Next Article in Journal
Polyphyllin VI Induces Caspase-1-Mediated Pyroptosis via the Induction of ROS/NF-κB/NLRP3/GSDMD Signal Axis in Non-Small Cell Lung Cancer
Next Article in Special Issue
Nicotine Induces IL-8 Secretion from Pancreatic Cancer Stroma and Worsens Cancer-Induced Cachexia
Previous Article in Journal
Current Status and Future Perspectives of Immunotherapy for Locally Advanced or Metastatic Urothelial Carcinoma: A Comprehensive Review
Previous Article in Special Issue
Upregulation of ZIP14 and Altered Zinc Homeostasis in Muscles in Pancreatic Cancer Cachexia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Systemic Actions of Breast Cancer Facilitate Functional Limitations

by
Ruizhong Wang
1 and
Harikrishna Nakshatri
1,2,3,*
1
Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
2
Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
3
VA Roudebush Medical Center, Indianapolis, IN 46202, USA
*
Author to whom correspondence should be addressed.
Cancers 2020, 12(1), 194; https://doi.org/10.3390/cancers12010194
Submission received: 4 December 2019 / Revised: 6 January 2020 / Accepted: 9 January 2020 / Published: 13 January 2020
(This article belongs to the Special Issue Cancer Cachexia)

Abstract

:
Breast cancer is a disease of a specific organ, but its effects are felt throughout the body. The systemic effects of breast cancer can lead to functional limitations in patients who suffer from muscle weakness, fatigue, pain, fibromyalgia, or many other dysfunctions, which hasten cancer-associated death. Mechanistic studies have identified quite a few molecular defects in skeletal muscles that are associated with functional limitations in breast cancer. These include circulating cytokines such as TNF-α, IL-1, IL-6, and TGF-β altering the levels or function of myogenic molecules including PAX7, MyoD, and microRNAs through transcriptional regulators such as NF-κB, STAT3, and SMADs. Molecular defects in breast cancer may also include reduced muscle mitochondrial content and increased extracellular matrix deposition leading to energy imbalance and skeletal muscle fibrosis. This review highlights recent evidence that breast cancer-associated molecular defects mechanistically contribute to functional limitations and further provides insights into therapeutic interventions in managing functional limitations, which in turn may help to improve quality of life in breast cancer patients.

1. Prevalence of Functional Limitations in Breast Cancer

Breast cancer is one of the most common malignancies affecting one in eight women in the United States. With disease progression, breast cancer causes dramatic systemic effects. Myopenia and cachexia under metastatic settings are observed in ~25% of breast cancer patients, particularly in women with triple-negative breast cancer [1,2]. Functional limitations are observed even before cancer diagnosis and are the major reasons for increased no-cancer related deaths in these patients [3,4]. In a recent study, a cohort of 2202 women with breast cancer was examined and approximately 40% breast cancer patients demonstrated at least one of the functional limitations [5]. Patients with functional limitations exhibit decreased volitional activity, whole body weakness, and fatigue [6,7,8,9,10]. The prevalence of functional limitations in breast cancer patients is dependent on disease stage, age, ethnicity, demographics, and physical compositions [5]. Indeed, functional limitations are more common among older breast cancer patients [5,11]. Women with functional limitations are more likely to be overweight or obese with less physically activity [5,12]. More critically, preclinical and clinical data demonstrate that functional limitations result in significantly shorter survival due to non-cancer causes of death [5,13].

2. Functional Limitation is Likely Due to Skeletal Muscle Dysfunction in Breast Cancer

Growing evidence points to skeletal muscle dysfunction as a major cause of cancer-associated functional limitations. Skeletal muscles represent 40%–50% of the total body mass of a healthy human, making them collectively one of the largest organ systems [14,15]. A defining characteristic of skeletal muscles is its remarkable capacity of generating power for motion of an individual to maintain activities and to ensure the quality of life [16,17]. Thus, muscle force production, fatigue, and twitch characteristics are functional properties that define skeletal muscle quality. Life quality change associated with aging, Duchenne muscular dystrophy (DMD), chronic obstructive pulmonary disease (COPD), and cancer is directly related to these muscle parameters [18,19,20,21,22]. Recently, specific decrements to skeletal muscle’s contractile quality have been identified in cachectic patients and tumor-bearing mice. Women with breast cancer are found to have reduced cross-sectional areas of a single muscle fiber compared with cancer-free controls. In addition, women with breast cancer have reduced fractional content of both subsarcolemmal and intermyofibrillar mitochondria [23]. This observation is consistent with a higher risk of dying due to low levels of muscle mass in women with breast cancer compared to women who have adequate muscle mass [24]. Furthermore, women with higher muscle mass demonstrate improved response to cancer treatment and overall survival [25,26]. Preclinical data from animal models of breast cancer demonstrated that mice with primary mammary tumors have reduced grip strength and declined motor activity [13]. In line with functional limitations, reduction of body fat and increased body free water were observed in the transgenic PyMT mouse model, which represents the luminal B subtype of human breast cancer. Tumor progression also caused decrement of grip strength and reduction of specific force generated by muscle contraction [27], which might be related to the decreased size of muscle fibers and muscle mitochondria [23]. Moreover, breast cancer-associated functional limitations, including muscle weakness and reduced muscle force, correlate with dynamic systemic changes in cytokine/chemokine levels and myogenic regulation (Figure 1). This process involves complex gene expression regulatory networks in the skeletal muscle differentiation hierarchy, which is influenced by the chemokines and cytokines (Figure 1). In breast cancer bone metastasis models, body weight loss is associated with skeletal muscle atrophy and specific changes in the activity of skeletal muscle ryanodine receptors and the calcium (Ca2+) release channel [27].

3. Altered Cytokines Contribute to Functional Limitations in Breast Cancer

Cytokines are a large family of polypeptides and small proteins, including interleukin (e.g., IL-1, IL-2, IL-4), interferon (IFN-α, β, γ), chemokine, and tumor necrosis factor (TNF) families. Cytokines produced by a variety of cell types exert their actions locally (autocrine and paracrine) or systemically by directly interacting with their specific membrane receptors [28,29]. It has been well established that cytokines secreted by host immune cells in response to tumors and/or by the tumor itself are associated with muscle wasting and functional limitations [30,31]. High plasma levels of IL-6, TNF-α, INF-γ, and IL-1β have been observed in both tumor-bearing animals and cachectic cancer patients [32,33]. Fatigued women who survive breast cancer have significantly higher serum levels of IL-1ra, sTNF-RII, and neopterin than non-fatigued women [34]. This cancer-associated increase in pro-inflammatory activity is common across many diseases including autoimmune, inflammatory, and infectious diseases [35,36,37,38]. In our preclinical models, we identified several cytokines/chemokines secreted by mammary tumor lines derived from PyMT+ and Neu+ mice. Both cell lines secreted Gm-csf, Tnf-α, Ccl-2, Ifn-γ, and IL-1α while Timp1 was uniquely secreted by the PyMT+ tumor line. Neu+ mammary tumor line secreted G-csf, Ccl-1, Ccl-5, Cxcl-1, Cxcl-2, Cxcl-10, and Il-1ra. Recently, we measured 33 circulating cytokines/chemokines in plasma of PyMT+ mice by using the Milliplex kit and reported that Tnf-α, Tgf-α, β, G-csf, Il-6, and Gm-csf are elevated, whereas Mip-2 Cxcl-2, Il-9, Cxcl-5, and Il-1α are decreased in tumor-bearing mice compared to age and sex-matched control animals [13]. Thus, the cancer genome has an influence on the type of cytokines/chemokines secreted by cancer cells and consequently on the type and severity of functional limitations. In addition, Peake and colleagues [29] reported that skeletal muscle itself expresses a variety of cytokines, including IFN-γ, TNF-α, LIF, TGF-β, and IL-6. Thus, cytokines produced locally and delivered systemically act on muscles through their specific receptors on myogenic cells. Indeed, myogenic cells express receptors that interact with cytokines including TNF-α, IL-1, IL-6, IFN-γ, and G-CSF [28,39,40].
Many cytokines have been well studied for their actions on cancer-associated muscle wasting and functional limitations. TNF-α directly binds to type 1 TNF-α receptor (TNFR1) on muscle cells resulting in increased reactive oxygen species production via mitochondrial electron transport and activation of NF-κB signaling pathways. NF-κB increases the activity of the ubiquitin/proteasome pathway, which accelerates the regulated degradation of muscle proteins and hence causes muscle weakness [41]. Indeed, our recent report showed that increased levels of Tnf-α, Tgf-β, and G-csf in plasma of PyMT+ mice were accompanied by lower muscle mitochondrial activity and muscle dysfunction, which were restored upon inhibition of NF-κB signaling pathways [13]. Interestingly, TNF-α and several other cytokines have been shown to modulate peripheral insulin sensitivity with a mechanism that affects the activation of the insulin receptor and downstream signaling molecules in the skeletal muscles. In addition, TNF-α downregulated insulin-like growth factor-1-dependent signaling pathways that reduced muscle anabolic capacity and enhanced pro-catabolic stimuli [32]. In animal models, treatment with Tgf-β results in muscle atrophy and fibrosis, which causes reduced muscle contraction force [40,42]. In breast cancer with bone metastasis, Tgf-β causes muscle weakness and reduces muscle contractibility [27,43]. Similarly, TGF-β and TNF-α enhance the expression of zinc transporter ZIP14, which blocks the differentiation of muscle stem cells, also called satellite cells (MuSCs), and enhances the loss of myosin heavy chain in the skeletal muscles [44]. Taken together, breast cancer-associated alterations in circulating cytokines contribute to tumor-associated muscle dysfunction (Figure 1). Cancer-specific genomic aberrations may determine the type of cytokines/chemokines produced by cancer cells and pathways affected in the skeletal muscles.

4. Impaired Myogenesis Contributes to Functional Limitations in Breast Cancer

Healthy skeletal muscles have self-renewal capacity via MuSCs to replace injured or dead myofibers [45,46]. Generally, MuSCs are located in niche reservoirs between muscle fibers and extracellular matrix (ECM) and maintained in a quiescent state through activities of several factors including MuSC-enriched transcription factor PAX7 [47,48]. When muscles are injured, MuSCs are activated as myoblasts expressing both PAX7 and MyoD. Myoblasts have proliferating ability to generate as many cells as needed to repair injury. While a portion of myoblasts expressing PAX7, but lacking MyoD, return to quiescent state, the other major portion expressing MyoD, but lacking PAX7, will go on to differentiate and form multinucleated myofibers, and finally replace injured muscle fibers physically and functionally by expressing various contractile proteins, such as myosin heavy chain (MyHC) and skeletal muscle alpha actin [47,49]. Tumors have a pronounced impact on the number of regenerating myofibers in tumor-bearing mdx mice, a popular model to study DMD, compared to the number of regenerating myofibers in tumor-free mdx mice [50]. Skeletal muscle in PDX mouse cancer models also shows reduced regenerative capacity due to dysregulation of Pax7. Pax7 is of importance during myogenesis at any age as Pax7 deficiency in mice results in cell cycle arrest and precocious differentiation of MuSCs, which leads to impaired muscle regeneration [51,52]. As reported previously, Pax7 not only contributes to MuSC activation and self-renewal but also regulates MuSC proliferation and differentiation by acting on its target genes such as Myf5 and MyoD [53,54]. Indeed, MyoD and Myf5 along with other myogenic regulatory factors Myf6 and MyoG, determine the specification of myogenesis and influence MyHC levels and metabolic properties of myofibers [55]. In several of these models, through the P38 MAPK signaling pathway, TNF-α negatively regulates Pax7 expression and contributes to impaired muscle regeneration [56]. In the PyMT+ mammary tumor model, tumor-bearing mice contained lower skeletal muscle Pax7 as well as lower levels of MyoD along with the upregulation of circulating Tnf-α [13]. As illustrated schematically in Figure 1, tumor-derived cytokines/chemokines can impair specific steps of the myogenic program and contribute to functional limitations [13].
In animals that lack inducible nitric oxide synthase (Nos2) expression, MuSCs fail to proliferate and differentiate as a muscle damage-induced increase of Pax7+ MyoD+ double-positive cells is significantly delayed in iNOS-deficient mice along with a significant decrease of MyoG compared to wild type mice [57], indicating that Nos2 is required for effective regeneration of muscle. Nos2 is an enzyme that regulates the synthesis of nitric oxide (NO), which is involved in adult skeletal muscle homeostasis, particularly by mediating muscle regeneration after injury [58,59,60,61]. NO promotes activation, fusion, and maintenance of the pool of MuSCs in acutely and chronically damaged muscles [62]. Skeletal muscle Nos2 expression in PyMT+ mammary tumor mice compared to wild type mice was significantly lower [13], which may have an impact on recovery from injury.
Both BCL-2 and caspase-3 are key regulators of cell apoptosis [63,64]. BCL-2 prevents programmed cell death by inhibiting shuttle proteins BAX and BAK, as enhanced activity of BAX and BAK leads to caspase-3 activation that results in cell death [65,66]. In this regard, in human disease of DMD, enhanced expression of caspase-3 is responsible for myofiber cell death and muscle atrophy [67]. However, a complex role of caspase 3 in skeletal muscle homeostasis is emerging [68]. Caspase 3 null mice compared to wild type mice have reduced muscle mass and myoblasts due to defects in differentiation to myofibers with a parallel downregulation of MyoD and MyoG. These findings are further confirmed in the C2C12 cell line by pharmacologically inhibiting caspase-3 activity. A recent in vivo study showed that inhibition of caspase-3 activity results in a profound disruption in skeletal muscle regeneration [69]. Taken together, caspase-3 is required for MuSC differentiation. With respect to the role of Bcl-2 in myogenesis, Bcl-2+ C2C12 cells co-express markers of early stages of myogenesis, including desmin, MyoD, and Myf-5, and expression of Bcl-2 promotes clonal expansion as the muscle colonies produced by cloned Bcl-2–null cells contain only about half as many cells as the colonies produced by cells with Bcl-2 [70]. Since cytokines such as TNF can influence the expression and/or activity of Bcl-2 and caspase 3, circulating chemokine/cytokine-mediated changes in pro-apoptotic and anti-apoptotic proteins in skeletal muscle could have a profound impact on skeletal muscle function in cancer patients.
Skeletal muscle expresses many miRNAs [71,72,73,74,75,76] that regulate MuSC proliferation, differentiation, and even muscle force production by modulating gene expression via transcriptional, post-transcriptional, epigenetic mechanisms, and nuclear genome organization [71,77,78,79,80]. Cancer affecting the expression of muscle-specific miRNAs has been reported recently in breast cancer [38], rhabdomyosarcoma [81], pancreatic and colorectal cancer [82], gastric cancer [83], and lung cancer [84]. We reported that circulating levels of cardiac and skeletal muscle-enriched miR-486 were lower in breast cancer patients with metastasis compared to healthy individuals [38]. In animal models of breast cancer, lower circulating miR-486 levels correlate with reduced miR-486 expression in skeletal muscle. Furthermore, proteins in the myogenesis (Pax7/MyoD/MyoG) and myofiber survival (Dock3/Pten/Akt) networks, which are regulated by and/or regulate miR-486, are deregulated in skeletal muscles of tumor-bearing transgenic mice compared to controls [38]. In mammary tumor-bearing animals, the downregulation of miR-486 was correlated with the upregulation of circulating Tnf-α, a cytokine that suppresses the expression of miR-486 in myoblasts in culture potentially via NF-κB signaling pathways [13,38]. Thus, miR-486 is an integral part of the myogenesis signaling network that involves Pax7, MyoD, myostatin, and NF-κB [15,85,86,87] and a potential target of cancer-induced chemokines/cytokines. Reduced skeletal muscle miR-486 expression is a major defect in DMD and transgenic expression of miR-486 in muscle could rescue muscular dystrophy phenotype in animal models [88]. In this regard, based on our preclinical studies, we had previously proposed that loss of miR-486 is responsible for functional limitations in breast cancer patients.
miR-206 is another one of the widely studied miRNAs in skeletal muscles [89]. miR-206 promotes myoblast differentiation of C2C12 cells by blocking cell cycle progression and by inducing myofiber formation [90] via its upstream regulator MyoD and downstream targets Pax7, Pax3, and estrogen receptor-alpha [91]. During myoblast differentiation, MyoD activates miR-206 expression that, in turn, downregulates Pax7 and Pax3 [92,93,94]. TGF-β and myostatin, which are upregulated in cancer patients, downregulate miR-206 and MyoD levels in skeletal muscle. Skeletal muscles of myostatin knockout mice compared to wild type mice have significantly higher levels of miR-206, while Tgf-β treatment reduces the expression of miR-206 in C2C12 myoblasts [95,96]. Local injection of miR-206 into injured skeletal muscles in rats enhanced muscle regeneration both morphologically and physiologically and effectively inhibited muscle fibrosis [97]. These observations indicate the role of miR-206 in muscle regeneration and muscle function. Thus, it is not surprising that elevated circulating Tgf-β in a PyMT+ mammary tumor model correlated with lower miR-206 in muscles and reduced muscle function (Figure 1) [13]. In addition, decreased circulating miR-206 is observed in breast cancer [98], renal cell carcinoma [99], melanoma [100], and osteosarcoma [101] patients.

5. Energetic Inefficiency in Skeletal Muscle Contributes to Functional Limitations in Breast Cancer

Cachexia, myopenia, and functional limitations in cancer patients are generally associated with a negative energy balance resulting from either reduced energy production or increased energy expenditure during disease progression [102,103,104]. Advanced cancer patients have enhanced thermogenesis that increases energy expenditure, which is consistent with the observation in cachectic tumor-bearing animals [105]. It is also known that impairment of the mitochondrial compartment results in decreased ATP production, leading to an energy deficit that becomes even worse since it is coupled with steadily increased energy expenditure. Consistent with this possibility, we have shown reduced muscle mitochondrial content in a PyMT+ mammary tumor model [13], and others have shown reduced ATP levels and increased activity of the energy sensor AMPK [106,107], implying that breast cancer decreases energy production in skeletal muscles. Thus, it is not surprising that skeletal muscles of PyMT+ animals contained lower levels of Atp2a1 [13], an energy transfer enzyme that regulates cellular calcium required for muscle contraction and muscle force production [108]. Interestingly, miR-486 and miR-206 are localized in mitochondria [109], and it is known that miR-486 protects the membrane potential of mitochondria to maintain cell integrity [110,111] through anti-apoptotic BCL-2 family proteins [112,113]. Thus, reduced miR-486 in skeletal muscles of tumor-bearing mice may affect mitochondrial activity that regulates energy production. Collectively, current literature suggests impaired energy production in skeletal muscle as a consequence of cancer, which may be responsible for functional limitation (Figure 1).

6. Altered Extracellular Matrix Contributes to Functional Limitations in Breast Cancer

Extracellular matrix (ECM) consists of a variety of collagens, elastins, fibronectins, tenascins, proteoglycans, and glycosaminoglycans [114]. ECM has dynamic influence on myofibers and muscle function through ECM-directed remodeling events [115,116]. Metalloproteinases (MMPs) play an essential role in ECM remodeling. MMP-2 and MMP-9 degrade collagen type IV of the basement membranes, but also activate focal adhesion kinase that may lead to changes in integrin function and modifications of the cytoskeleton [117]. During early disease stage of DMD, MMP-9, which is elevated due to NF-kB activity, enhances myogenesis. However, during the later disease stage of DMD, MMP-9 level is decreased, leading to the accumulation of fibroadipose tissues and reduced muscle strength [118]. These findings indicate the dual actions of MMP-9 in ECM remodeling and muscle regeneration. We reported that skeletal muscle of mammary tumor-bearing mice compared to wild type mice had elevated ECM deposition, which could be due to a reduction of Mmp-9, [13]. The specific role of Mmp-9 in skeletal muscle remodeling is evident from transgenic and knockout mouse models. Overexpression of Mmp-9 results in myofiber hypertrophy with increased fiber size and contractile force [119]. On the other hand, knockout of the Mmp-9 gene causes muscle atrophy by decreasing fiber cross-sectional areas and altering myofiber type distribution in mouse hindlimb skeletal muscles [120]. Thus, enhanced ECM and decreased Mmp-9 in the skeletal muscle may directly or indirectly contribute to muscle-associated functional limitations (Figure 1). Although MMP-9 has pro-tumorigenic and pro-metastatic roles, MMP-9 targeted therapies failed in clinical studies, potentially due to its requirement for proper skeletal muscle function [121]. MMP members other than MMP-9 could also play a role in skeletal muscle remodeling as the expression of laminin A, a key component of basal membrane in muscle and an Mmp-11 target, which is downregulated in muscles [122]. How cancer causes changes in skeletal muscle ECM is unknown but TGF-β is the likely culprit as TGF-β is implicated in multiple fibrotic diseases [123,124,125]. TGF-β functions to promote ECM preservation by enhancing collagen synthesis, expression of ECM and profibrotic genes, and inhibiting ECM degradation [126,127] through reduced MMP activity [128,129,130]. Interestingly, MMP-9 is a positive regulator of TGF-β as MMP-9 is required for the release of active TGF-β [131,132]. Therefore, in cancer, it is likely that circulating TGF-β rather than locally produced TGF-β contributes to enhanced ECM deposition in skeletal muscle.

7. STAT3 Signaling Contributes to Muscle Wasting in Cancer Cachexia

The role of STAT3 in muscle function has been reviewed [133,134]. It is reported that STAT3 contributes to muscle wasting in PDX animal models, as constitutively active STAT3 induces muscle fiber atrophy and exacerbates wasting in cancer cachexia [135,136] in an NF-kB dependent manner [137]. Conversely, inhibition of STAT3 reduces muscle atrophy in cancer [135,136]. These observations are consistent with STAT3 signaling negatively regulating MuSC expansion and skeletal muscle regeneration. Pharmacological inhibition of Stat3 activity enhances MuSC proliferation and muscle repair after injury or during diseased conditions [138,139]. Interestingly, a recent study demonstrated that Stat3 promotes the progression of MuSCs to myogenic lineage through mitochondrial respiration [140], indicating the involvement of mitochondria in muscle repair or muscle wasting in disease conditions. Indeed, JAK2/STAT3 signaling improves cardiac dysfunction by normalizing mitochondrial respiratory function in vivo and in vitro [141] through regulating reactive oxygen species (ROS) formation [142]. Mitochondria are one of the most important sources of ROS in skeletal muscles [143]. The role of ROS and redox signaling in muscle function and muscle regeneration has been reviewed [143,144,145]. Mitochondrial STAT3 has also been implicated in modulating mitochondrial DNA, mitochondrial transcription, and electron transport chain activity [146,147,148,149,150], further influencing cell survival and metabolism [151,152].

8. NF-κB is a Critical Signaling Relay Engaged in Breast Cancer Associated Functional Limitations

NF-κB represses myogenesis [153,154] and enhances muscle atrophy in various skeletal muscle diseases [155,156,157,158]. Activation of NF-κB in response to TNF-α blocks muscle differentiation by enhancing the degradation of the skeletal muscle-specific transcription factor MyoD [154,159]. Apart from TNF-α, increased TGF-β levels in circulation of breast cancer patients [160] can activate TAK1 [161,162], which phosphorylates IKK2 as well as NIK [163,164] and inhibits myogenesis by repressing the expression of MyoD [75,165].
It is noted that the exact role of the NF-κB signaling pathway depends on the state of the skeletal muscle disease and the nature of the disease. For instance, normal skeletal muscles have lower NF-κB activity with different transcriptional profiles compared to stressed muscles. During the late-stage of muscle differentiation, NF-κB acts as a negative regulator of differentiation through the transcriptional repressor YinYang1 [166]. During skeletal muscle atrophy, NF-κB induces murine ring finger-1 [155], an E3 ubiquitin ligase known to be involved in multiple models of skeletal muscle atrophy [167]. NF-κB represses MyoD by inducing its negative regulator PAX7 [50]. Recent studies have demonstrated that myostatin, another protein induced by NF-κB, represses miR-486 expression and inhibitors of this molecule can overcome aging-associated sarcopenia [87,168]. By contrast, miR-486 increases MyoD and its own expression by repressing Pax7 [169]. Furthermore, miR-486 itself controls NF-κB [86]. Mmp-9 regulates NF-κB activity, which further modulates Mmp-9 expression in skeletal muscles. NF-κB mediated induction of nitric oxide has been reported recently [170]. Therefore, there is an intricate signaling network with feed-forward and feedback loops comprising of NF-κB, PAX7, MyoD, myostatin, miR-486, and MMP-9 that regulate skeletal muscle homeostasis, which is disrupted in cancer (Figure 1).
In preclinical studies, activation of NF-κB within muscles results in severe muscle atrophy in mice [50,155]. Inhibition of NF-κB activity preserves muscle mass and reduces oxidative stress in transgenic and PDX tumor-bearing mice [171,172]. In response to denervation-associated muscle degeneration, IKK2 depletion prevents muscle atrophy, maintains fiber type, size, and strength, increases protein synthesis, and reduces protein degradation [156]. Treatment of tumor-bearing animals with a non-specific NF-κB inhibitor prevents tumor-associated muscle wasting via increased MyoD expression and inhibition of ubiquitin-conjugating enzyme E214K in skeletal muscles [173]. Thus, NF-κB and its associated signaling pathways are potential therapeutic targets to treat cancer-related muscle dysfunction (Figure 1). Indeed, we reported that the NF-κB inhibitor, dimethylaminoparthenolide (DMAPT), restored skeletal muscle functions including grip strength and motion performance in mammary tumor-bearing mice, accompanied by amelioration of few of the tumor-associated skeletal muscle molecular defects [13].

9. Perspective and Conclusions

Although there has been recent progress in understanding functional limitations in breast cancer, many areas may need to be further explored. The limited progress is mainly due to the perception that breast cancer patients, compared to other cancer patients, rarely experience cachexia. However, considering one in eight women in the United States is diagnosed with breast cancer and 25% of breast cancer patients, particularly those with triple-negative breast cancer, experience cachexia [2,174], additional studies are needed. Furthermore, myopenic obesity, which not only provides a misguided outlook but also impacts chemotherapy tolerability [26,175], is common in breast cancer patients. Lean muscle mass instead of BMI may need to be measured in breast cancer patients to ensure that there is no muscle loss. Mechanistic insights into myopenic obesity are needed to develop therapeutic intervention. In certain cases, adipose atrophy is also a concern as chemokines/cytokines that cause skeletal muscle dysfunction in cancer also mediate adipose atrophy [176]. Healthy interaction between skeletal muscle and adipocytes is essential as adipokines from adipocytes have a direct influence on myocytes, particularly with respect to metabolic pathways. Cancer-associated changes in neuromuscular and bone-skeletal muscle interaction are gaining attention. Recently, Baraldo and her colleagues reported that permanent deletion of the mTORC1 gene results in instability in the neuromuscular junction, which correlates with reduced treadmill performance [177]. Thus, it might be worthwhile to examine neuromuscular junction structure integrity and stability in skeletal muscles of breast cancer patients or animals with mammary tumors, as innervated skeletal muscle by tissue constructs has higher contractile forces compared to non-innervated skeletal muscle [178]. Gut microbiome is another upcoming area of research and it has recently been shown to affect skeletal muscle size, composition, and function via a proposed gut–muscle axis [179,180]. Since cancer-induced cytokines/chemokines as well as cancer treatments can affect the gut microbiome, it will be interesting to determine an association between the gut microbiome and muscle dysfunction in breast cancer patients. Consistent with this possibility, animals that lack a microbiota in gut have reduced muscle weight, reduced transcription of genes in mitochondria, and reduced neurotransmitter for neuromuscular junction [181]. Since breast cancer is not a single disease and broadly classified into five subtypes [182,183], how each subtype affects skeletal muscle function is unknown. Whether molecular defects are different among subtypes and disease stages of breast cancer remains unknown due to scarce clinical information and limited translational knowledge. Therefore, clinical management of cancer-associated functional limitations remains a great challenge for improving the life quality of cancer patients. In this regard, the majority of the anti-cachexia drugs tested in clinical trials so far have proven ineffective in improving muscle functional performance although a few of them had effects in increasing body weight and lean body mass [184,185].
Overall, our review outlines recent advances in breast cancer-induced functional limitations, highlighting the role of circulating cytokines/chemokines, microRNAs, mitochondria, ECM, and impaired myogenesis, as depicted in Figure 1. Molecular defects outlined here, as well as advances being made in the field of skeletal muscle biology, offer an opportunity to develop pharmacological interventions for the treatment of functional limitations in breast cancer in the near future.

Funding

This work is supported by Department of Veterans Affairs grant BX002764 to HN.

Conflicts of Interest

All authors declare no conflict of interest.

Abbreviations

AKTThymoma viral proto-oncogene, also as known protein kinase B (PKB)
AMPKAMP-activated protein kinase
BAK Bcl-2 homologous antagonist killer
BAXBcl-2-associated X protein
BCL-2 B-cell lymphoma 2
CCL1, 2, or 5 C-C motif chemokine ligand 1, 2, or 5
CCR3 C-C chemokine receptor type 3
CM Conditioned media
COX-IV Cytochrome c oxidase subunit 4 isoform 1
CXCL1, 2, 5 or 10 C-X-C motif chemokine ligand 1, 2, 5, or 10
DMAPT Dimethylaminoparthenolide
DMDDuchenne muscular dystrophy
DOCK3 Dedicator of cytokinesis 3
ECM Extracellular matrix
G-CSF Granulocyte-colony stimulating factor
GM-CSFGranulocyte-macrophage colony stimulating factor
GTex, Genotype-tissue expression
HOXA9Homeobox A9
IFN-α, β or γInterferon alpha, beta or gamma
IKK Inhibitor of kappaB kinase
IL-1, 2, 4, 6 or 9 Interleukin 1, 2, 4, 6 or 9
IL-1RAInterleukin 1 receptor antagonist
MAPK Mitogen-activated protein kinase
MMPsMatrix metalloproteinases
MMTV Mouse mammary tumor virus
MIP2 Macrophage inflammatory protein 2
mTORC1Mammalian target of rapamycin complex 1
MuSC Muscle stem cell
MYF5 or 6 Myogenic factor 5 or 6
MyHC Myosin heavy chain
MyoD Myoblast determination protein 1
MyoGMyogenin
NF-κB Nuclear factor-kappa B
NIK NF-kappa B-inducing kinase
NO Nitric oxide
Nos2 Nitric oxide synthase
Pax3Paired box 3
Pax7 Paired box 7
PDX Patient-derived xenograft
PGC-1β Peroxisome proliferator-activated receptor gamma co-activator 1 beta
PTEN Phosphatase and tensin homolog
PyMT Polyoma middle tumor antigen
ROSReactive oxygen species
STAT3Signal transducer and activator of transcription 3
sTNFRII Soluble tumor necrosis factor receptor 2
TAK1 Transforming growth factor beta activated kinase 1
TGF-βTransforming growth factor beta
TIMP1 TIMP metallopeptidase inhibitor 1
TNF Tumor necrosis factor
TNFR1TNF-α receptor type 1
ZIP14 ZRT/IRT-like protein 14

References

  1. Consul, N.; Guo, X.; Coker, C.; Lopez-Pintado, S.; Hibshoosh, H.; Zhao, B.; Kalinsky, K.; Acharyya, S. Monitoring metastasis and cachexia in a patient with breast cancer: A case study. Clin. Med. Insights Oncol. 2016, 10, 83–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Tomasin, R.; Martin, A.; Cominetti, M.R. Metastasis and cachexia: Alongside in clinics, but not so in animal models. J. Cachexia Sarcopenia Muscle 2019, 10, 1183–1194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Hendren, E.; Vinik, O.; Faragalla, H.; Haq, R. Breast cancer and dermatomyositis: A case study and literature review. Curr. Oncol. 2017, 24, e429–e433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Khoo, H.Y.; Tan, W.J.; Cheong, Y.T. Breast cancer with dermatomyositis as initial presentation. Med. J. Malays. 2018, 73, 44–45. [Google Scholar]
  5. Braithwaite, D.; Satariano, W.A.; Sternfeld, B.; Hiatt, R.A.; Ganz, P.A.; Kerlikowske, K.; Moore, D.H.; Slattery, M.L.; Tammemagi, M.; Castillo, A.; et al. Long-term prognostic role of functional limitations among women with breast cancer. J. Natl. Cancer Inst. 2010, 102, 1468–1477. [Google Scholar] [CrossRef]
  6. Baltgalvis, K.A.; Berger, F.G.; Pena, M.M.; Mark Davis, J.; White, J.P.; Carson, J.A. Activity level, apoptosis, and development of cachexia in apc(min/+) mice. J. Appl. Physiol. 2010, 109, 1155–1161. [Google Scholar] [CrossRef] [Green Version]
  7. Murphy, K.T.; Chee, A.; Trieu, J.; Naim, T.; Lynch, G.S. Importance of functional and metabolic impairments in the characterization of the c-26 murine model of cancer cachexia. Dis. Model Mech. 2012, 5, 533–545. [Google Scholar] [CrossRef] [Green Version]
  8. Puppa, M.J.; Gao, S.; Narsale, A.A.; Carson, J.A. Skeletal muscle glycoprotein 130’s role in lewis lung carcinoma-induced cachexia. FASEB J. 2014, 28, 998–1009. [Google Scholar] [CrossRef] [Green Version]
  9. Roberts, B.M.; Frye, G.S.; Ahn, B.; Ferreira, L.F.; Judge, A.R. Cancer cachexia decreases specific force and accelerates fatigue in limb muscle. Biochem. Biophys. Res. Commun. 2013, 435, 488–492. [Google Scholar] [CrossRef] [Green Version]
  10. Toth, M.J.; Callahan, D.M.; Miller, M.S.; Tourville, T.W.; Hackett, S.B.; Couch, M.E.; Dittus, K. Skeletal muscle fiber size and fiber type distribution in human cancer: Effects of weight loss and relationship to physical function. Clin. Nutr. 2016, 35, 1359–1365. [Google Scholar] [CrossRef]
  11. Sweeney, C.; Schmitz, K.H.; Lazovich, D.; Virnig, B.A.; Wallace, R.B.; Folsom, A.R. Functional limitations in elderly female cancer survivors. J. Natl. Cancer Inst. 2006, 98, 521–529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Arnold, M.; Jiang, L.; Stefanick, M.L.; Johnson, K.C.; Lane, D.S.; LeBlanc, E.S.; Prentice, R.; Rohan, T.E.; Snively, B.M.; Vitolins, M.; et al. Duration of adulthood overweight, obesity, and cancer risk in the women’s health initiative: A longitudinal study from the united states. PLoS Med. 2016, 13, e1002081. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, R.; Bhat-Nakshatri, P.; Padua, M.B.; Prasad, M.S.; Anjanappa, M.; Jacobson, M.; Finnearty, C.; Sefcsik, V.; McElyea, K.; Redmond, R.; et al. Pharmacological dual inhibition of tumor and tumor-induced functional limitations in transgenic model of breast cancer. Mol. Cancer Ther. 2017, 16, 2747–2758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Janssen, I.; Heymsfield, S.B.; Wang, Z.M.; Ross, R. Skeletal muscle mass and distribution in 468 men and women aged 18-88 yr. J. Appl. Physiol. 2000, 89, 81–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Small, E.M.; O’Rourke, J.R.; Moresi, V.; Sutherland, L.B.; McAnally, J.; Gerard, R.D.; Richardson, J.A.; Olson, E.N. Regulation of pi3-kinase/akt signaling by muscle-enriched microrna-486. Proc. Natl. Acad. Sci. USA 2010, 107, 4218–4223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Frontera, W.R.; Ochala, J. Skeletal muscle: A brief review of structure and function. Calcif. Tissue Int. 2015, 96, 183–195. [Google Scholar] [CrossRef]
  17. Rowland, L.A.; Bal, N.C.; Periasamy, M. The role of skeletal-muscle-based thermogenic mechanisms in vertebrate endothermy. Biol. Rev. Camb. Philos. Soc. 2015, 90, 1279–1297. [Google Scholar] [CrossRef] [Green Version]
  18. Al-Majid, S.; McCarthy, D.O. Cancer-induced fatigue and skeletal muscle wasting: The role of exercise. Biol. Res. Nurs. 2001, 2, 186–197. [Google Scholar] [CrossRef]
  19. Barreiro, E.; Gea, J. Respiratory and limb muscle dysfunction in copd. COPD 2015, 12, 413–426. [Google Scholar] [CrossRef]
  20. Siegel, I.M. Update on duchenne muscular dystrophy. Compr. Ther. 1989, 15, 45–52. [Google Scholar]
  21. Winningham, M.L.; Nail, L.M.; Burke, M.B.; Brophy, L.; Cimprich, B.; Jones, L.S.; Pickard-Holley, S.; Rhodes, V.; St Pierre, B.; Beck, S.; et al. Fatigue and the cancer experience: The state of the knowledge. Oncol. Nurs. Forum 1994, 21, 23–36. [Google Scholar] [PubMed]
  22. Wolfe, R.R. The underappreciated role of muscle in health and disease. Am. J. Clin. Nutr. 2006, 84, 475–482. [Google Scholar] [CrossRef] [PubMed]
  23. Guigni, B.A.; Callahan, D.M.; Tourville, T.W.; Miller, M.S.; Fiske, B.; Voigt, T.; Korwin-Mihavics, B.; Anathy, V.; Dittus, K.; Toth, M.J. Skeletal muscle atrophy and dysfunction in breast cancer patients: Role for chemotherapy-derived oxidant stress. Am. J. Physiol. Cell Physiol. 2018, 315, C744–C756. [Google Scholar] [CrossRef] [PubMed]
  24. Caan, B.J.; Cespedes Feliciano, E.M.; Prado, C.M.; Alexeeff, S.; Kroenke, C.H.; Bradshaw, P.; Quesenberry, C.P.; Weltzien, E.K.; Castillo, A.L.; Olobatuyi, T.A.; et al. Association of muscle and adiposity measured by computed tomography with survival in patients with nonmetastatic breast cancer. JAMA Oncol. 2018, 4, 798–804. [Google Scholar] [CrossRef]
  25. Shiroyama, T.; Nagatomo, I.; Koyama, S.; Hirata, H.; Nishida, S.; Miyake, K.; Fukushima, K.; Shirai, Y.; Mitsui, Y.; Takata, S.; et al. Impact of sarcopenia in patients with advanced non-small cell lung cancer treated with pd-1 inhibitors: A preliminary retrospective study. Sci. Rep. 2019, 9, 2447. [Google Scholar] [CrossRef] [Green Version]
  26. Cespedes Feliciano, E.; Chen, W.Y. Clinical implications of low skeletal muscle mass in early-stage breast and colorectal cancer. Proc. Nutr. Soc. 2018, 77, 382–387. [Google Scholar] [CrossRef]
  27. Waning, D.L.; Mohammad, K.S.; Reiken, S.; Xie, W.; Andersson, D.C.; John, S.; Chiechi, A.; Wright, L.E.; Umanskaya, A.; Niewolna, M.; et al. Excess tgf-beta mediates muscle weakness associated with bone metastases in mice. Nat. Med. 2015, 21, 1262–1271. [Google Scholar] [CrossRef]
  28. Zhang, Y.; Pilon, G.; Marette, A.; Baracos, V.E. Cytokines and endotoxin induce cytokine receptors in skeletal muscle. Am. J. Physiol. Endocrinol. Metab. 2000, 279, E196–E205. [Google Scholar] [CrossRef]
  29. Peake, J.M.; Della Gatta, P.; Suzuki, K.; Nieman, D.C. Cytokine expression and secretion by skeletal muscle cells: Regulatory mechanisms and exercise effects. Exerc. Immunol. Rev. 2015, 21, 8–25. [Google Scholar]
  30. Argiles, J.M.; Busquets, S.; Lopez-Soriano, F.J. Cytokines in the pathogenesis of cancer cachexia. Curr. Opin. Clin. Nutr. Metab. Care 2003, 6, 401–406. [Google Scholar] [CrossRef]
  31. Fearon, K.; Arends, J.; Baracos, V. Understanding the mechanisms and treatment options in cancer cachexia. Nat. Rev. Clin. Oncol. 2013, 10, 90–99. [Google Scholar] [CrossRef] [PubMed]
  32. Tisdale, M.J. Cancer cachexia. Curr. Opin. Gastroenterol. 2010, 26, 146–151. [Google Scholar] [CrossRef] [PubMed]
  33. Hogan, K.A.; Cho, D.S.; Arneson, P.C.; Samani, A.; Palines, P.; Yang, Y.; Doles, J.D. Tumor-derived cytokines impair myogenesis and alter the skeletal muscle immune microenvironment. Cytokine 2018, 107, 9–17. [Google Scholar] [CrossRef] [PubMed]
  34. Bower, J.E.; Ganz, P.A.; Aziz, N.; Fahey, J.L. Fatigue and proinflammatory cytokine activity in breast cancer survivors. Psychosom. Med. 2002, 64, 604–611. [Google Scholar] [CrossRef] [Green Version]
  35. Arend, W.P.; Malyak, M.; Guthridge, C.J.; Gabay, C. Interleukin-1 receptor antagonist: Role in biology. Annu. Rev. Immunol. 1998, 16, 27–55. [Google Scholar] [CrossRef]
  36. Diez-Ruiz, A.; Tilz, G.P.; Zangerle, R.; Baier-Bitterlich, G.; Wachter, H.; Fuchs, D. Soluble receptors for tumour necrosis factor in clinical laboratory diagnosis. Eur. J. Haematol. 1995, 54, 1–8. [Google Scholar] [CrossRef]
  37. Fuchs, D.; Hausen, A.; Reibnegger, G.; Werner, E.R.; Dierich, M.P.; Wachter, H. Neopterin as a marker for activated cell-mediated immunity: Application in hiv infection. Immunol. Today 1988, 9, 150–155. [Google Scholar] [CrossRef]
  38. Chen, D.; Goswami, C.P.; Burnett, R.M.; Anjanappa, M.; Bhat-Nakshatri, P.; Muller, W.; Nakshatri, H. Cancer affects microrna expression, release, and function in cardiac and skeletal muscle. Cancer Res. 2014, 74, 4270–4281. [Google Scholar] [CrossRef] [Green Version]
  39. Hara, M.; Yuasa, S.; Shimoji, K.; Onizuka, T.; Hayashiji, N.; Ohno, Y.; Arai, T.; Hattori, F.; Kaneda, R.; Kimura, K.; et al. G-csf influences mouse skeletal muscle development and regeneration by stimulating myoblast proliferation. J. Exp. Med. 2011, 208, 715–727. [Google Scholar] [CrossRef] [Green Version]
  40. Ismaeel, A.; Kim, J.S.; Kirk, J.S.; Smith, R.S.; Bohannon, W.T.; Koutakis, P. Role of transforming growth factor-beta in skeletal muscle fibrosis: A review. Int. J. Mol. Sci. 2019, 20, 2446. [Google Scholar] [CrossRef] [Green Version]
  41. Reid, M.B.; Li, Y.P. Tumor necrosis factor-alpha and muscle wasting: A cellular perspective. Respir. Res. 2001, 2, 269–272. [Google Scholar] [CrossRef] [PubMed]
  42. Mendias, C.L.; Gumucio, J.P.; Davis, M.E.; Bromley, C.W.; Davis, C.S.; Brooks, S.V. Transforming growth factor-beta induces skeletal muscle atrophy and fibrosis through the induction of atrogin-1 and scleraxis. Muscle Nerve 2012, 45, 55–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Regan, J.N.; Trivedi, T.; Guise, T.A.; Waning, D.L. The role of tgfbeta in bone-muscle crosstalk. Curr. Osteoporos Rep. 2017, 15, 18–23. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, G.; Biswas, A.K.; Ma, W.; Kandpal, M.; Coker, C.; Grandgenett, P.M.; Hollingsworth, M.A.; Jain, R.; Tanji, K.; Lomicronpez-Pintado, S.; et al. Metastatic cancers promote cachexia through zip14 upregulation in skeletal muscle. Nat. Med. 2018, 24, 770–781. [Google Scholar] [CrossRef] [PubMed]
  45. Feige, P.; Brun, C.E.; Ritso, M.; Rudnicki, M.A. Orienting muscle stem cells for regeneration in homeostasis, aging, and disease. Cell Stem Cell 2018, 23, 653–664. [Google Scholar] [CrossRef] [PubMed]
  46. Rayagiri, S.S.; Ranaldi, D.; Raven, A.; Mohamad Azhar, N.I.F.; Lefebvre, O.; Zammit, P.S.; Borycki, A.G. Basal lamina remodeling at the skeletal muscle stem cell niche mediates stem cell self-renewal. Nat. Commun. 2018, 9, 1075. [Google Scholar] [CrossRef] [Green Version]
  47. Dumont, N.A.; Wang, Y.X.; Rudnicki, M.A. Intrinsic and extrinsic mechanisms regulating satellite cell function. Development 2015, 142, 1572–1581. [Google Scholar] [CrossRef] [Green Version]
  48. Dueweke, J.J.; Awan, T.M.; Mendias, C.L. Regeneration of skeletal muscle after eccentric injury. J. Sport Rehabil. 2017, 26, 171–179. [Google Scholar] [CrossRef] [Green Version]
  49. Zammit, P.S.; Relaix, F.; Nagata, Y.; Ruiz, A.P.; Collins, C.A.; Partridge, T.A.; Beauchamp, J.R. Pax7 and myogenic progression in skeletal muscle satellite cells. J. Cell Sci. 2006, 119, 1824–1832. [Google Scholar] [CrossRef] [Green Version]
  50. He, W.A.; Berardi, E.; Cardillo, V.M.; Acharyya, S.; Aulino, P.; Thomas-Ahner, J.; Wang, J.; Bloomston, M.; Muscarella, P.; Nau, P.; et al. Nf-kappab-mediated pax7 dysregulation in the muscle microenvironment promotes cancer cachexia. J. Clin. Investig. 2013, 123, 4821–4835. [Google Scholar] [CrossRef] [Green Version]
  51. Von Maltzahn, J.; Jones, A.E.; Parks, R.J.; Rudnicki, M.A. Pax7 is critical for the normal function of satellite cells in adult skeletal muscle. Proc. Natl. Acad. Sci. USA 2013, 110, 16474–16479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Seale, P.; Sabourin, L.A.; Girgis-Gabardo, A.; Mansouri, A.; Gruss, P.; Rudnicki, M.A. Pax7 is required for the specification of myogenic satellite cells. Cell 2000, 102, 777–786. [Google Scholar] [CrossRef] [Green Version]
  53. Bajard, L.; Relaix, F.; Lagha, M.; Rocancourt, D.; Daubas, P.; Buckingham, M.E. A novel genetic hierarchy functions during hypaxial myogenesis: Pax3 directly activates myf5 in muscle progenitor cells in the limb. Genes Dev. 2006, 20, 2450–2464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. McKinnell, I.W.; Ishibashi, J.; Le Grand, F.; Punch, V.G.; Addicks, G.C.; Greenblatt, J.F.; Dilworth, F.J.; Rudnicki, M.A. Pax7 activates myogenic genes by recruitment of a histone methyltransferase complex. Nat. Cell. Biol. 2008, 10, 77–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Zammit, P.S. Function of the myogenic regulatory factors myf5, myod, myogenin and mrf4 in skeletal muscle, satellite cells and regenerative myogenesis. Semin. Cell Dev. Biol. 2017, 72, 19–32. [Google Scholar] [CrossRef] [PubMed]
  56. Palacios, D.; Mozzetta, C.; Consalvi, S.; Caretti, G.; Saccone, V.; Proserpio, V.; Marquez, V.E.; Valente, S.; Mai, A.; Forcales, S.V.; et al. Tnf/p38alpha/polycomb signaling to pax7 locus in satellite cells links inflammation to the epigenetic control of muscle regeneration. Cell Stem Cell 2010, 7, 455–469. [Google Scholar] [CrossRef] [Green Version]
  57. Rigamonti, E.; Touvier, T.; Clementi, E.; Manfredi, A.A.; Brunelli, S.; Rovere-Querini, P. Requirement of inducible nitric oxide synthase for skeletal muscle regeneration after acute damage. J. Immunol. 2013, 190, 1767–1777. [Google Scholar] [CrossRef] [Green Version]
  58. De Palma, C.; Clementi, E. Nitric oxide in myogenesis and therapeutic muscle repair. Mol. Neurobiol. 2012, 46, 682–692. [Google Scholar] [CrossRef] [Green Version]
  59. Filippin, L.I.; Moreira, A.J.; Marroni, N.P.; Xavier, R.M. Nitric oxide and repair of skeletal muscle injury. Nitric Oxide 2009, 21, 157–163. [Google Scholar] [CrossRef]
  60. Stamler, J.S.; Meissner, G. Physiology of nitric oxide in skeletal muscle. Physiol. Rev. 2001, 81, 209–237. [Google Scholar] [CrossRef]
  61. Nisoli, E.; Clementi, E.; Carruba, M.O.; Moncada, S. Defective mitochondrial biogenesis: A hallmark of the high cardiovascular risk in the metabolic syndrome? Circ. Res. 2007, 100, 795–806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Buono, R.; Vantaggiato, C.; Pisa, V.; Azzoni, E.; Bassi, M.T.; Brunelli, S.; Sciorati, C.; Clementi, E. Nitric oxide sustains long-term skeletal muscle regeneration by regulating fate of satellite cells via signaling pathways requiring vangl2 and cyclic gmp. Stem Cells 2012, 30, 197–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Porter, A.G.; Janicke, R.U. Emerging roles of caspase-3 in apoptosis. Cell Death Differ. 1999, 6, 99–104. [Google Scholar] [CrossRef] [PubMed]
  64. Elmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef] [PubMed]
  65. Tsujimoto, Y. Role of bcl-2 family proteins in apoptosis: Apoptosomes or mitochondria? Genes Cells 1998, 3, 697–707. [Google Scholar] [CrossRef] [PubMed]
  66. Edlich, F. Bcl-2 proteins and apoptosis: Recent insights and unknowns. Biochem. Biophys. Res. Commun. 2018, 500, 26–34. [Google Scholar] [CrossRef] [PubMed]
  67. Sandri, M.; El Meslemani, A.H.; Sandri, C.; Schjerling, P.; Vissing, K.; Andersen, J.L.; Rossini, K.; Carraro, U.; Angelini, C. Caspase 3 expression correlates with skeletal muscle apoptosis in duchenne and facioscapulo human muscular dystrophy. A potential target for pharmacological treatment? J. Neuropathol. Exp. Neurol. 2001, 60, 302–312. [Google Scholar] [CrossRef] [Green Version]
  68. Fernando, P.; Kelly, J.F.; Balazsi, K.; Slack, R.S.; Megeney, L.A. Caspase 3 activity is required for skeletal muscle differentiation. Proc. Natl. Acad. Sci. USA 2002, 99, 11025–11030. [Google Scholar] [CrossRef] [Green Version]
  69. Arnon-Friedman, R.; Dupuis, F.; Fawzi, O.; Renner, R.; Vidick, T. Practical device-independent quantum cryptography via entropy accumulation. Nat. Commun. 2018, 9, 459. [Google Scholar] [CrossRef] [Green Version]
  70. Dominov, J.A.; Dunn, J.J.; Miller, J.B. Bcl-2 expression identifies an early stage of myogenesis and promotes clonal expansion of muscle cells. J. Cell Biol. 1998, 142, 537–544. [Google Scholar] [CrossRef] [Green Version]
  71. Sohi, G.; Dilworth, F.J. Noncoding rnas as epigenetic mediators of skeletal muscle regeneration. FEBS J. 2015, 282, 1630–1646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Ahmad, A.; Negri, I.; Oliveira, W.; Brown, C.; Asiimwe, P.; Sammons, B.; Horak, M.; Jiang, C.; Carson, D. Transportable data from non-target arthropod field studies for the environmental risk assessment of genetically modified maize expressing an insecticidal double-stranded rna. Transgenic Res. 2016, 25, 1–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Simionescu-Bankston, A.; Kumar, A. Noncoding rnas in the regulation of skeletal muscle biology in health and disease. J. Mol. Med. 2016, 94, 853–866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Kusakabe, R.; Inoue, K. Developmental regulation and evolution of muscle-specific micrornas. Semin. Cell Dev. Biol. 2015, 47–48, 9–16. [Google Scholar] [CrossRef] [PubMed]
  75. Liu, D.; Black, B.L.; Derynck, R. Tgf-beta inhibits muscle differentiation through functional repression of myogenic transcription factors by smad3. Genes Dev. 2001, 15, 2950–2966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Ha, M.; Kim, V.N. Regulation of microrna biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef] [PubMed]
  77. Catalanotto, C.; Cogoni, C.; Zardo, G. Microrna in control of gene expression: An overview of nuclear functions. Int. J. Mol. Sci. 2016, 17, 1712. [Google Scholar] [CrossRef] [Green Version]
  78. Cai, L.; Chang, H.; Fang, Y.; Li, G. A comprehensive characterization of the function of lincrnas in transcriptional regulation through long-range chromatin interactions. Sci. Rep. 2016, 6, 36572. [Google Scholar] [CrossRef]
  79. Goncalves, T.J.M.; Armand, A.S. Non-coding rnas in skeletal muscle regeneration. Noncoding RNA Res. 2017, 2, 56–67. [Google Scholar] [CrossRef]
  80. D’Souza, R.F.; Bjornsen, T.; Zeng, N.; Aasen, K.M.M.; Raastad, T.; Cameron-Smith, D.; Mitchell, C.J. Micrornas in muscle: Characterizing the powerlifter phenotype. Front. Physiol. 2017, 8, 383. [Google Scholar] [CrossRef] [Green Version]
  81. Li, L.; Sarver, A.L.; Alamgir, S.; Subramanian, S. Downregulation of micrornas mir-1, -206 and -29 stabilizes pax3 and ccnd2 expression in rhabdomyosarcoma. Lab. Investig. 2012, 92, 571–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Narasimhan, A.; Ghosh, S.; Stretch, C.; Greiner, R.; Bathe, O.F.; Baracos, V.; Damaraju, S. Small rnaome profiling from human skeletal muscle: Novel mirnas and their targets associated with cancer cachexia. J. Cachexia Sarcopenia Muscle 2017, 8, 405–416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Yang, Q.; Zhang, C.; Huang, B.; Li, H.; Zhang, R.; Huang, Y.; Wang, J. Downregulation of microrna-206 is a potent prognostic marker for patients with gastric cancer. Eur. J. Gastroenterol. Hepatol. 2013, 25, 953–957. [Google Scholar] [CrossRef] [PubMed]
  84. Lee, D.E.; Brown, J.L.; Rosa-Caldwell, M.E.; Blackwell, T.A.; Perry, R.A., Jr.; Brown, L.A.; Khatri, B.; Seo, D.; Bottje, W.G.; Washington, T.A.; et al. Cancer cachexia-induced muscle atrophy: Evidence for alterations in micrornas important for muscle size. Physiol. Genom. 2017, 49, 253–260. [Google Scholar] [CrossRef]
  85. Buckingham, M.; Rigby, P.W. Gene regulatory networks and transcriptional mechanisms that control myogenesis. Dev. Cell 2014, 28, 225–238. [Google Scholar] [CrossRef] [Green Version]
  86. Song, L.; Lin, C.; Gong, H.; Wang, C.; Liu, L.; Wu, J.; Tao, S.; Hu, B.; Cheng, S.Y.; Li, M.; et al. Mir-486 sustains nf-kappab activity by disrupting multiple nf-kappab-negative feedback loops. Cell Res. 2013, 23, 274–289. [Google Scholar] [CrossRef]
  87. Hitachi, K.; Nakatani, M.; Tsuchida, K. Myostatin signaling regulates akt activity via the regulation of mir-486 expression. Int. J. Biochem. Cell Biol. 2014, 47, 93–103. [Google Scholar] [CrossRef]
  88. Alexander, M.S.; Casar, J.C.; Motohashi, N.; Vieira, N.M.; Eisenberg, I.; Marshall, J.L.; Gasperini, M.J.; Lek, A.; Myers, J.A.; Estrella, E.A.; et al. Microrna-486-dependent modulation of dock3/pten/akt signaling pathways improves muscular dystrophy-associated symptoms. J. Clin. Investig. 2014, 124, 2651–2667. [Google Scholar] [CrossRef]
  89. Ma, Y.; Ren, Y.; Dai, Z.J.; Wu, C.J.; Ji, Y.H.; Xu, J. Il-6, il-8 and tnf-alpha levels correlate with disease stage in breast cancer patients. Adv. Clin. Exp. Med. 2017, 26, 421–426. [Google Scholar] [CrossRef] [Green Version]
  90. Kim, H.K.; Lee, Y.S.; Sivaprasad, U.; Malhotra, A.; Dutta, A. Muscle-specific microrna mir-206 promotes muscle differentiation. J. Cell Biol. 2006, 174, 677–687. [Google Scholar] [CrossRef]
  91. Adams, B.D.; Cowee, D.M.; White, B.A. The role of mir-206 in the epidermal growth factor (egf) induced repression of estrogen receptor-alpha (eralpha) signaling and a luminal phenotype in mcf-7 breast cancer cells. Mol. Endocrinol. 2009, 23, 1215–1230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Chen, J.F.; Tao, Y.; Li, J.; Deng, Z.; Yan, Z.; Xiao, X.; Wang, D.Z. Microrna-1 and microrna-206 regulate skeletal muscle satellite cell proliferation and differentiation by repressing pax7. J. Cell Biol. 2010, 190, 867–879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Hirai, H.; Verma, M.; Watanabe, S.; Tastad, C.; Asakura, Y.; Asakura, A. Myod regulates apoptosis of myoblasts through microrna-mediated down-regulation of pax3. J. Cell Biol. 2010, 191, 347–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Rosenberg, M.I.; Georges, S.A.; Asawachaicharn, A.; Analau, E.; Tapscott, S.J. Myod inhibits fstl1 and utrn expression by inducing transcription of mir-206. J. Cell Biol. 2006, 175, 77–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Winbanks, C.E.; Wang, B.; Beyer, C.; Koh, P.; White, L.; Kantharidis, P.; Gregorevic, P. Tgf-beta regulates mir-206 and mir-29 to control myogenic differentiation through regulation of hdac4. J. Biol. Chem. 2011, 286, 13805–13814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Rachagani, S.; Cheng, Y.; Reecy, J.M. Myostatin genotype regulates muscle-specific mirna expression in mouse pectoralis muscle. BMC Res. Notes 2010, 3, 297. [Google Scholar] [CrossRef] [Green Version]
  97. Nakasa, T.; Ishikawa, M.; Shi, M.; Shibuya, H.; Adachi, N.; Ochi, M. Acceleration of muscle regeneration by local injection of muscle-specific micrornas in rat skeletal muscle injury model. J. Cell Mol. Med. 2010, 14, 2495–2505. [Google Scholar] [CrossRef]
  98. Samaeekia, R.; Adorno-Cruz, V.; Bockhorn, J.; Chang, Y.F.; Huang, S.; Prat, A.; Ha, N.; Kibria, G.; Huo, D.; Zheng, H.; et al. Mir-206 inhibits stemness and metastasis of breast cancer by targeting mkl1/il11 pathway. Clin. Cancer Res. 2017, 23, 1091–1103. [Google Scholar] [CrossRef] [Green Version]
  99. Heinemann, F.G.; Tolkach, Y.; Deng, M.; Schmidt, D.; Perner, S.; Kristiansen, G.; Muller, S.C.; Ellinger, J. Serum mir-122-5p and mir-206 expression: Non-invasive prognostic biomarkers for renal cell carcinoma. Clin. Epigenet. 2018, 10, 11. [Google Scholar] [CrossRef] [Green Version]
  100. Tian, R.; Liu, T.; Qiao, L.; Gao, M.; Li, J. Decreased serum microrna-206 level predicts unfavorable prognosis in patients with melanoma. Int. J. Clin. Exp. Pathol. 2015, 8, 3097–3103. [Google Scholar]
  101. Zhang, C.; Yao, C.; Li, H.; Wang, G.; He, X. Serum levels of microrna-133b and microrna-206 expression predict prognosis in patients with osteosarcoma. Int. J. Clin. Exp. Pathol. 2014, 7, 4194–4203. [Google Scholar] [PubMed]
  102. Argiles, J.M.; Fontes-Oliveira, C.C.; Toledo, M.; Lopez-Soriano, F.J.; Busquets, S. Cachexia: A problem of energetic inefficiency. J. Cachexia Sarcopenia Muscle 2014, 5, 279–286. [Google Scholar] [CrossRef] [PubMed]
  103. Penna, F.; Ballaro, R.; Beltra, M.; De Lucia, S.; Costelli, P. Modulating metabolism to improve cancer-induced muscle wasting. Oxid. Med. Cell Longev. 2018, 2018, 7153610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Hardee, J.P.; Montalvo, R.N.; Carson, J.A. Linking cancer cachexia-induced anabolic resistance to skeletal muscle oxidative metabolism. Oxid. Med. Cell Longev. 2017, 2017, 8018197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Harfmann, B.D.; Schroder, E.A.; Kachman, M.T.; Hodge, B.A.; Zhang, X.; Esser, K.A. Muscle-specific loss of bmal1 leads to disrupted tissue glucose metabolism and systemic glucose homeostasis. Skelet. Muscle 2016, 6, 12. [Google Scholar] [CrossRef] [Green Version]
  106. Leskawa, K.C.; Erwin, R.E.; Buse, P.E.; Hogan, E.L. Glycosphingolipid biosynthesis during myogenesis of rat l6 cells in vitro. Mol. Cell Biochem. 1988, 83, 47–54. [Google Scholar] [CrossRef]
  107. Anastasia, L.; Papini, N.; Colazzo, F.; Palazzolo, G.; Tringali, C.; Dileo, L.; Piccoli, M.; Conforti, E.; Sitzia, C.; Monti, E.; et al. Neu3 sialidase strictly modulates gm3 levels in skeletal myoblasts c2c12 thus favoring their differentiation and protecting them from apoptosis. J. Biol. Chem. 2008, 283, 36265–36271. [Google Scholar] [CrossRef] [Green Version]
  108. Gehlert, S.; Bloch, W.; Suhr, F. Ca2+-dependent regulations and signaling in skeletal muscle: From electro-mechanical coupling to adaptation. Int. J. Mol. Sci. 2015, 16, 1066–1095. [Google Scholar] [CrossRef]
  109. Barrey, E.; Saint-Auret, G.; Bonnamy, B.; Damas, D.; Boyer, O.; Gidrol, X. Pre-microrna and mature microrna in human mitochondria. PLoS ONE 2011, 6, e20220. [Google Scholar] [CrossRef]
  110. Zhang, Y.; Lei, W.; Yan, W.; Li, X.; Wang, X.; Zhao, Z.; Hui, J.; Shen, Z.; Yang, J. Microrna-206 is involved in survival of hypoxia preconditioned mesenchymal stem cells through targeting pim-1 kinase. Stem Cell Res. Ther. 2016, 7, 61. [Google Scholar] [CrossRef] [Green Version]
  111. Sun, Y.; Su, Q.; Li, L.; Wang, X.; Lu, Y.; Liang, J. Mir-486 regulates cardiomyocyte apoptosis by p53-mediated bcl-2 associated mitochondrial apoptotic pathway. BMC Cardiovasc. Disord. 2017, 17, 119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Deng, M.; Qin, Y.; Chen, X.; Wang, Q.; Wang, J. Mir-206 inhibits proliferation, migration, and invasion of gastric cancer cells by targeting the muc1 gene. Onco Targets Ther. 2019, 12, 849–859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Hao, W.; Luo, W.; Bai, M.; Li, J.; Bai, X.; Guo, J.; Wu, J.; Wang, M. Microrna-206 inhibited the progression of glioblastoma through bcl-2. J. Mol. Neurosci. 2016, 60, 531–538. [Google Scholar] [CrossRef] [PubMed]
  114. Frantz, C.; Stewart, K.M.; Weaver, V.M. The extracellular matrix at a glance. J. Cell Sci. 2010, 123, 4195–4200. [Google Scholar] [CrossRef] [Green Version]
  115. Thorsteinsdottir, S.; Deries, M.; Cachaco, A.S.; Bajanca, F. The extracellular matrix dimension of skeletal muscle development. Dev. Biol. 2011, 354, 191–207. [Google Scholar] [CrossRef] [Green Version]
  116. Sternlicht, M.D.; Werb, Z. How matrix metalloproteinases regulate cell behavior. Annu. Rev. Cell Dev. Biol. 2001, 17, 463–516. [Google Scholar] [CrossRef] [Green Version]
  117. Desiderio, M.A. Hepatocyte growth factor in invasive growth of carcinomas. Cell Mol. Life Sci. 2007, 64, 1341–1354. [Google Scholar] [CrossRef]
  118. Shiba, N.; Miyazaki, D.; Yoshizawa, T.; Fukushima, K.; Shiba, Y.; Inaba, Y.; Imamura, M.; Takeda, S.; Koike, K.; Nakamura, A. Differential roles of mmp-9 in early and late stages of dystrophic muscles in a mouse model of duchenne muscular dystrophy. Biochim. Biophys. Acta 2015, 1852, 2170–2182. [Google Scholar] [CrossRef] [Green Version]
  119. Dahiya, S.; Bhatnagar, S.; Hindi, S.M.; Jiang, C.; Paul, P.K.; Kuang, S.; Kumar, A. Elevated levels of active matrix metalloproteinase-9 cause hypertrophy in skeletal muscle of normal and dystrophin-deficient mdx mice. Hum. Mol. Genet. 2011, 20, 4345–4359. [Google Scholar] [CrossRef] [Green Version]
  120. Mehan, R.S.; Greybeck, B.J.; Emmons, K.; Byrnes, W.C.; Allen, D.L. Matrix metalloproteinase-9 deficiency results in decreased fiber cross-sectional area and alters fiber type distribution in mouse hindlimb skeletal muscle. Cells Tissues Organs 2011, 194, 510–520. [Google Scholar] [CrossRef] [Green Version]
  121. Winer, A.; Adams, S.; Mignatti, P. Matrix metalloproteinase inhibitors in cancer therapy: Turning past failures into future successes. Mol. Cancer Ther. 2018, 17, 1147–1155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Newby, A.C. Matrix metalloproteinases regulate migration, proliferation, and death of vascular smooth muscle cells by degrading matrix and non-matrix substrates. Cardiovasc. Res. 2006, 69, 614–624. [Google Scholar] [CrossRef] [PubMed]
  123. Phanish, M.K.; Winn, S.K.; Dockrell, M.E. Connective tissue growth factor-(ctgf, ccn2)—A marker, mediator and therapeutic target for renal fibrosis. Nephron Exp. Nephrol. 2010, 114, e83–e92. [Google Scholar] [CrossRef] [PubMed]
  124. Leask, A.; Parapuram, S.K.; Shi-Wen, X.; Abraham, D.J. Connective tissue growth factor (ctgf, ccn2) gene regulation: A potent clinical bio-marker of fibroproliferative disease? J. Cell Commun. Signal. 2009, 3, 89–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Rameshwar, P.; Narayanan, R.; Qian, J.; Denny, T.N.; Colon, C.; Gascon, P. Nf-kappa b as a central mediator in the induction of tgf-beta in monocytes from patients with idiopathic myelofibrosis: An inflammatory response beyond the realm of homeostasis. J. Immunol. 2000, 165, 2271–2277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Grande, J.P.; Melder, D.C.; Zinsmeister, A.R. Modulation of collagen gene expression by cytokines: Stimulatory effect of transforming growth factor-beta1, with divergent effects of epidermal growth factor and tumor necrosis factor-alpha on collagen type i and collagen type iv. J. Lab. Clin. Med. 1997, 130, 476–486. [Google Scholar] [CrossRef]
  127. Ignotz, R.A.; Massague, J. Transforming growth factor-beta stimulates the expression of fibronectin and collagen and their incorporation into the extracellular matrix. J. Biol. Chem. 1986, 261, 4337–4345. [Google Scholar]
  128. Micallef, L.; Vedrenne, N.; Billet, F.; Coulomb, B.; Darby, I.A.; Desmouliere, A. The myofibroblast, multiple origins for major roles in normal and pathological tissue repair. Fibrogenes. Tissue Repair. 2012, 5, S5. [Google Scholar] [CrossRef] [Green Version]
  129. Massague, J. Tgfbeta signalling in context. Nat. Rev. Mol. Cell Biol. 2012, 13, 616–630. [Google Scholar] [CrossRef]
  130. Leask, A.; Abraham, D.J. Tgf-beta signaling and the fibrotic response. FASEB J. 2004, 18, 816–827. [Google Scholar] [CrossRef]
  131. Yu, Q.; Stamenkovic, I. Cell surface-localized matrix metalloproteinase-9 proteolytically activates tgf-beta and promotes tumor invasion and angiogenesis. Genes Dev. 2000, 14, 163–176. [Google Scholar] [PubMed]
  132. Imai, K.; Hiramatsu, A.; Fukushima, D.; Pierschbacher, M.D.; Okada, Y. Degradation of decorin by matrix metalloproteinases: Identification of the cleavage sites, kinetic analyses and transforming growth factor-beta1 release. Biochem. J. 1997, 322, 809–814. [Google Scholar] [CrossRef] [PubMed]
  133. Zimmers, T.A.; Fishel, M.L.; Bonetto, A. Stat3 in the systemic inflammation of cancer cachexia. Semin. Cell Dev. Biol. 2016, 54, 28–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Guadagnin, E.; Mazala, D.; Chen, Y.W. Stat3 in skeletal muscle function and disorders. Int. J. Mol. Sci. 2018, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Bonetto, A.; Aydogdu, T.; Kunzevitzky, N.; Guttridge, D.C.; Khuri, S.; Koniaris, L.G.; Zimmers, T.A. Stat3 activation in skeletal muscle links muscle wasting and the acute phase response in cancer cachexia. PLoS ONE 2011, 6, e22538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Bonetto, A.; Aydogdu, T.; Jin, X.; Zhang, Z.; Zhan, R.; Puzis, L.; Koniaris, L.G.; Zimmers, T.A. Jak/stat3 pathway inhibition blocks skeletal muscle wasting downstream of il-6 and in experimental cancer cachexia. Am. J. Physiol. Endocrinol. Metab. 2012, 303, E410–E421. [Google Scholar] [CrossRef] [Green Version]
  137. Ma, J.F.; Sanchez, B.J.; Hall, D.T.; Tremblay, A.K.; Di Marco, S.; Gallouzi, I.E. Stat3 promotes ifngamma/tnfalpha-induced muscle wasting in an nf-kappab-dependent and il-6-independent manner. EMBO Mol. Med. 2017, 9, 622–637. [Google Scholar] [CrossRef]
  138. Price, F.D.; von Maltzahn, J.; Bentzinger, C.F.; Dumont, N.A.; Yin, H.; Chang, N.C.; Wilson, D.H.; Frenette, J.; Rudnicki, M.A. Inhibition of jak-stat signaling stimulates adult satellite cell function. Nat. Med. 2014, 20, 1174–1181. [Google Scholar] [CrossRef] [Green Version]
  139. Tierney, M.T.; Aydogdu, T.; Sala, D.; Malecova, B.; Gatto, S.; Puri, P.L.; Latella, L.; Sacco, A. Stat3 signaling controls satellite cell expansion and skeletal muscle repair. Nat. Med. 2014, 20, 1182–1186. [Google Scholar] [CrossRef] [Green Version]
  140. Sala, D.; Cunningham, T.J.; Stec, M.J.; Etxaniz, U.; Nicoletti, C.; Dall’Agnese, A.; Puri, P.L.; Duester, G.; Latella, L.; Sacco, A. The stat3-fam3a axis promotes muscle stem cell myogenic lineage progression by inducing mitochondrial respiration. Nat. Commun. 2019, 10, 1796. [Google Scholar] [CrossRef]
  141. Qaed, E.; Wang, J.; Almoiliqy, M.; Song, Y.; Liu, W.; Chu, P.; Alademi, S.; Alademi, M.; Li, H.; Alshwmi, M.; et al. Phosphocreatine improves cardiac dysfunction by normalizing mitochondrial respiratory function through jak2/stat3 signaling pathway in vivo and in vitro. Oxid. Med. Cell Longev. 2019, 2019, 6521218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Boengler, K.; Ungefug, E.; Heusch, G.; Schulz, R. The stat3 inhibitor stattic impairs cardiomyocyte mitochondrial function through increased reactive oxygen species formation. Curr. Pharm. Des. 2013, 19, 6890–6895. [Google Scholar] [CrossRef] [PubMed]
  143. Le Moal, E.; Pialoux, V.; Juban, G.; Groussard, C.; Zouhal, H.; Chazaud, B.; Mounier, R. Redox control of skeletal muscle regeneration. Antioxid. Redox. Signal 2017, 27, 276–310. [Google Scholar] [CrossRef] [PubMed]
  144. Powers, S.K.; Ji, L.L.; Kavazis, A.N.; Jackson, M.J. Reactive oxygen species: Impact on skeletal muscle. Compr. Physiol. 2011, 1, 941–969. [Google Scholar] [PubMed] [Green Version]
  145. Barbieri, E.; Sestili, P. Reactive oxygen species in skeletal muscle signaling. J. Signal Transduct. 2012, 2012, 982794. [Google Scholar] [CrossRef] [Green Version]
  146. Gianotti, T.F.; Castano, G.; Gemma, C.; Burgueno, A.L.; Rosselli, M.S.; Pirola, C.J.; Sookoian, S. Mitochondrial DNA copy number is modulated by genetic variation in the signal transducer and activator of transcription 3 (stat3). Metabolism 2011, 60, 1142–1149. [Google Scholar] [CrossRef]
  147. Vassilev, A.O.; Lorenz, D.R.; Tibbles, H.E.; Uckun, F.M. Role of the leukemia-associated transcription factor stat3 in platelet physiology. Leuk. Lymphoma 2002, 43, 1461–1467. [Google Scholar] [CrossRef]
  148. Phillips, D.; Reilley, M.J.; Aponte, A.M.; Wang, G.; Boja, E.; Gucek, M.; Balaban, R.S. Stoichiometry of stat3 and mitochondrial proteins: Implications for the regulation of oxidative phosphorylation by protein-protein interactions. J. Biol. Chem. 2010, 285, 23532–23536. [Google Scholar] [CrossRef] [Green Version]
  149. Wegrzyn, J.; Potla, R.; Chwae, Y.J.; Sepuri, N.B.; Zhang, Q.; Koeck, T.; Derecka, M.; Szczepanek, K.; Szelag, M.; Gornicka, A.; et al. Function of mitochondrial stat3 in cellular respiration. Science 2009, 323, 793–797. [Google Scholar] [CrossRef] [Green Version]
  150. Gough, D.J.; Corlett, A.; Schlessinger, K.; Wegrzyn, J.; Larner, A.C.; Levy, D.E. Mitochondrial stat3 supports ras-dependent oncogenic transformation. Science 2009, 324, 1713–1716. [Google Scholar] [CrossRef] [Green Version]
  151. Boengler, K.; Hilfiker-Kleiner, D.; Heusch, G.; Schulz, R. Inhibition of permeability transition pore opening by mitochondrial stat3 and its role in myocardial ischemia/reperfusion. Basic Res. Cardiol. 2010, 105, 771–785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Shaw, P.E. Could stat3 provide a link between respiration and cell cycle progression? Cell Cycle 2010, 9, 4294–4296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Acharyya, S.; Villalta, S.A.; Bakkar, N.; Bupha-Intr, T.; Janssen, P.M.; Carathers, M.; Li, Z.W.; Beg, A.A.; Ghosh, S.; Sahenk, Z.; et al. Interplay of ikk/nf-kappab signaling in macrophages and myofibers promotes muscle degeneration in duchenne muscular dystrophy. J. Clin. Investig. 2007, 117, 889–901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Guttridge, D.C.; Mayo, M.W.; Madrid, L.V.; Wang, C.Y.; Baldwin, A.S., Jr. Nf-kappab-induced loss of myod messenger rna: Possible role in muscle decay and cachexia [see comments]. Science 2000, 289, 2363–2366. [Google Scholar] [CrossRef] [Green Version]
  155. Cai, D.; Frantz, J.D.; Tawa, N.E., Jr.; Melendez, P.A.; Oh, B.C.; Lidov, H.G.; Hasselgren, P.O.; Frontera, W.R.; Lee, J.; Glass, D.J.; et al. Ikkbeta/nf-kappab activation causes severe muscle wasting in mice. Cell 2004, 119, 285–298. [Google Scholar] [CrossRef] [Green Version]
  156. Mourkioti, F.; Kratsios, P.; Luedde, T.; Song, Y.H.; Delafontaine, P.; Adami, R.; Parente, V.; Bottinelli, R.; Pasparakis, M.; Rosenthal, N. Targeted ablation of ikk2 improves skeletal muscle strength, maintains mass, and promotes regeneration. J. Clin. Investig. 2006, 116, 2945–2954. [Google Scholar] [CrossRef] [Green Version]
  157. Mourkioti, F.; Rosenthal, N. Nf-kappab signaling in skeletal muscle: Prospects for intervention in muscle diseases. J. Mol. Med. 2008, 86, 747–759. [Google Scholar] [CrossRef] [Green Version]
  158. Li, H.; Malhotra, S.; Kumar, A. Nuclear factor-kappa b signaling in skeletal muscle atrophy. J. Mol. Med. 2008, 86, 1113–1126. [Google Scholar] [CrossRef] [Green Version]
  159. Dogra, C.; Changotra, H.; Mohan, S.; Kumar, A. Tumor necrosis factor-like weak inducer of apoptosis inhibits skeletal myogenesis through sustained activation of nuclear factor-kappab and degradation of myod protein. J. Biol. Chem. 2006, 281, 10327–10336. [Google Scholar] [CrossRef] [Green Version]
  160. Kong, F.M.; Anscher, M.S.; Murase, T.; Abbott, B.D.; Iglehart, J.D.; Jirtle, R.L. Elevated plasma transforming growth factor-beta 1 levels in breast cancer patients decrease after surgical removal of the tumor. Ann. Surg. 1995, 222, 155–162. [Google Scholar] [CrossRef]
  161. Nakano, H.; Shindo, M.; Sakon, S.; Nishinaka, S.; Mihara, M.; Yagita, H.; Okumura, K. Differential regulation of ikappab kinase alpha and beta by two upstream kinases, nf-kappab-inducing kinase and mitogen-activated protein kinase/erk kinase kinase-1. Proc. Natl. Acad. Sci. USA 1998, 95, 3537–3542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Zhao, Q.; Lee, F.S. Mitogen-activated protein kinase/erk kinase kinases 2 and 3 activate nuclear factor-kappab through ikappab kinase-alpha and ikappab kinase-beta. J. Biol. Chem. 1999, 274, 8355–8358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Ninomiya-Tsuji, J.; Kishimoto, K.; Hiyama, A.; Inoue, J.; Cao, Z.; Matsumoto, K. The kinase tak1 can activate the nik-i kappab as well as the map kinase cascade in the il-1 signalling pathway. Nature 1999, 398, 252–256. [Google Scholar] [CrossRef] [PubMed]
  164. Wang, C.; Deng, L.; Hong, M.; Akkaraju, G.R.; Inoue, J.; Chen, Z.J. Tak1 is a ubiquitin-dependent kinase of mkk and ikk. Nature 2001, 412, 346–351. [Google Scholar] [CrossRef] [PubMed]
  165. Furutani, Y.; Umemoto, T.; Murakami, M.; Matsui, T.; Funaba, M. Role of endogenous tgf-beta family in myogenic differentiation of c2c12 cells. J. Cell Biochem. 2011, 112, 614–624. [Google Scholar] [CrossRef] [Green Version]
  166. Wang, H.; Hertlein, E.; Bakkar, N.; Sun, H.; Acharyya, S.; Wang, J.; Carathers, M.; Davuluri, R.; Guttridge, D.C. Nf-kappab regulation of yy1 inhibits skeletal myogenesis through transcriptional silencing of myofibrillar genes. Mol. Cell Biol. 2007, 27, 4374–4387. [Google Scholar] [CrossRef] [Green Version]
  167. Bodine, S.C.; Latres, E.; Baumhueter, S.; Lai, V.K.; Nunez, L.; Clarke, B.A.; Poueymirou, W.T.; Panaro, F.J.; Na, E.; Dharmarajan, K.; et al. Identification of ubiquitin ligases required for skeletal muscle atrophy. Science 2001, 294, 1704–1708. [Google Scholar] [CrossRef]
  168. Siriett, V.; Salerno, M.S.; Berry, C.; Nicholas, G.; Bower, R.; Kambadur, R.; Sharma, M. Antagonism of myostatin enhances muscle regeneration during sarcopenia. Mol. Ther. 2007, 15, 1463–1470. [Google Scholar] [CrossRef]
  169. Dey, B.K.; Gagan, J.; Dutta, A. Mir-206 and -486 induce myoblast differentiation by downregulating pax7. Mol. Cell Biol. 2011, 31, 203–214. [Google Scholar] [CrossRef] [Green Version]
  170. Rathnasamy, G.; Sivakumar, V.; Rangarajan, P.; Foulds, W.S.; Ling, E.A.; Kaur, C. Nf-kappab-mediated nitric oxide production and activation of caspase-3 cause retinal ganglion cell death in the hypoxic neonatal retina. Investig. Ophthalmol. Vis. Sci. 2014, 55, 5878–5889. [Google Scholar] [CrossRef]
  171. Gil da Costa, R.M.; Aragao, S.; Moutinho, M.; Alvarado, A.; Carmo, D.; Casaca, F.; Silva, S.; Ribeiro, J.; Sousa, H.; Ferreira, R.; et al. Hpv16 induces a wasting syndrome in transgenic mice: Amelioration by dietary polyphenols via nf-kappab inhibition. Life Sci. 2017, 169, 11–19. [Google Scholar] [CrossRef] [PubMed]
  172. Zhang, L.; Tang, H.; Kou, Y.; Li, R.; Zheng, Y.; Wang, Q.; Zhou, X.; Jin, L. Mg132-mediated inhibition of the ubiquitin-proteasome pathway ameliorates cancer cachexia. J. Cancer Res. Clin. Oncol. 2013, 139, 1105–1115. [Google Scholar] [CrossRef] [PubMed]
  173. Moore-Carrasco, R.; Busquets, S.; Almendro, V.; Palanki, M.; Lopez-Soriano, F.J.; Argiles, J.M. The ap-1/nf-kappab double inhibitor sp100030 can revert muscle wasting during experimental cancer cachexia. Int. J. Oncol. 2007, 30, 1239–1245. [Google Scholar] [PubMed]
  174. Prado, C.M.; Baracos, V.E.; McCargar, L.J.; Reiman, T.; Mourtzakis, M.; Tonkin, K.; Mackey, J.R.; Koski, S.; Pituskin, E.; Sawyer, M.B. Sarcopenia as a determinant of chemotherapy toxicity and time to tumor progression in metastatic breast cancer patients receiving capecitabine treatment. Clin. Cancer Res. 2009, 15, 2920–2926. [Google Scholar] [CrossRef] [Green Version]
  175. Sheng, J.Y.; Sharma, D.; Jerome, G.; Santa-Maria, C.A. Obese breast cancer patients and survivors: Management considerations. Oncology 2018, 32, 410–417. [Google Scholar]
  176. Vegiopoulos, A.; Rohm, M.; Herzig, S. Adipose tissue: Between the extremes. EMBO J. 2017, 36, 1999–2017. [Google Scholar] [CrossRef]
  177. Baraldo, M.; Geremia, A.; Pirazzini, M.; Nogara, L.; Solagna, F.; Turk, C.; Nolte, H.; Romanello, V.; Megighian, A.; Boncompagni, S.; et al. Skeletal muscle mtorc1 regulates neuromuscular junction stability. J. Cachexia Sarcopenia Muscle 2019. [Google Scholar] [CrossRef] [Green Version]
  178. Arifuzzaman, M.; Ito, A.; Ikeda, K.; Kawabe, Y.; Kamihira, M. Fabricating muscle-neuron constructs with improved contractile force generation. Tissue Eng. Part A 2019, 25, 563–574. [Google Scholar] [CrossRef]
  179. Grosicki, G.J.; Fielding, R.A.; Lustgarten, M.S. Gut microbiota contribute to age-related changes in skeletal muscle size, composition, and function: Biological basis for a gut-muscle axis. Calcif. Tissue Int. 2018, 102, 433–442. [Google Scholar] [CrossRef] [Green Version]
  180. Ticinesi, A.; Lauretani, F.; Tana, C.; Nouvenne, A.; Ridolo, E.; Meschi, T. Exercise and immune system as modulators of intestinal microbiome: Implications for the gut-muscle axis hypothesis. Exerc. Immunol. Rev. 2019, 25, 84–95. [Google Scholar]
  181. Lahiri, S.; Kim, H.; Garcia-Perez, I.; Reza, M.M.; Martin, K.A.; Kundu, P.; Cox, L.M.; Selkrig, J.; Posma, J.M.; Zhang, H.; et al. The gut microbiota influences skeletal muscle mass and function in mice. Sci. Transl. Med. 2019, 11, eaan5662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Sotiriou, C.; Neo, S.Y.; McShane, L.M.; Korn, E.L.; Long, P.M.; Jazaeri, A.; Martiat, P.; Fox, S.B.; Harris, A.L.; Liu, E.T. Breast cancer classification and prognosis based on gene expression profiles from a population-based study. Proc. Natl. Acad. Sci. USA 2003, 100, 10393–10398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Sharma, G.N.; Dave, R.; Sanadya, J.; Sharma, P.; Sharma, K.K. Various types and management of breast cancer: An overview. J. Adv. Pharm. Technol. Res. 2010, 1, 109–126. [Google Scholar] [PubMed]
  184. Advani, S.M.; Advani, P.G.; VonVille, H.M.; Jafri, S.H. Pharmacological management of cachexia in adult cancer patients: A systematic review of clinical trials. BMC Cancer 2018, 18, 1174. [Google Scholar] [CrossRef] [Green Version]
  185. Penna, F.; Ballaro, R.; Beltra, M.; De Lucia, S.; Garcia Castillo, L.; Costelli, P. The skeletal muscle as an active player against cancer cachexia. Front. Physiol. 2019, 10, 41. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Systemic actions of breast cancer on skeletal muscles lead to functional limitations. Tumors in the breast release growth factors and cytokines into the circulating system. In response to tumors, host immune cells in breast tissue and other organs also release growth factors and cytokines into circulation. These growth factors and cytokines activate NF-κB in tumors, which further enhance the expression and release of growth factors and cytokines into circulation. Circulating growth factors and cytokines such as TGF-β and TNF-α are transported into skeletal muscles where, through their selective receptors, activate signaling molecules such as NF-κB in myogenic cells. Activated NF-κB regulates a series of molecules through its downstream signaling, feedback regulations, and cross-talk interactions. Tumor-induced reduction of Cox-IV, Prka-γ and Atp2a1 in skeletal muscles results in defective energetic regulation leading to fatigue in breast cancer patients. miR-486 and miR-206 are expressed in the mitochondria of myofibers, but their role in regulating energy remains unknown. Reduced expression of Pax7, MyoD, Myf5/6, and MyoG in skeletal muscles impairs proliferation and differentiation of MuSCs, which is exacerbated by downregulation of Bcl-2, miR-486, and miR-206. These actions lead to myogenic defects such as reduced number and size of myofibers, which is associated with myoatrophy. Downregulation of Mmp-9 and laminin A in skeletal muscles could contribute to increased ECM deposition and myofibrosis. Decreased expression of muscle-specific miR-486 and miR-206 may contribute to their lower levels in circulation. Whether the neuromuscular junction is affected by breast cancer is unknown. Furthermore, whether the tumor itself or cancer treatment affects the gut microbiome, which then changes muscle function, is unknown. Overall, breast cancer-induced impairment of myogenesis, energetic inefficiency, and ECM remodeling leads to functional limitations, which may be manifested clinically as fatigue, muscle weakness, fibromyalgia with and without overt cachexia. Pharmacological intervention through inhibitors of NF-κB such as DMAPT is a promising therapeutic strategy.
Figure 1. Systemic actions of breast cancer on skeletal muscles lead to functional limitations. Tumors in the breast release growth factors and cytokines into the circulating system. In response to tumors, host immune cells in breast tissue and other organs also release growth factors and cytokines into circulation. These growth factors and cytokines activate NF-κB in tumors, which further enhance the expression and release of growth factors and cytokines into circulation. Circulating growth factors and cytokines such as TGF-β and TNF-α are transported into skeletal muscles where, through their selective receptors, activate signaling molecules such as NF-κB in myogenic cells. Activated NF-κB regulates a series of molecules through its downstream signaling, feedback regulations, and cross-talk interactions. Tumor-induced reduction of Cox-IV, Prka-γ and Atp2a1 in skeletal muscles results in defective energetic regulation leading to fatigue in breast cancer patients. miR-486 and miR-206 are expressed in the mitochondria of myofibers, but their role in regulating energy remains unknown. Reduced expression of Pax7, MyoD, Myf5/6, and MyoG in skeletal muscles impairs proliferation and differentiation of MuSCs, which is exacerbated by downregulation of Bcl-2, miR-486, and miR-206. These actions lead to myogenic defects such as reduced number and size of myofibers, which is associated with myoatrophy. Downregulation of Mmp-9 and laminin A in skeletal muscles could contribute to increased ECM deposition and myofibrosis. Decreased expression of muscle-specific miR-486 and miR-206 may contribute to their lower levels in circulation. Whether the neuromuscular junction is affected by breast cancer is unknown. Furthermore, whether the tumor itself or cancer treatment affects the gut microbiome, which then changes muscle function, is unknown. Overall, breast cancer-induced impairment of myogenesis, energetic inefficiency, and ECM remodeling leads to functional limitations, which may be manifested clinically as fatigue, muscle weakness, fibromyalgia with and without overt cachexia. Pharmacological intervention through inhibitors of NF-κB such as DMAPT is a promising therapeutic strategy.
Cancers 12 00194 g001

Share and Cite

MDPI and ACS Style

Wang, R.; Nakshatri, H. Systemic Actions of Breast Cancer Facilitate Functional Limitations. Cancers 2020, 12, 194. https://doi.org/10.3390/cancers12010194

AMA Style

Wang R, Nakshatri H. Systemic Actions of Breast Cancer Facilitate Functional Limitations. Cancers. 2020; 12(1):194. https://doi.org/10.3390/cancers12010194

Chicago/Turabian Style

Wang, Ruizhong, and Harikrishna Nakshatri. 2020. "Systemic Actions of Breast Cancer Facilitate Functional Limitations" Cancers 12, no. 1: 194. https://doi.org/10.3390/cancers12010194

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop