mRNA in EDL

↑: MYF5 mRNA in soleus

de Lima et al. 2018[47]

C57BL/6 mice

Gastrocnemius

2.5

12

↓: Body weight; fat & lean body mass; muscle weight; physical capacity; CSA; protein synthesis

↑; Basal glycemia; corticosterone


#

Doxorubicin induces oxidative stress

It has been reported that doxorubicin increases ROS production, which is responsible for increased oxidative stress and subsequent cell death [5] [31]. More specifically, doxorubicin decreases the mitochondrial respiratory capacity by inhibiting complex I- and II-supported respiration and by increasing H2O2 release, ending in reduced electron transport [6] [36] [37]. This effect on electron transport sets the basis of increased ROS [38]. Increased rate of mitochondrial H2O2 emission in skeletal muscle is possibly brought about by redox changes in the matrix such as the electron transport system and the redox-buffering system [36]. In turn, depressed oxidant scavenging within the mitochondria and increased H2O2 emission may push the redox state of the fiber to a more oxidized state. Electron transfer from O2 to aglycone in doxorubicin produces ROS (i. e. superoxide and subsequently H2O2), giving rise to potent hydroxyl radicals when reacting with iron. Doxorubicin can also form complexes with iron, disrupting iron homeostasis, leading to impaired electron flow, reduced oxygen consumption and membrane potential [39]. In addition, the respiratory control ratio (a mitochondrial uncoupling and dysfunction indicator) is decreased in response to doxorubicin. Increased ROS production by mitochondria during doxorubicin administration causes oxidative damage to DNA and to protein [38]. Under the same mechanism, in the presence of doxorubicin, lipid peroxidation forms active aldehydes such as 4–hydroxy-2–nonenal (4-HNE, a lipid peroxidation biomarker) that forms adducts with muscle proteins to exacerbate oxidative damage [40]. In line with the augmented ROS, heat shock proteins (HSPs), which are important for protein synthesis and cell protection against oxidative stress, are reduced in response to doxorubicin [6]. Persistent oxidant damage induces activation of proteases and loss of muscle tissue [39]. Taken together, the disruption of mitochondrial respiration caused by doxorubicin leads to augmented synthesis of ROS, which in turn plays a key role in the induction of cell death and skeletal muscle atrophy ([Fig. 1]).


#

Doxorubicin induces proteolysis and apoptosis and impairs protein synthesis

The ubiquitin-proteasome pathway is the main system for muscle protein degradation [16] [41]. During doxorubicin treatment, Forkhead-box (Fox) O1 and FoxO3 in muscle are elevated, which are directly associated with the activation of the E3 ligases, Atrogin-1 and MuRF-1, regulated by protein kinase B (Akt). E3 ligases regulate polyubiquitination, a key step in the ubiquitin-proteasome system which is involved in skeletal muscle proteolysis by targeting proteins for degradation [15] [16] [41] [42]. Additional studies supported this data by revealing that doxorubicin inhibits Akt phosphorylation in muscle [5] [42]. Conversely, another study, failed to identify any alterations in genes related to induced proteolysis that lead to muscle atrophy during doxorubicin administration. However, when gene set enrichment analysis (GSEA) was done to identify minor gene changes, it was shown that FoxO1, ubiquitin-proteasome pathway and apoptosis were induced [18]. Furthermore, myostatin, a potent inhibitor of myogenesis, and upstream regulator of atrogin-1 and FoxO signaling, is increased in the skeletal muscle upon doxorubicin administration [41]. These data suggest that impairment of myogenesis and muscle repair due to increased proteolysis by upregulation of ubiquitin-proteasome pathway may induce muscle atrophy.

Furthermore, doxorubicin induces muscle atrophy by increasing the activity of calpain-1 and caspase-3 proteases [43] [44]. Calpain-1 and caspase-3 are enzymes that breakdown intact myofibrillar proteins and cleave structural proteins of the skeletal muscle to cause atrophy [43] [44]. Additionally, the doxorubicin-increased mitochondrial ROS formation induces proteolysis through oxidative alterations that augments myofibrillar protein (i. e. myosin, actin, troponin I, α-actinin) exposure towards calpain-1 and caspase-3 degradation [43] [44]. In line with this, myonuclear DNA damage by doxorubicin causes apoptosis via calpain-1 and caspase-3 activation, as indicated by the high number of TUNEL-positive nuclei (i. e. apoptosis marker) in skeletal muscle [5] [42] [45]. Apoptosis is induced by doxorubicin and skeletal muscle cellular damage, which is combined with muscle atrophy to cause a decrease of muscle cell number and muscle dysfunction [5]. Therefore, calpain-1 and caspase-3 upregulation by doxorubicin induces muscle protein degradation and leads to muscle cells apoptosis ([Fig. 1]).

Doxorubicin has the potential to reduce protein synthesis through the mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway, which is related to muscle size control. Evidence suggests that, during doxorubicin treatment (i. e. four injections administered every third day for two weeks), the phosphorylation of ERK1/2 is reduced but it returns to baseline after 2–4 weeks [5] [18]. Moreover, REDD1, a protein which has a role in muscle atrophy and is associated to reduced protein synthesis, is increased in response to doxorubicin [18]. It has been shown that REDD1 is induced through the p53-p21-REDD1 pathway, as it is activated by doxorubicin [15]. Subsequently, protein synthesis is negatively affected by doxorubicin and possibly involved in muscle atrophy ([Fig. 1]).


#

Doxorubicin induces autophagy

Autophagy is a process characterized by the fusion of the autophagosome, a closed double-membrane vesicle containing a part of cytoplasm, with the lysosome, to degrade damaged organelles and protein aggregates to preserve the healthy function of cells [16]. However, it is believed that induced autophagy may result in cell death via apoptosis [6]. During doxorubicin administration, autophagy markers including Beclin-1 mRNA and protein, Atg12 mRNA and protein, Atg12–Atg5 protein, Atg7 protein and LC3 mRNA, which are essentials for the autophagosome formation and maturation, are significantly increased [42]. Increased levels of autophagic genes Atg9B and Atg18 are indicative of increased activation of autophagy in the doxorubicin-induced cellular senescence [46]. Moreover, LC3 II-to-LC3 I ratio, which indicates the formation of autophagosomes, is elevated [42]. Transient increases in autophagic signaling of autophagosome formation are reported as early as one day following a single dose of doxorubicin and return to baseline five days post-administration [5]. Furthermore, cathepsin L is augmented, whereas cathepsin B and D shows no response to doxorubicin treatment. Cathepsin B, D and L are proteases found in lysosomal, and their abundance is very high during muscle atrophy [42]. Additionally, following doxorubicin administration, BCL2/adenovirus E1B 19 kDa interacting protein 3 (BNIP3) found in skeletal muscle was increased, which is an autophagy activation protein that also induces apoptosis and mitochondrial dysfunction [41]. Taken together, it appears that elevated activation of autophagy occurs at 24 hours after doxorubicin treatment, it is normalized 5 days post-doxorubicin administration and it may induce skeletal muscle atrophy [5] [41] [42]. The molecular events of doxorubicin-induced autophagy that lead to muscle atrophy are presented in [Fig. 1].


#

Doxorubicin disrupts insulin signaling

Evidence suggests that doxorubicin chemotherapy can indirectly cause muscle atrophy through glucose intolerance. High glucose, free fatty acids and insulin levels have been detected in plasma, three days post-doxorubicin administration [1]. Even though increased insulin resistance has been detected in response to doxorubicin treatment, the insulin-like growth factor (IGF) 1 receptor, the phosphoinositide 3-kinase (PI3–K) and the Akt protein expression in skeletal muscle remained unaltered. Despite that, proteins of the insulin pathway such as insulin receptor substrate 1 (IRS-1) and glycogen synthase kinase 3 beta (GSK3-B), and protein and mRNA levels of the glucose transporter type 4 (GLUT4) and the AMP-activated protein kinase-alpha (-α) were reduced [1]. However, the AMPK modulation by doxorubicin remains controversial, as other studies showed that doxorubicin induces AMPK activation and is related to the increased cell death, apoptosis and ROS production [11] [14]. According to these studies, muscle atrophy due to doxorubicin can affect the insulin signaling pathway which in turn further induces muscle atrophy, as protein synthesis is reduced due to impaired expression of proteins related to glucose uptake [1] [6] [47]. Therefore, the doxorubicin-induced muscle atrophy disrupts the insulin signaling pathway, which in turn mediates the disruption and induces muscle atrophy ([Fig. 1]).


#

Doxorubicin induces cellular senescence

Several studies (see [Table 2]) have shown that doxorubicin induces cellular senescence in various cell types including skeletal muscle cells, embryonic ventricular myocardial cells, endothelial progenitor cells (EPCs) and vascular smooth muscle cells (VSMCs) [11] [12] [13] [14]. Some studies showed that doxorubicin induces the activation of the AMPK which leads to increased cell death and apoptosis through: the increased ROS production that damages cell DNA; the increased activation of p53 (cell death and apoptosis regulator) and JNK (apoptosis marker); and the inhibition of mammalian target of Rapamycin (mTORC)1 [11] [14]. However, increased mTOR signaling may induce senescence through reduced autophagy by the increased activation of the senescence markers p53/p21/p16. p53/p21/p16 act as tumor suppressors inducing senescence through cell cycle arrest [13]. Furthermore, increased expression of the transcription factor E2F1 has been reported in p16-defective cells showing that apoptosis due to p16 is mediated through the E2F1. Activation of E2F1 is related to cell proliferation as an oncogene or to cell death as a tumor suppressor. p16, as a tumor suppressor, has the ability to modulate the E2F1 by negative control of the mRNA decay-promoting AUF1 protein. Also increased expression of the E2F1 was seen in cells ectopically expressing p16, representing that p16 sensitizes the cells to doxorubicin through E2F1 [48]. In addition, Spallarossa et al. reported increased activity of p16 and JNK, and reduction in proliferation and cell viability [12]. Also, telomeric repeat-binding factor 2 (TRF2), a protein responsible for preserving the t-loop telomeric structure that governs chromosomal stability, was reduced. This leads to senescence by telomere shortening and dysfunction [12]. Moreover, increased miR-375 expression reduced the proliferation of K562 cells. An inversely proportional relationship was observed between miR-375 against 14–3–3zeta (anti-apoptotic gene) and SP1 genes (transcriptional regulator), which are related to cancer development and progression [46]. Reduced miR-375 expression leads in an upregulation of 14–3–3zeta and SP1 and promotes a survival effect for cancer cells. On the other hand, increased miR-375 and downregulated 14–3–3zeta and SP1 induces cellular senescence. Finally, doxorubicin induces skeletal muscle senescence through an upregulation of apoptotic and senescence markers such as JNK, p16 and p53 and reduction of anti-apoptotic markers and telomere preserving proteins such as 14–3–3zeta, SP1 genes and TRF2. Augmented proteolysis due to oxidative stress, autophagy and ubiquitin-proteasome pathway activation, in line with decreased protein synthesis due to changes in response of growth-promoting pathways, can lead to muscle atrophy and cellular senescence. Therefore, strategies including exercise and growth factor administration as potential tools against the doxorubicin-induced muscle atrophy and toxicity are critically discussed in the following section.

Table 2 Characteristics and outcomes from doxorubicin studies inducing cellular senescence.

Reference

Species

Dose (μM)

Doxorubicin effect

Yoon et al. 2019[14]

C2C12 skeletal myoblast cells

1

↑: AMPK phosphorylation; cell death; β-galactosidase

Chen et al. 2011[11]

Rat embryonic ventricular myocardial H9c2 cells

0.17, 0.52, 0.85, & 1.71

↑: AMPKα phosphorylation; cell death and apoptosis; ROS-dependent LKB1 activation; JNK activation; mTORC1 inhibition; p53 activation

Al-Khalaf et al. 2011[48]

U2OS, EH1, EH2, MEFs p16 (WT) & their p16-specific knockout counterpart, Huh7 (hepatocarcinoma cell line) & HFSN1 (primary normal human skin fibroblast)

2

↑: Apoptosis; Bax; cleaved caspase-3; E2F1

↓: NF-kB; Bcl-2 & Bcl-xL

Spallarossa et al. 2010[12]

Cord Blood (CB), (EPCs)

0.1, 0.25, 0.5, 1.0

↑: Apoptosis; β-galactosidase; p16INK4A with perinuclear accumulation; activation of p38 & JNK; F-actin disorganization

↓: TRF2 protein; proliferation; cell viability

Sung et al. 2018[13]

VSMCs

0.5

↑: mTOR signaling; p70S6K; 4E-BP1; β-galactosidase; LC3 II; expression of p53/p21/p16

Yang et al. 2012[46]

Chronic myeloid leukemic cell line K-562

0.05

↑: β-galactosidase; miR-375; miR-652; miR-22; miR139–5p; ATG9B & ATG18


#
#

Section II: The role of exercise and growth factors in mitigating the deleterious effects of doxorubicin on skeletal muscle

The Effect of Exercise on Doxorubicin-Induced Myotoxicity

Regarding the effect of exercise against tumor growth and cancer progression, the reader is directed to other relevant reviews [49] [50] [51] [52] [53]. Here, in an attempt to gain mechanistic insights of how exercise training prevents the doxorubicin-induced atrophy on the skeletal muscle, recent studies (see [Table 3]) have focused on the soleus and EDL muscles of rats exposed to endurance exercise using a treadmill [42] [54] [55]. It was previously believed that exercise-induced ROS production would aggravate the toxicity of doxorubicin. However, this is not supported by data illustrating the therapeutic effects of exercise against doxorubicin toxicity [25] [56].

Table 3 Characteristics and outcomes from exercise studies having therapeutic effects against doxorubicin.

Reference

Tissue/cell

Type of exercise

Effect of exercise on skeletal muscle

Bredahl et al. 2016[57]

Rat soleus and EDL muscle

Resistance and endurance training

Resistance training maintained maximal twitch force and maximal rate of force decline in the soleus; Endurance training reduced doxorubicin-induced fatigue in the soleus but not EDL

Bredahl et al. 2020[54]

Rat soleus and EDL muscle

Resistance training and creatine

Resistance training delayed doxorubicin-induced fatigue by 20 s in the soleus and 10 s in the EDL; Resistance training and Creatine combined, delayed doxorubicin-induced fatigue by 50 s in the soleus and 20 s in the EDL

De Lima et al. 2018[47]

Murine gastrocnemius

Aerobic exercise

↑: Maximal aerobic capacity; AMPKα phT172/total AMPKα expression

↓: Doxorubicin effect to reduce protein synthesis

Dickinson et al. 2017[58]

Rat soleus muscle

Endurance exercise

Prevents doxorubicin-induced: REDD1 mRNA; mTOR and 4E-BP1 phosphorylation reduction; LC3BII/I ratio reduction and MHC I fiber size loss

Huang et al. 2017[9]

Rat soleus muscle

Eccentric exercise

Prevents doxorubicin-induced: increased inflammation score; increased M1 macrophage

↑: M2 macrophage (CD163+)

↓: Necrotic fibers; centronucleation; TNF-α mRNA

Quinn et al. 2017[55]

Rat soleus, EDL and diaphragm muscle

Endurance exercise

↑: Myf5 in soleus and diaphragm; MyoD & Mrf4 in soleus

Kavazis et al. 2014[41]

Rat heart and soleus muscles

Short-term endurance exercise

Prevents doxorubicin-induced: increases of FoxO1 and MuRF-1 in cardiac muscle; increases of FoxO3, MuRF-1 and BNIP3 in soleus muscle

↑: PGC-1α in heart and soleus muscle

Smuder et al. 2011[42]

Rat soleus muscle

Endurance exercise

Prevents doxorubicin-induced: damaged myofiber ultrastructure; cell apoptosis; Beclin-1; Atg12 mRNA & protein; Atg7 protein; LC3 mRNA; LC3II/LC3I ratio

Smuder et al. 2011[44]

Rat soleus muscle

Endurance exercise

Prevents doxorubicin-induced: protein carbonyls; 4-HNE; calpain-to-calpastatin ratio; calpain and caspase-3 activity; degradation of actin; proteolysis

↑: GPX1 protein; HSP72 protein

Guigni et al. 2019[60]

Murine C2C12 myotubes

Muscle contraction by electrical stimulation

Prevents doxorubicin-induced: myosin loss; increased Murf1; decreased mitochondrial & Akt and FoxO3a phosphorylation.

Yoon et al. 2019[14]

Murine C2C12 cells

AICAR (endurance exercise)

↑: Phosphorylation of AMPK; cell viability

↓: Cell death

Kwon 2020[59]

Murine soleus muscle

Endurance exercise

Prevents doxorubicin-induced: irregular myofiber size; centronucleation dislocation; MHC type IIa isoform and type I composition reduction; Inhibition of the Z-line expression of α-ACTN protein; FOXO3α activation.

Bredahl et al. examined the effects of resistance training on the soleus and EDL muscle of rats against doxorubicin treatment [57]. Resistance training was achieved by a model of chronic hind limb loading while endurance training was performed on a treadmill at various speeds, inclines and durations. It was found that the maximal twitch force and the maximal rate of force decline were maintained in the soleus muscle of the doxorubicin resistance training group compared to the doxorubicin sedentary group. Bredahl et al., in a more recent study, investigated the effects of resistance training combined with creatine monohydrate administration on the soleus and EDL muscle of rats against doxorubicin-induced myotoxicity [54]. The same model of chronic hind limb loading was used and creatine was administrated after the muscles were isolated. They have shown that doxorubicin-induced fatigue was delayed by 20 s in the soleus and 10 s in the EDL post resistance training compared to the sedentary group. In addition, the doxorubicin-induced fatigue was delayed by 50 s in the soleus and 20 s in the EDL when resistance training and creatine treatment were combined [54].

Huang et al. accessed the effects of eccentric exercise (downhill running) on rat soleus muscle administered with doxorubicin [9]. The eccentric exercise protocol was an acute bout of decline treadmill running. The exercise prevented the increased inflammation score and increased M1 macrophage, which is involved in the phagocytic events during the early phase of inflammation, in the doxorubicin-treated exercised rats compared to the sedentary. M2 macrophage, which is involved in the regenerative phase of inflammation, was increased in the exercised group. Moreover, the amount of necrotic and centrally nucleated fibers was decreased [9] .

Dickinson et al. reported that interval exercise prevented the doxorubicin-induced REDD1 protein on the rat soleus muscle [58]. REDD1 negatively affects the muscle size by inhibiting mTOR signaling, compared to sedentary animals treated with doxorubicin. Moreover, it was found that exercise maintained the MHC I fiber size and the phosphorylation of the mTORC1 and its related 4E-BP1 protein which regulate muscle protein synthesis. The LC3BII/I ratio was also maintained in the exercise group compared to the sedentary group, which is related to slower mitochondrial turnover due to the maintained rate of LC3 lipidation that may eventually result in ROS formation [58].

Quinn et al. investigated the effects of short-term endurance exercise (treadmill for 2 weeks with increasing duration, steady speed and 0% grad slope) and doxorubicin treatment on the myogenic regulatory factors using the soleus, EDL and diaphragm muscles of rat [55]. They found that exercise augments the myogenic regulatory factor Myf5 in soleus and diaphragm muscles and MyoD & Mrf4 in soleus muscle compared to sedentary group treated with doxorubicin [55]. Kavazis et al. examined the effects of short-term endurance exercise (treadmill for 2 weeks with increasing duration, steady speed and 0% grad slope) on acute doxorubicin-induced FoxO transcription in cardiac and skeletal muscle of rats [41]. Exercise prevented the doxorubicin-induced increase of FoxO1 and Murf1 in cardiac muscle and the increase of FoxO3, Murf1 and BNIP3 in soleus muscle compared to the sedentary group treated with doxorubicin. Activated FoxO signaling is induced by increased ROS formation caused by doxorubicin. Upregulation of FoxO signaling leads to an increased expression of FoxO target genes including Murf1 and BNIP3 that are associated with muscle degradation and atrophy [41]. In addition, exercise increases peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α), which promotes mitochondria biogenesis and can inhibit FoxO transcriptional activity, thus protecting muscle from doxorubicin-induced atrophy [41]. Smuder et al. accessed the effects of short-term endurance exercise (treadmill for 2 weeks with increasing duration, steady speed and 0% grad slope) on doxorubicin-induced markers of autophagy signaling in the soleus muscle of rats [42]. This study found that exercise prevents the doxorubicin-induced damaged myofiber ultrastructure and cell apoptosis compared to the sedentary group administered with doxorubicin. Moreover, autophagic regulators involved in autophagosome formation and maturation such as Beclin-1, Atg12 mRNA and protein, Atg7 protein and LC3 mRNA or the LC3 II-to-LC3 I ratio, a marker of autophagosomes formation, which were induced in the control group, were prevented by the endurance exercise [42]. In another study, Smuder et al. studied the effects of short-term endurance exercise (treadmill for 2 weeks with increasing duration, steady speed and 0% grad slope) on doxorubicin-induced oxidative stress and proteolysis in the soleus muscle of rats [44]. Exercise protected muscle from preventing doxorubicin-induced proteins carbonyls and 4-HNE which increase oxidative damage. Moreover, degradation of actin and proteolysis were prevented as the calpain and caspase-3 activity, which are responsible for this damage and are related to muscle atrophy, were prevented by exercise [44]. The HSP72 and the GPX1 proteins which are responsible protein synthesis and protection against oxidative stress were upregulated by exercise [44].

Bredahl et al. examined the effects of endurance training (treadmill for 10 weeks with increasing speed, duration and slope) on the soleus and EDL muscle of rats against doxorubicin treatment [57]. The doxorubicin-induced fatigue was reduced in the soleus, but not EDL, in the endurance training group compared to the sedentary group [57]. Kwon examined the effects of endurance exercise (60 min daily for 4 weeks) on skeletal muscle remodeling against doxorubicin-induced myotoxicity in murine soleus muscle [59]. The protective effects of exercise originated in the prevention of doxorubicin to induce irregular myofiber size and central nucleation and a fiber type I transition favorable for oxidative metabolism. Exercise restored the FoxO3α to basal levels, as it was activated by doxorubicin and restored the expression of α-ACTN, a structural protein of the Z-line inhibited by doxorubicin [59]. De Lima et al. studied the impact of endurance exercise (treadmill for 6 weeks with increasing speed until exhaustion) on the murine gastrocnemius against the deleterious effects induced by the doxorubicin administration [47]. They showed that exercise increases the maximal aerobic capacity of the mice treated with doxorubicin and mitigates the negative effect of doxorubicin on protein synthesis and the doxorubicin-induced fatigue compared to the control group treated with doxorubicin. Furthermore, it was found that exercise activates AMPK, which is reduced by doxorubicin administration [47]. AMPK has a significant role in cellular metabolism regulation and when inhibited by doxorubicin impairs glucose uptake [1] [47].

Guigni et al. examined the effect of exercise using an in vitro model of contraction and mechanotransduction by electrical stimulation in C2C12 myotubes treated with doxorubicin [60]. It was shown that electrical stimulation prevents doxorubicin-induced myotube myosin content loss and increased Murf1, an E3 ligase related to muscle proteolysis compared to non-electrical stimulated cells. Additionally, the in vitro model of exercise preserved the mitochondria content and the phosphorylation of Akt and FoxO3a (Akt is activated during muscle contraction which then phosphorylates the FoxO3a) [60]. Yoon et al. studied the effects of endurance exercise on murine C2C12 cells [14]. The addition of the pharmacological AMPK agonist, 5-aminoimidazole-4-carboxamide-1-b-D-ribofuranoside (AICAR) to the cells represented an in vitro exercise mimetic model as AICAR shows similar effects to exercise including reduction of fat mass, augmentation of oxygen consumption and improvement of endurance capacity. They found that AICAR decreased cell apoptosis and increased cell viability and activation of AMPK [14]. Taken together, many forms of exercise such as endurance, resistance and eccentric exercise protects skeletal muscle from doxorubicin-induced atrophy via multiple pathways.


#

The effect of growth factors against doxorubicin-induced myotoxicity

A different approach than physical exercise to reverse doxorubicin-induced toxicity has been the administration of growth factors. The potent mitogenic, angiogenic and migration properties of growth factors that are essential in tissue regeneration are well documented. In fact, therapeutic effects of various growth factors such as PDGF, EGF, VEGF, FGF and HGF against doxorubicin-induced myotoxicity have been reported [26] [27] [28] [29]. In the next sections we discuss the findings of genetic and pharmacological studies using growth factors against doxorubicin toxicity (see [Table 4]). However, given that growth factors are also involved in tumor growth and survival, their use has to be refined to minimize potential side effects.

Table 4 Characteristics and outcomes from growth factor studies that induce myogenesis against doxorubicin myotoxicity.

Reference

Tissue/cell type

Growth factor

Dosage (Administration)

Condition (Dosage)

Effect of growth factor

Chen et al. 2018[65]

Rat cardiomyocytes

MSCs-induced VEGF release

(pharmacological)

Doxorubicin (0.5 μM)

↑: Cell viability; proliferation

↓: p53; p16; telomere shortening; telomerase activity

Lawrence et al. 1986[28]

Rat cephalad and caudad chamber

TGF-β, EGF, PDGF

100 ng/ml (pharmacological)

Doxorubicin (8 mg/kg)

TGF-β accelerates wound healing; TGF-β, EGF & PDGF combined reverse completely the inhibition of wound repair induced by doxorubicin

Yao et al. 2015[61]

Murine C2C12, human embryonic kidney 293 cells and A549 lung adenocarcinoma epithelial cells

EGF

500 or 20 ng/ml (pharmacological)

Doxorubicin (0.3 μM)

↑: GATA4 expression; Cell cycle-associated protein cyclin D1; cell viability

↓: Dox-mediated growth arrest

Koleini et al. 2017[27]

Rat cardiomyocytes

FGF-2

10 ng/ml (pharmacological)

Doxorubicin (0.5 μM)

Prevents: mitochondrial permeability transition pores mPTP formation; downregulation of transcription factor EB and lysosomal associated membrane protein-1 (LAMP-1) and cell death caused by dox.

↑: ATP; Nrf-2 protein and mRNA; HO-1 mRNA and protein; p62/SQSTM1; (p-Ser2448)-mTORC1/total mTORC1 ratio

↓: LDH activity; caspase-3; p53; Bnip-3 protein; ADP levels; ROS levels

Koleini et al. 2018[62]

Rat cardiomyocytes

non-mitogenic FGF-2

10 ng/ml (pharmacological)

Doxorubicin (0.5 μM)

Protects against Dox-induced: oxidative stress; upregulation of fragmented and non-fragmented oxidized phosphatidylcholine species

↑: P-ERK; P-p38; P-AKT

↓: Cardiomyocyte damage; cell death

Sontag et al. 2013[63]

Murine heart

FGF-2 and FGF-16

10 μg (pharmacological)

Doxorubicin (10 μM)

↓: deleterious effect of doxorubicin on left ventricular developed pressure

Wang et al. 2017[64]

Rat cardiomyocytes (vitro), mice heart (vivo)

FGF21

50 ng/ml, 100 μg/kg (pharmacological)

Doxorubicin (5 μg/m, 5 mg/kg)

↑: SIRT1) binding to liver kinase B1 (LKB1); AMPK activation

↓: TNF-α; IL6; ROS formation; apoptotic cells; Bax/Bcl-2 expression; LKB1 acetylation

Wang et al. 2018[67]

Rat heart and cardiomyocytes

FGF-16

Gene therapy (AdV transfection)

Doxorubicin (1 μM)

↑: Resistance to DOX-induced cardiomyocyte damage

↓: Annexin-V+cells; LDH activity

Räsänen et al. 2016[29]

Murine liver, heart, epididymal adipose tissue, endothelial cells, cardiac microvasculature

VEGF-B

Gene therapy (AdV transfection)

Doxorubicin (6 mg/kg)

Prevents decrease of: heart weight; cardiomyocyte size; left ventricle posterior wall; septum thickness; body mass; coronary capillary area; ERK1/2 phosphorylation by doxorubicin

Prevents damage of: microvasculature cardiac from doxorubicin

Protects from: apoptosis; endothelial dysfunction induced by doxorubicin

↑: Cytoskeleton biogenesis; angiogenesis; cell cycle-related transcripts; left ventricle mass systolic and diastolic volumes; mitochondrial DNA (mtDNA) content;

↓: DOX-induced DNA damage

Chen et al. 2010[66]

Rat cardiomyocytes

VEGF165

Gene therapy (AdV transfection)

Doxorubicin (2 μM)

↑: Bcl-2; Akt/nF-kB/Bcl-2 signaling pathway

↓: Caspase-3; apoptotic cells; FADD/caspase-8

Esaki et al. 2008[26]

Murine heart, cardiomyocytes

HGF

Gene therapy (AdV transfection)

Doxorubicin (15 mg/kg ip)

↑: Myocardial expression of GATA4; MHC; activation of ERK; c-Met/HGF receptor

↓: Left ventricular dilatation and dysfunction; cardiomyocyte atrophy/degeneration; myocardial fibrosis


#

Pharmacological administration of growth factors in doxorubicin-treated cells

Among other studies, growth factors were used against doxorubicin-induced impaired wound healing. Lawrence et al. tested the effect of transforming growth factor beta (TGF-β), EGF and PDGF, individually and synergistically, on wound chamber models extracted from doxorubicin-treated rats by incubating them with 100 ng/ml of each growth factor. It was found that TGF-β accelerates wound healing and a combination of TGF-β, EGF and PDGF was able to completely reverse the impairment of wound repair caused by doxorubicin [28]. Yao et al. used either 500 or 20 ng/ml of EGF to investigate any properties against doxorubicin toxicity on various cell lines such as murine C2C12, human embryonic kidney 293 cells and lung adenocarcinoma epithelial A549 cells [61]. It was found that cell viability was increased by the EGF as doxorubicin-mediated growth arrest was diminished by the promotion of the cell cycle-associated protein cyclin D1, which induces proliferation. The induced GATA Binding Protein 4 (GATA4) expression contributed to this augmented cell survival, as it promotes the cyclin D1 expression [61].

Koleini et al. demonstrated that FGF-2 has the capacity to protect cardiomyocytes from the cardiotoxic effects of doxorubicin via the mTOR/Nrf-2/HO-1 pathway, by incubating rat cardiomyocytes with 10 ng/ml of FGF-2 [27]. FGF-2 decreased the lactate dehydrogenase (LDH) activity (i. e. an indicator of disruption of cardiomyocyte plasma membrane integrity) and reduced ROS production and the pro-apoptotic markers such as p53, caspase-3 and BNIP3 [27]. In addition, FGF-2 reversed cell death and mitochondrial permeability transition pores (mPTP) formation caused by doxorubicin. FGF-2 increased the mRNA and protein expressions of Nrf-2 and HO-1 which are endogenous cytoprotective antioxidant regulators, and induced the mTOR activity, which controls cell growth and inhibits the initiation of autophagy [27]. Koleini et al. have also demonstrated an experiment using non-mitogenic FGF-2 against doxorubicin-induced cardiomyocyte toxicity [62]. By incubating rat cardiomyocytes with 10 ng/ml non-mitogenic FGF-2 they were able to identify a protective effect against doxorubicin toxicity. Non-mitogenic FGF-2 was able to prevent augmentation of ROS and upregulation of fragmented and non-fragmented oxidized phosphatidylcholine species. Moreover, cardiomyocyte damage and cell death were reduced whereas the phosphorylation of ERK (cardiac pro-survival kinase) was increased. Finally, they showed that the protective effect of non-mitogenic FGF2 is mediated through the FGFR1/ERK signaling [62].

Sontag et al. demonstrated the effect of 10 μg of either FGF2 or FGF16 on murine heart under doxorubicin conditions. Both FGFs had the same protecting properties of mitigating the doxorubicin-induced poisoning effect in the left ventricular developed pressure [63]. Wang et al. used 50 ng/ml of FGF21 on rat cardiomyocytes in vitro and 100 ng/ml FGF21 on mice heart in vivo to test any regenerative properties against doxorubicin-induced toxicity [64]. They showed that FGF21 induces the activation of Sirt1/liver kinase B1 (LKB1)/AMPK pathway which through this activation, doxorubicin-induced toxicity is prevented, as inflammation in heart, apoptosis and oxidative stress are suppressed. On the other hand, inflammatory cytokines related to heart dysfunction such as tumor necrosis factor (TNF-α) and IL6, and cell death related ROS production and Bax/Bcl-2 expression, were all decreased [64].

Chen et al. proved that mesenchymal stem cells (MSCs) induced the release of VEGF against the doxorubicin-induced cellular senescence on cardiomyocytes and was able to rescue the affected cells [65]. This MSCs-induced VEGF release increased the cell viability and the proliferation, decreased the p53 and p16 expression, and reduced the telomere shortening and telomerase activity compared to the control [65]. Therefore, the augmented presence of growth factors such as PDGF, EGF, FGF and VEGF either by cell incubation or boosting via a mediator, increased the wound healing process and presented protective effects on the heart against doxorubicin-induced injury and toxicity.


#

Gene therapy delivering growth factors in doxorubicin-treated cells

Gene therapy using growth factors has been used as an alternative procedure for pharmacological induction. Adenovirus (Adv) transfection was used to induce the experimental cells with the desired growth factor. Räsänen et al. used VEGF-B gene therapy in tumor-bearing mice to prevent doxorubicin-induced cardiotoxicity [29]. VEGF-B gene therapy was found to prevent the decrease of heart weight, cardiomyocyte size, left ventricle posterior wall, septum thickness, body mass, coronary capillary area and ERK1/2 phosphorylation caused by doxorubicin administration [29]. Furthermore VEGF-B prevented microvasculature cardiac damage and protected from apoptosis and endothelial dysfunction induced by doxorubicin. Cytoskeleton biogenesis, angiogenesis, cell cycle-related transcripts, left ventricle mass and systolic and diastolic volumes and mitochondrial DNA content where increased, while DNA damage induced by doxorubicin was reduced by VEGF-B gene therapy [29]. Chen et al. examined the effect of VEGF165 expression by Adv delivery on rat cardiomyocytes against doxorubicin administration [66]. According to their outcome, VEGF165 increased the Bcl-2 protein and induced the Akt/nF-kB/Bcl-2 signaling pathway. Chen et al. reported that Bcl2 is an anti-apoptotic factor as it prevents the release of cytochrome c, which can activate apoptotic factors such as caspase-9. The effects of VEGF165 on cell survival may have been brought about by the Akt/nF-kB/Bcl-2 signaling pathway. Caspase-3 and Fas-Associated protein with Death Domain (FADD)/caspase-8 which are apoptotic markers, were reduced by the VEGF165 [66]. FGF-16 has revealed protective effect on heart and cardiomyocytes against doxorubicin administration [60]. It was shown that FGF-16 increased the resistance to doxorubicin-induced cardiomyocyte damage and decreased the LDH activity as well as the apoptotic marker, annexin-V+cells [67]. Esaki et al. investigated the effect of Adv HGF delivery on mouse cardiac muscle and cardiomyocytes [26]. HGF reduced the left ventricular dilatation and dysfunction of heart, the cardiomyocyte atrophy and the myocardial fibrosis. It was also found that HGF induced the expression of GATA4 and MHC. GATA4 protein is related with antiatrophic effects on heart as it promotes cardiac growth [26]. The activation of ERK and the c-Met/HGF receptor were induced by the HGF. The ERK/MAPK pathway, which is related to heart hypertrophy, is activated through c-Met/HGF receptor signaling [26]. Overall, delivery of growth factors by cell transfection is protective against doxorubicin chemotherapy and may reverse atrophy and boost cardiac myogenesis.

A point of concern with using growth factors against doxorubicin-induced myotoxicity originates in their potent mitogenic, angiogenic and migration properties which may promote tumor proliferation and tumor angiogenesis. Several studies using growth factors to alleviate doxorubicin toxicity do not appear to have assessed their impact on tumor growth e. g. [26] [27] [28] [61] [64] [65] [66] [67]. However, VEGF-B gene therapy inhibited doxorubicin-induced cardiotoxicity without promoting tumor growth or affecting the therapeutic levels of doxorubicin [29]. Similarly, a mutated form of FGF2, carrying a serine-to-alanine substitution retained acute cardioprotective potential lacking mitogenic and angiogenic activity [62] [68] [69]. Therefore, refinements on growth factors for retaining protective properties against chemotherapy-induced toxicity are essential [62] [63] for reducing the risk of adverse effects on tumor growth or cancer cell survival.


#
#
#

Conclusion

In conclusion, many of the articles cited here have contributed to our understanding of how doxorubicin’s mechanism of action induces myotoxicity. Furthermore, other articles cited in this review have expanded our knowledge in terms of how exercise and growth factors are individually used against doxorubicin-induced myotoxicity to prevent or reverse this effect. However, as cancer mortality has reduced in the recent years, the patients suffering from the long-term effects of myotoxic antineoplastic drugs are continuously increasing. Thus, future strategies such as combination of exercise and growth factor administration remain to be established to combat the doxorubicin-induced myotoxicity. At present, evidence from exercise interventions as an adjunctive therapy, is more abundant against chemotherapy-induced myotoxicity compared to the possible side effects of growth factors, which require further research for eliminating their impact on tumor cell growth.


#

Future directions

Platelet releasate collected from aggregated platelets after the removal of cellular debris, contains multiple growth factors such as endothelial cell growth factor (ECGF), IGF, TGF-b, PDGF, VEGF, EGF and FGF known to enhance proliferation, migration and angiogenesis [70] [71]. Accumulating evidence suggests that platelet releasate can be used as an autologous biomaterial to promote skeletal myogenesis and accelerate skeletal muscle regeneration after acute injury, reduce markers of inflammation and apoptosis and promote myocellular proliferation [70] [72] [73] [74] [75]. We have recently shown that platelet releasate regulates skeletal myogenesis and muscle stem cell fate in a dose-dependent manner via the PDGF/VEGF-Cyclin D1–MyoD-Scrib-Myogenin axis and maintains the ability of satellite cell to differentiate [73] [74] [76]. However, the role of platelet releasate in myogenesis after doxorubicin administration remains undetermined. Therefore, future research is needed to determine the muscle regenerative properties of platelet releasate post-doxorubicin treatment. Furthermore, the synergistic effects of the exercise and growth factors have not experimentally addressed yet. Thus, the combination of the two strategies against doxorubicin-induced myotoxicity remains to be examined in future work to identify any synergistic effects.


#
#

Conflict of Interest

The authors declare that they have no conflict of interest.

Acknowledgements

Part of this article has been included in a dissertation submitted to the University of Hull. We apologise to those colleagues whose work could not be cited due to space limitations.

  • References

  • 1 de Lima Junior EA, Yamashita AS, Pimentel GD. et al. Doxorubicin caused severe hyperglycaemia and insulin resistance, mediated by inhibition in AMPk signalling in skeletal muscle. J Cachexia Sarcopenia Muscle 2016; 7: 615-625
  • 2 Gibson NM, Quinn CJ, Pfannenstiel KB. et al. Effects of age on multidrug resistance protein expression and doxorubicin accumulation in cardiac and skeletal muscle. Xenobiotica 2014; 44: 472-479
  • 3 Gajadeera C, Willby MJ, Green KD. et al. Antimycobacterial activity of DNA intercalator inhibitors of Mycobacterium tuberculosis primase DnaG. J Antibiot (Tokyo) 2015; 68: 153-157
  • 4 Slingerland M, Guchelaar HJ, Gelderblom H. Liposomal drug formulations in cancer therapy: 15 years along the road. Drug Discov Today 2012; 17: 160-166
  • 5 Yu AP, Pei XM, Sin TK. et al. Acylated and unacylated ghrelin inhibit doxorubicin-induced apoptosis in skeletal muscle. Acta Physiol (Oxf) 2014; 211: 201-213
  • 6 Hiensch AE, Bolam KA, Mijwel S. et al. Doxorubicin-induced skeletal muscle atrophy: Elucidating the underlying molecular pathways. Acta Physiol (Oxf) 2020; 229: e13400
  • 7 Rivankar S. An overview of doxorubicin formulations in cancer therapy. J Cancer Res Ther 2014; 10: 853-858
  • 8 Baxter-Holland M, Dass CR. Doxorubicin, mesenchymal stem cell toxicity and antitumour activity: implications for clinical use. J Pharm Pharmacol 2018; 70: 320-327
  • 9 Huang SC, Wu JF, Saovieng S. et al. Doxorubicin inhibits muscle inflammation after eccentric exercise. J Cachexia Sarcopenia Muscle 2017; 8: 277-284
  • 10 Shrestha B, Pokhrel AR, Darsandhari S et al. Engineering streptomyces peucetius for doxorubicin and daunorubicin biosynthesis. In: Arora D, Sharma C, Jaglan S et al. (Eds.). Pharmaceuticals from Microbes: The Bioengineering Perspective. Springer Nature; 2019: 191–209
  • 11 Chen MB, Wu XY, Gu JH. et al. Activation of AMP-activated protein kinase contributes to doxorubicin-induced cell death and apoptosis in cultured myocardial H9c2 cells. Cell Biochem Biophys 2011; 60: 311-322
  • 12 Spallarossa P, Altieri P, Barisione C. et al. p38 MAPK and JNK antagonistically control senescence and cytoplasmic p16INK4A expression in doxorubicin-treated endothelial progenitor cells. PLoS One 2010; 5: e15583
  • 13 Sung JY, Lee KY, Kim JR. et al. Interaction between mTOR pathway inhibition and autophagy induction attenuates adriamycin-induced vascular smooth muscle cell senescence through decreased expressions of p53/p21/p16. Exp Gerontol 2018; 109: 51-58
  • 14 Yoon KJ, Zhang D, Kim SJ. et al. Exercise-induced AMPK activation is involved in delay of skeletal muscle senescence. Biochem Biophys Res Commun 2019; 512: 604-610
  • 15 Hulmi JJ, Nissinen TA, Rasanen M. et al. Prevention of chemotherapy-induced cachexia by ACVR2B ligand blocking has different effects on heart and skeletal muscle. J Cachexia Sarcopenia Muscle 2018; 9: 417-432
  • 16 Jiao J, Demontis F. Skeletal muscle autophagy and its role in sarcopenia and organismal aging. Curr Opin Pharmacol 2017; 34: 1-6
  • 17 Yang J, Cao RY, Li Q. et al. Muscle Atrophy in Cancer. Adv Exp Med Biol 2018; 1088: 329-346
  • 18 Nissinen TA, Degerman J, Rasanen M. et al. Systemic blockade of ACVR2B ligands prevents chemotherapy-induced muscle wasting by restoring muscle protein synthesis without affecting oxidative capacity or atrogenes. Sci Rep 2016; 6: 32695
  • 19 Tisdale MJ. Mechanisms of cancer cachexia. Physiol Rev 2009; 89: 381-410
  • 20 Powers SK, Criswell D, Lawler J. et al. Influence of exercise and fiber type on antioxidant enzyme activity in rat skeletal muscle. Am J Physiol 1994; 266: R375-R380
  • 21 Powers SK, Lawler J, Criswell D. et al. Endurance-training-induced cellular adaptations in respiratory muscles. J Appl Physiol (1985) 1990; 68: 2114-2118
  • 22 Camera DM, Smiles WJ, Hawley JA. Exercise-induced skeletal muscle signaling pathways and human athletic performance. Free Radic Biol Med 2016; 98: 131-143
  • 23 Powers SK, Duarte JA, Le Nguyen B. et al. Endurance exercise protects skeletal muscle against both doxorubicin-induced and inactivity-induced muscle wasting. Pflugers Arch 2019; 471: 441-453
  • 24 Powers SK, Bomkamp M, Ozdemir M. et al. Mechanisms of exercise-induced preconditioning in skeletal muscles. Redox Biol 2020; 35: 101462
  • 25 Smuder AJ. Exercise stimulates beneficial adaptations to diminish doxorubicin-induced cellular toxicity. Am J Physiol Regul Integr Comp Physiol 2019; 317: R662-R672
  • 26 Esaki M, Takemura G, Kosai K. et al. Treatment with an adenoviral vector encoding hepatocyte growth factor mitigates established cardiac dysfunction in doxorubicin-induced cardiomyopathy. Am J Physiol Heart Circ Physiol 2008; 294: H1048-H1057
  • 27 Koleini N, Nickel BE, Wang J. et al. Fibroblast growth factor-2-mediated protection of cardiomyocytes from the toxic effects of doxorubicin requires the mTOR/Nrf-2/HO-1 pathway. Oncotarget 2017; 8: 87415-87430
  • 28 Lawrence WT, Norton JA, Sporn MB. et al. The reversal of an Adriamycin induced healing impairment with chemoattractants and growth factors. Ann Surg 1986; 203: 142-147
  • 29 Rasanen M, Degerman J, Nissinen TA. et al. VEGF-B gene therapy inhibits doxorubicin-induced cardiotoxicity by endothelial protection. Proc Natl Acad Sci U S A 2016; 113: 13144-13149
  • 30 Harriss DJ, MacSween A, Atkinson G. Ethical Standards in Sport and Exercise Science Research: 2020 Update. Int J Sports Med 2019; 40: 813-817
  • 31 Bredahl EC, Hydock DS. Creatine Supplementation and Doxorubicin-Induced Skeletal Muscle Dysfunction: An Ex Vivo Investigation. Nutr Cancer 2017; 69: 607-615
  • 32 Hydock DS, Lien CY, Jensen BT. et al. Characterization of the effect of in vivo doxorubicin treatment on skeletal muscle function in the rat. Anticancer Res 2011; 31: 2023-2028
  • 33 Gilliam LA, Moylan JS, Callahan LA. et al. Doxorubicin causes diaphragm weakness in murine models of cancer chemotherapy. Muscle Nerve 2011; 43: 94-102
  • 34 Hayward R, Hydock D, Gibson N. et al. Tissue retention of doxorubicin and its effects on cardiac, smooth, and skeletal muscle function. J Physiol Biochem 2013; 69: 177-187
  • 35 D’Lugos AC, Fry CS, Ormsby JC. et al. Chronic doxorubicin administration impacts satellite cell and capillary abundance in a muscle-specific manner. Physiol Rep 2019; 7: e14052
  • 36 Gilliam LA, Lark DS, Reese LR. et al. Targeted overexpression of mitochondrial catalase protects against cancer chemotherapy-induced skeletal muscle dysfunction. Am J Physiol Endocrinol Metab 2016; 311: E293-E301
  • 37 Gouspillou G, Scheede-Bergdahl C, Spendiff S. et al. Anthracycline-containing chemotherapy causes long-term impairment of mitochondrial respiration and increased reactive oxygen species release in skeletal muscle. Sci Rep 2015; 5: 8717
  • 38 Gilliam LAA, Fisher-Wellman KH, Lin CT. et al. The anticancer agent doxorubicin disrupts mitochondrial energy metabolism and redox balance in skeletal muscle. Free Radic Biol Med 2013; 65: 988-996
  • 39 Tarpey MD, Amorese AJ, Balestrieri NP. et al. Doxorubicin causes lesions in the electron transport system of skeletal muscle mitochondria that are associated with a loss of contractile function. J Biol Chem 2019; 294: 19709-19722
  • 40 Min K, Kwon OS, Smuder AJ. et al. Increased mitochondrial emission of reactive oxygen species and calpain activation are required for doxorubicin-induced cardiac and skeletal muscle myopathy. J Physiol 2015; 593: 2017-2036
  • 41 Kavazis AN, Smuder AJ, Powers SK. Effects of short-term endurance exercise training on acute doxorubicin-induced FoxO transcription in cardiac and skeletal muscle. J Appl Physiol (1985) 2014; 117: 223-230
  • 42 Smuder AJ, Kavazis AN, Min K. et al. Exercise protects against doxorubicin-induced markers of autophagy signaling in skeletal muscle. J Appl Physiol (1985) 2011; 111: 1190-1198
  • 43 Smuder AJ, Kavazis AN, Hudson MB. et al. Oxidation enhances myofibrillar protein degradation via calpain and caspase-3. Free Radic Biol Med 2010; 49: 1152-1160
  • 44 Smuder AJ, Kavazis AN, Min K. et al. Exercise protects against doxorubicin-induced oxidative stress and proteolysis in skeletal muscle. J Appl Physiol (1985) 2011; 110: 935-942
  • 45 Sin TK, Tam BT, Yu AP. et al. Acute Treatment of Resveratrol Alleviates Doxorubicin-Induced Myotoxicity in Aged Skeletal Muscle Through SIRT1-Dependent Mechanisms. J Gerontol A Biol Sci Med Sci 2016; 71: 730-739
  • 46 Yang MY, Lin PM, Liu YC. et al. Induction of cellular senescence by doxorubicin is associated with upregulated miR-375 and induction of autophagy in K562 cells. PLoS One 2012; 7: e37205
  • 47 de Lima EA, de Sousa LGO, de STAA. et al. Aerobic exercise, but not metformin, prevents reduction of muscular performance by AMPk activation in mice on doxorubicin chemotherapy. J Cell Physiol 2018; 233: 9652-9662
  • 48 Al-Khalaf HH, Colak D, Al-Saif M. et al. p16( INK4a) positively regulates cyclin D1 and E2F1 through negative control of AUF1. PLoS One 2011; 6: e21111
  • 49 Idorn M, Thor Straten P. Exercise and cancer: from “healthy” to “therapeutic”?. Cancer Immunol Immunother 2017; 66: 667-671
  • 50 Kim JS, Galvao DA, Newton RU. et al. Exercise-induced myokines and their effect on prostate cancer. Nat Rev Urol 2021; 18: 519-542
  • 51 Pedersen L, Christensen JF, Hojman P. Effects of exercise on tumor physiology and metabolism. Cancer J 2015; 21: 111-116
  • 52 Perez Regalado S, Leon J, Feriche B. Therapeutic approach for digestive system cancers and potential implications of exercise under hypoxia condition: what little is known? a narrative review. J Cancer Res Clin Oncol 2022; 148: 1107-1121
  • 53 Ruiz-Casado A, Martin-Ruiz A, Perez LM. et al. Exercise and the hallmarks of cancer. Trends Cancer 2017; 3: 423-441
  • 54 Bredahl EC, Busekrus RB, Hydock DS. The combined effect of creatine and resistance training on doxorubicin-induced muscle dysfunction. Nutr Cancer 2020; 72: 939-947
  • 55 Quinn CJ, Hydock DS. Effects of endurance exercise and doxorubicin on skeletal muscle myogenic regulatory factor expression. Muscles Ligaments Tendons J 2017; 7: 418-425
  • 56 Combs AB, Hudman SL, Bonner HW. Effect of exercise stress upon the acute toxicity of adriamycin in mice. Res Commun Chem Pathol Pharmacol 1979; 23: 395-398
  • 57 Bredahl EC, Pfannenstiel KB, Quinn CJ. et al. Effects of exercise on doxorubicin-induced skeletal muscle dysfunction. Med Sci Sports Exerc 2016; 48: 1468-1473
  • 58 Dickinson JM, D’Lugos AC, Mahmood TN. et al. Exercise protects skeletal muscle during chronic doxorubicin administration. Med Sci Sports Exerc 2017; 49: 2394-2403
  • 59 Kwon I. Protective effects of endurance exercise on skeletal muscle remodeling against doxorubicin-induced myotoxicity in mice. Phys Act Nutr 2020; 24: 11-21
  • 60 Guigni BA, Fix DK, Bivona JJ. et al. Electrical stimulation prevents doxorubicin-induced atrophy and mitochondrial loss in cultured myotubes. Am J Physiol Cell Physiol 2019; 317: C1213-C1228
  • 61 Yao CX, Shi JC, Ma CX. et al. EGF protects cells against dox-induced growth arrest through activating cyclin d1 expression. J Cell Biochem 2015; 116: 1755-1765
  • 62 Koleini N, Nickel BE, Edel AL. et al. Non-mitogenic FGF2 protects cardiomyocytes from acute doxorubicin-induced toxicity independently of the protein kinase CK2/heme oxygenase-1 pathway. Cell Tissue Res 2018; 374: 607-617
  • 63 Sontag DP, Wang J, Kardami E. et al. FGF-2 and FGF-16 protect isolated perfused mouse hearts from acute doxorubicin-induced contractile dysfunction. Cardiovasc Toxicol 2013; 13: 244-253
  • 64 Wang S, Wang Y, Zhang Z. et al. Cardioprotective effects of fibroblast growth factor 21 against doxorubicin-induced toxicity via the SIRT1/LKB1/AMPK pathway. Cell Death Dis 2017; 8: e3018
  • 65 Chen L, Xia W, Hou M. Mesenchymal stem cells attenuate doxorubicininduced cellular senescence through the VEGF/Notch/TGFbeta signaling pathway in H9c2 cardiomyocytes. Int J Mol Med 2018; 42: 674-684
  • 66 Chen T, Zhou G, Zhu Q. et al. Overexpression of vascular endothelial growth factor 165 (VEGF165) protects cardiomyocytes against doxorubicin-induced apoptosis. J Chemother 2010; 22: 402-406
  • 67 Wang J, Xiang B, Dolinsky VW. et al. Cardiac fgf-16 expression supports cardiomyocyte survival and increases resistance to doxorubicin cytotoxicity. DNA Cell Biol 2018; 37: 866-877
  • 68 Bailly K, Soulet F, Leroy D. et al. Uncoupling of cell proliferation and differentiation activities of basic fibroblast growth factor. FASEB J 2000; 14: 333-344
  • 69 Jiang ZS, Srisakuldee W, Soulet F. et al. Non-angiogenic FGF-2 protects the ischemic heart from injury, in the presence or absence of reperfusion. Cardiovasc Res 2004; 62: 154-166
  • 70 Scully D, Naseem KM, Matsakas A. Platelet biology in regenerative medicine of skeletal muscle. Acta Physiol (Oxf) 2018; 223: e13071
  • 71 Stellos K, Kopf S, Paul A. et al. Platelets in regeneration. Semin Thromb Hemost 2010; 36: 175-184
  • 72 Li H, Usas A, Poddar M. et al. Platelet-rich plasma promotes the proliferation of human muscle derived progenitor cells and maintains their stemness. PLoS One 2013; 8: e64923
  • 73 Scully D, Sfyri P, Verpoorten S. et al. Platelet releasate promotes skeletal myogenesis by increasing muscle stem cell commitment to differentiation and accelerates muscle regeneration following acute injury. Acta Physiol (Oxf) 2019; 225: e13207
  • 74 Scully D, Sfyri P, Wilkinson HN. et al. Optimising platelet secretomes to deliver robust tissue-specific regeneration. J Tissue Eng Regen Med 2020; 14: 82-98
  • 75 Tsai WC, Yu TY, Chang GJ. et al. Platelet-rich plasma releasate promotes regeneration and decreases inflammation and apoptosis of injured skeletal muscle. Am J Sports Med 2018; 46: 1980-1986
  • 76 Barlow J, Sfyri PP, Mitchell R. et al. Platelet releasate normalises the compromised muscle regeneration in a mouse model of hyperlipidaemia. Exp Physiol 2021; 106: 700-713

Correspondence

Dr. Antonios Matsakas
Molecular Physiology Laboratory
Centre for Atherothrombosis & Metabolic Disease
Hull York Medical School
Cottingham Road
Hull, HU6 7RX
United Kingdom   
Phone: +44(0)1482465008   

Publication History

Received: 12 December 2021

Accepted: 14 March 2022

Accepted Manuscript online:
14 March 2022

Article published online:
06 May 2022

© 2022. Thieme. All rights reserved.

Georg Thieme Verlag
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 de Lima Junior EA, Yamashita AS, Pimentel GD. et al. Doxorubicin caused severe hyperglycaemia and insulin resistance, mediated by inhibition in AMPk signalling in skeletal muscle. J Cachexia Sarcopenia Muscle 2016; 7: 615-625
  • 2 Gibson NM, Quinn CJ, Pfannenstiel KB. et al. Effects of age on multidrug resistance protein expression and doxorubicin accumulation in cardiac and skeletal muscle. Xenobiotica 2014; 44: 472-479
  • 3 Gajadeera C, Willby MJ, Green KD. et al. Antimycobacterial activity of DNA intercalator inhibitors of Mycobacterium tuberculosis primase DnaG. J Antibiot (Tokyo) 2015; 68: 153-157
  • 4 Slingerland M, Guchelaar HJ, Gelderblom H. Liposomal drug formulations in cancer therapy: 15 years along the road. Drug Discov Today 2012; 17: 160-166
  • 5 Yu AP, Pei XM, Sin TK. et al. Acylated and unacylated ghrelin inhibit doxorubicin-induced apoptosis in skeletal muscle. Acta Physiol (Oxf) 2014; 211: 201-213
  • 6 Hiensch AE, Bolam KA, Mijwel S. et al. Doxorubicin-induced skeletal muscle atrophy: Elucidating the underlying molecular pathways. Acta Physiol (Oxf) 2020; 229: e13400
  • 7 Rivankar S. An overview of doxorubicin formulations in cancer therapy. J Cancer Res Ther 2014; 10: 853-858
  • 8 Baxter-Holland M, Dass CR. Doxorubicin, mesenchymal stem cell toxicity and antitumour activity: implications for clinical use. J Pharm Pharmacol 2018; 70: 320-327
  • 9 Huang SC, Wu JF, Saovieng S. et al. Doxorubicin inhibits muscle inflammation after eccentric exercise. J Cachexia Sarcopenia Muscle 2017; 8: 277-284
  • 10 Shrestha B, Pokhrel AR, Darsandhari S et al. Engineering streptomyces peucetius for doxorubicin and daunorubicin biosynthesis. In: Arora D, Sharma C, Jaglan S et al. (Eds.). Pharmaceuticals from Microbes: The Bioengineering Perspective. Springer Nature; 2019: 191–209
  • 11 Chen MB, Wu XY, Gu JH. et al. Activation of AMP-activated protein kinase contributes to doxorubicin-induced cell death and apoptosis in cultured myocardial H9c2 cells. Cell Biochem Biophys 2011; 60: 311-322
  • 12 Spallarossa P, Altieri P, Barisione C. et al. p38 MAPK and JNK antagonistically control senescence and cytoplasmic p16INK4A expression in doxorubicin-treated endothelial progenitor cells. PLoS One 2010; 5: e15583
  • 13 Sung JY, Lee KY, Kim JR. et al. Interaction between mTOR pathway inhibition and autophagy induction attenuates adriamycin-induced vascular smooth muscle cell senescence through decreased expressions of p53/p21/p16. Exp Gerontol 2018; 109: 51-58
  • 14 Yoon KJ, Zhang D, Kim SJ. et al. Exercise-induced AMPK activation is involved in delay of skeletal muscle senescence. Biochem Biophys Res Commun 2019; 512: 604-610
  • 15 Hulmi JJ, Nissinen TA, Rasanen M. et al. Prevention of chemotherapy-induced cachexia by ACVR2B ligand blocking has different effects on heart and skeletal muscle. J Cachexia Sarcopenia Muscle 2018; 9: 417-432
  • 16 Jiao J, Demontis F. Skeletal muscle autophagy and its role in sarcopenia and organismal aging. Curr Opin Pharmacol 2017; 34: 1-6
  • 17 Yang J, Cao RY, Li Q. et al. Muscle Atrophy in Cancer. Adv Exp Med Biol 2018; 1088: 329-346
  • 18 Nissinen TA, Degerman J, Rasanen M. et al. Systemic blockade of ACVR2B ligands prevents chemotherapy-induced muscle wasting by restoring muscle protein synthesis without affecting oxidative capacity or atrogenes. Sci Rep 2016; 6: 32695
  • 19 Tisdale MJ. Mechanisms of cancer cachexia. Physiol Rev 2009; 89: 381-410
  • 20 Powers SK, Criswell D, Lawler J. et al. Influence of exercise and fiber type on antioxidant enzyme activity in rat skeletal muscle. Am J Physiol 1994; 266: R375-R380
  • 21 Powers SK, Lawler J, Criswell D. et al. Endurance-training-induced cellular adaptations in respiratory muscles. J Appl Physiol (1985) 1990; 68: 2114-2118
  • 22 Camera DM, Smiles WJ, Hawley JA. Exercise-induced skeletal muscle signaling pathways and human athletic performance. Free Radic Biol Med 2016; 98: 131-143
  • 23 Powers SK, Duarte JA, Le Nguyen B. et al. Endurance exercise protects skeletal muscle against both doxorubicin-induced and inactivity-induced muscle wasting. Pflugers Arch 2019; 471: 441-453
  • 24 Powers SK, Bomkamp M, Ozdemir M. et al. Mechanisms of exercise-induced preconditioning in skeletal muscles. Redox Biol 2020; 35: 101462
  • 25 Smuder AJ. Exercise stimulates beneficial adaptations to diminish doxorubicin-induced cellular toxicity. Am J Physiol Regul Integr Comp Physiol 2019; 317: R662-R672
  • 26 Esaki M, Takemura G, Kosai K. et al. Treatment with an adenoviral vector encoding hepatocyte growth factor mitigates established cardiac dysfunction in doxorubicin-induced cardiomyopathy. Am J Physiol Heart Circ Physiol 2008; 294: H1048-H1057
  • 27 Koleini N, Nickel BE, Wang J. et al. Fibroblast growth factor-2-mediated protection of cardiomyocytes from the toxic effects of doxorubicin requires the mTOR/Nrf-2/HO-1 pathway. Oncotarget 2017; 8: 87415-87430
  • 28 Lawrence WT, Norton JA, Sporn MB. et al. The reversal of an Adriamycin induced healing impairment with chemoattractants and growth factors. Ann Surg 1986; 203: 142-147
  • 29 Rasanen M, Degerman J, Nissinen TA. et al. VEGF-B gene therapy inhibits doxorubicin-induced cardiotoxicity by endothelial protection. Proc Natl Acad Sci U S A 2016; 113: 13144-13149
  • 30 Harriss DJ, MacSween A, Atkinson G. Ethical Standards in Sport and Exercise Science Research: 2020 Update. Int J Sports Med 2019; 40: 813-817
  • 31 Bredahl EC, Hydock DS. Creatine Supplementation and Doxorubicin-Induced Skeletal Muscle Dysfunction: An Ex Vivo Investigation. Nutr Cancer 2017; 69: 607-615
  • 32 Hydock DS, Lien CY, Jensen BT. et al. Characterization of the effect of in vivo doxorubicin treatment on skeletal muscle function in the rat. Anticancer Res 2011; 31: 2023-2028
  • 33 Gilliam LA, Moylan JS, Callahan LA. et al. Doxorubicin causes diaphragm weakness in murine models of cancer chemotherapy. Muscle Nerve 2011; 43: 94-102
  • 34 Hayward R, Hydock D, Gibson N. et al. Tissue retention of doxorubicin and its effects on cardiac, smooth, and skeletal muscle function. J Physiol Biochem 2013; 69: 177-187
  • 35 D’Lugos AC, Fry CS, Ormsby JC. et al. Chronic doxorubicin administration impacts satellite cell and capillary abundance in a muscle-specific manner. Physiol Rep 2019; 7: e14052
  • 36 Gilliam LA, Lark DS, Reese LR. et al. Targeted overexpression of mitochondrial catalase protects against cancer chemotherapy-induced skeletal muscle dysfunction. Am J Physiol Endocrinol Metab 2016; 311: E293-E301
  • 37 Gouspillou G, Scheede-Bergdahl C, Spendiff S. et al. Anthracycline-containing chemotherapy causes long-term impairment of mitochondrial respiration and increased reactive oxygen species release in skeletal muscle. Sci Rep 2015; 5: 8717
  • 38 Gilliam LAA, Fisher-Wellman KH, Lin CT. et al. The anticancer agent doxorubicin disrupts mitochondrial energy metabolism and redox balance in skeletal muscle. Free Radic Biol Med 2013; 65: 988-996
  • 39 Tarpey MD, Amorese AJ, Balestrieri NP. et al. Doxorubicin causes lesions in the electron transport system of skeletal muscle mitochondria that are associated with a loss of contractile function. J Biol Chem 2019; 294: 19709-19722
  • 40 Min K, Kwon OS, Smuder AJ. et al. Increased mitochondrial emission of reactive oxygen species and calpain activation are required for doxorubicin-induced cardiac and skeletal muscle myopathy. J Physiol 2015; 593: 2017-2036
  • 41 Kavazis AN, Smuder AJ, Powers SK. Effects of short-term endurance exercise training on acute doxorubicin-induced FoxO transcription in cardiac and skeletal muscle. J Appl Physiol (1985) 2014; 117: 223-230
  • 42 Smuder AJ, Kavazis AN, Min K. et al. Exercise protects against doxorubicin-induced markers of autophagy signaling in skeletal muscle. J Appl Physiol (1985) 2011; 111: 1190-1198
  • 43 Smuder AJ, Kavazis AN, Hudson MB. et al. Oxidation enhances myofibrillar protein degradation via calpain and caspase-3. Free Radic Biol Med 2010; 49: 1152-1160
  • 44 Smuder AJ, Kavazis AN, Min K. et al. Exercise protects against doxorubicin-induced oxidative stress and proteolysis in skeletal muscle. J Appl Physiol (1985) 2011; 110: 935-942
  • 45 Sin TK, Tam BT, Yu AP. et al. Acute Treatment of Resveratrol Alleviates Doxorubicin-Induced Myotoxicity in Aged Skeletal Muscle Through SIRT1-Dependent Mechanisms. J Gerontol A Biol Sci Med Sci 2016; 71: 730-739
  • 46 Yang MY, Lin PM, Liu YC. et al. Induction of cellular senescence by doxorubicin is associated with upregulated miR-375 and induction of autophagy in K562 cells. PLoS One 2012; 7: e37205
  • 47 de Lima EA, de Sousa LGO, de STAA. et al. Aerobic exercise, but not metformin, prevents reduction of muscular performance by AMPk activation in mice on doxorubicin chemotherapy. J Cell Physiol 2018; 233: 9652-9662
  • 48 Al-Khalaf HH, Colak D, Al-Saif M. et al. p16( INK4a) positively regulates cyclin D1 and E2F1 through negative control of AUF1. PLoS One 2011; 6: e21111
  • 49 Idorn M, Thor Straten P. Exercise and cancer: from “healthy” to “therapeutic”?. Cancer Immunol Immunother 2017; 66: 667-671
  • 50 Kim JS, Galvao DA, Newton RU. et al. Exercise-induced myokines and their effect on prostate cancer. Nat Rev Urol 2021; 18: 519-542
  • 51 Pedersen L, Christensen JF, Hojman P. Effects of exercise on tumor physiology and metabolism. Cancer J 2015; 21: 111-116
  • 52 Perez Regalado S, Leon J, Feriche B. Therapeutic approach for digestive system cancers and potential implications of exercise under hypoxia condition: what little is known? a narrative review. J Cancer Res Clin Oncol 2022; 148: 1107-1121
  • 53 Ruiz-Casado A, Martin-Ruiz A, Perez LM. et al. Exercise and the hallmarks of cancer. Trends Cancer 2017; 3: 423-441
  • 54 Bredahl EC, Busekrus RB, Hydock DS. The combined effect of creatine and resistance training on doxorubicin-induced muscle dysfunction. Nutr Cancer 2020; 72: 939-947
  • 55 Quinn CJ, Hydock DS. Effects of endurance exercise and doxorubicin on skeletal muscle myogenic regulatory factor expression. Muscles Ligaments Tendons J 2017; 7: 418-425
  • 56 Combs AB, Hudman SL, Bonner HW. Effect of exercise stress upon the acute toxicity of adriamycin in mice. Res Commun Chem Pathol Pharmacol 1979; 23: 395-398
  • 57 Bredahl EC, Pfannenstiel KB, Quinn CJ. et al. Effects of exercise on doxorubicin-induced skeletal muscle dysfunction. Med Sci Sports Exerc 2016; 48: 1468-1473
  • 58 Dickinson JM, D’Lugos AC, Mahmood TN. et al. Exercise protects skeletal muscle during chronic doxorubicin administration. Med Sci Sports Exerc 2017; 49: 2394-2403
  • 59 Kwon I. Protective effects of endurance exercise on skeletal muscle remodeling against doxorubicin-induced myotoxicity in mice. Phys Act Nutr 2020; 24: 11-21
  • 60 Guigni BA, Fix DK, Bivona JJ. et al. Electrical stimulation prevents doxorubicin-induced atrophy and mitochondrial loss in cultured myotubes. Am J Physiol Cell Physiol 2019; 317: C1213-C1228
  • 61 Yao CX, Shi JC, Ma CX. et al. EGF protects cells against dox-induced growth arrest through activating cyclin d1 expression. J Cell Biochem 2015; 116: 1755-1765
  • 62 Koleini N, Nickel BE, Edel AL. et al. Non-mitogenic FGF2 protects cardiomyocytes from acute doxorubicin-induced toxicity independently of the protein kinase CK2/heme oxygenase-1 pathway. Cell Tissue Res 2018; 374: 607-617
  • 63 Sontag DP, Wang J, Kardami E. et al. FGF-2 and FGF-16 protect isolated perfused mouse hearts from acute doxorubicin-induced contractile dysfunction. Cardiovasc Toxicol 2013; 13: 244-253
  • 64 Wang S, Wang Y, Zhang Z. et al. Cardioprotective effects of fibroblast growth factor 21 against doxorubicin-induced toxicity via the SIRT1/LKB1/AMPK pathway. Cell Death Dis 2017; 8: e3018
  • 65 Chen L, Xia W, Hou M. Mesenchymal stem cells attenuate doxorubicininduced cellular senescence through the VEGF/Notch/TGFbeta signaling pathway in H9c2 cardiomyocytes. Int J Mol Med 2018; 42: 674-684
  • 66 Chen T, Zhou G, Zhu Q. et al. Overexpression of vascular endothelial growth factor 165 (VEGF165) protects cardiomyocytes against doxorubicin-induced apoptosis. J Chemother 2010; 22: 402-406
  • 67 Wang J, Xiang B, Dolinsky VW. et al. Cardiac fgf-16 expression supports cardiomyocyte survival and increases resistance to doxorubicin cytotoxicity. DNA Cell Biol 2018; 37: 866-877
  • 68 Bailly K, Soulet F, Leroy D. et al. Uncoupling of cell proliferation and differentiation activities of basic fibroblast growth factor. FASEB J 2000; 14: 333-344
  • 69 Jiang ZS, Srisakuldee W, Soulet F. et al. Non-angiogenic FGF-2 protects the ischemic heart from injury, in the presence or absence of reperfusion. Cardiovasc Res 2004; 62: 154-166
  • 70 Scully D, Naseem KM, Matsakas A. Platelet biology in regenerative medicine of skeletal muscle. Acta Physiol (Oxf) 2018; 223: e13071
  • 71 Stellos K, Kopf S, Paul A. et al. Platelets in regeneration. Semin Thromb Hemost 2010; 36: 175-184
  • 72 Li H, Usas A, Poddar M. et al. Platelet-rich plasma promotes the proliferation of human muscle derived progenitor cells and maintains their stemness. PLoS One 2013; 8: e64923
  • 73 Scully D, Sfyri P, Verpoorten S. et al. Platelet releasate promotes skeletal myogenesis by increasing muscle stem cell commitment to differentiation and accelerates muscle regeneration following acute injury. Acta Physiol (Oxf) 2019; 225: e13207
  • 74 Scully D, Sfyri P, Wilkinson HN. et al. Optimising platelet secretomes to deliver robust tissue-specific regeneration. J Tissue Eng Regen Med 2020; 14: 82-98
  • 75 Tsai WC, Yu TY, Chang GJ. et al. Platelet-rich plasma releasate promotes regeneration and decreases inflammation and apoptosis of injured skeletal muscle. Am J Sports Med 2018; 46: 1980-1986
  • 76 Barlow J, Sfyri PP, Mitchell R. et al. Platelet releasate normalises the compromised muscle regeneration in a mouse model of hyperlipidaemia. Exp Physiol 2021; 106: 700-713

Zoom Image
Fig. 1 Doxorubicin affects multiple signaling pathways that induce muscle atrophy and senescence. Doxorubicin interferes with the DNA by intercalation and inhibits the topoisomerase II enzyme, impairing the repair of DNA chain breaks, DNA replication and transcription. Oxidation of doxorubicin to doxorubicin semiquinone and back, produces mitochondrial reactive oxygen species (ROS). The increased ROS production causes cell death via oxidative stress and DNA damage by p53-DNA binding. Increased ROS damages the cell membrane by lipid peroxidation. In the red pathway: mitochondrial degradation is increased due to augmented reactive oxygen species (ROS) production. Increased ROS activates calpain-1 and caspase-3 which results in proteolysis and eventually muscle atrophy (green pathway). In addition, increased ROS induces mitochondrial degradation and subsequent muscle atrophy. In the green pathway: protein degradation is increased as a result of calpain-caspase and ubiquitin-proteasome proteolysis and induced autophagy. As myostatin (Mstn) is increased, forkhead (FOXO) family transcription factors are activated and in turn upregulate atrogin-1 and MuRF-1. In the blue pathway: protein synthesis is reduced as a result of disrupted insulin pathway. Insulin-like growth factor 1 (IGF-1) enhances protein synthesis via Akt and mTOR however doxorubicin disrupts this pathway as well as glucose transporter type 4 (GLUT4) and AMP-activated protein kinase (AMPK) involved in glucose uptake, which ends in decreased protein synthesis. Finally, in the yellow pathway: protein synthesis is decreased due to increased REDD1 through activation of p53/p21 pathway by doxorubicin. Data derived from [6] [10].