Journal of Osteoporosis and Physical Activity

Journal of Osteoporosis and Physical Activity
Open Access

ISSN: 2329-9509

+44 1478 350008

Review Article - (2017) Volume 5, Issue 1

Why Age Matters: Inflammation, Cancer and Hormones in the Development of Sarcopenia

Saul D1*, Schilling AF1 and Kosinsky RL2
1Clinic for Trauma Surgery, Orthopaedic Surgery and Plastic Surgery, University Medical Center Goettingen, Göttingen, Germany
2Department of General, Visceral and Pediatric Surgery, University Medical Center Goettingen, Göttingen, Germany
*Corresponding Author: Saul D, Clinic for Trauma Surgery, Orthopaedic Surgery and Plastic Surgery, University Medical Center Goettingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany, Tel: 05513922462, Fax: 055139 8787 Email:

Abstract

In an aging population, the decline in muscle mass and strength in combination with a high prevalence of osteoporosis and cancer leads to a multitude of clinical manifestations. In the recent years, mouse models of wasting in cancer and inflammation, including xenograft, genetic and chemically induced models, allowed to uncover several key mechanisms underlying muscle loss. These include inflammation, hormone alterations and deregulated protein degradation. Inflammation is associated with increased expression of tumor necrosis factor α (TNF-α), nuclear factor κB (NF-κB), and interleukin (IL)-6 and is therefore linked to inflammatory bowel diseases or chronic obstructive pulmonary disease (COPD). Moreover, active NF-κB signaling and IL-6 secretion commonly occurs in malignancies and cancer-induced cachexia. The ubiquitin proteasome-mediated degradation of proteins represents a second pathway underlying sarcopenia and is partially initiated by inflammatory signaling. Consequently, increased levels of the E3 ligases Muscle RING-Finger Protein-1 (MuRF1), Atrogin-1/Muscle Atrophy F-box (MAFbx), and tumor necrosis factor α receptor adaptor protein 6 (TRAF6) are associated with high rates of protein degradation. Furthermore, hormonal alterations, such as the aging-related decline of growth hormone (GH) and insulin-like growth factor 1 (IGF-1), lead to a reduction of muscle mass.

Interestingly, experimental targeting of several of those sarcopenia-associated factors in vivo resulted in a rescue of muscle mass and function. While therapeutic options nowadays still need to be evaluated regarding their clinical practicability, IL-6 antibodies, inhibition of cyclooxygenases and inhibitors of myostatin appear promising.

<

Keywords: Osteoporosis; Sarcopenia; Cachexia; Myostatin; Muscle wasting; Inflammation; Baicalein.

Introduction

Definitions, epidemiology and costs

Loss of neuro-muscular function [1,2] leads to falls and fall-related injuries that protract the encumbrance for the elderly [3]. This is especially important in a population with a high prevalence of osteoporosis, an age-related loss of bone density and strength [4,5]. On the neural side, the basic functional unit of the neuromuscular system, the motor unit, and its neural inputs profoundly change with age (for review see [6]). On the muscular side, there are three major types of loss in muscle mass described, i.e., atrophy, and cachexia and sarcopenia. In atrophy, muscle fiber size decreases while the total number remains unchanged. In cachexia both adipose and muscle tissue are decreased, leading to diverse consequences dependent on the concomitant medical condition. In contrast, sarcopenia leads to a decrease in fiber size and number. Interestingly, the sarcopenia-related decline in muscle mass during aging is frequently accompanied by an increase in adipose tissue [7-10]. This correlation between obesity and sarcopenia, referred to as "sarcopenic obesity" (SO), was investigated in detail and confirmed in the InCHIANTI study in 1998 [11]. In addition, muscle wasting is often concomitant with miscellaneous severe diseases such as cancer, sepsis, liver cirrhosis, chronic obstructive pulmonary disease (COPD [12]) or chronic heart failure [13-17].

The forfeiture of muscles due to cancer cachexia has been estimated to affect >1.3 million [18,19] in the US population alone. On a worldwide prospect, more than 7.4 million deaths can be attributed to muscle wasting annually [20] with currently about 1% overall prevalence of cachexia in industrialized countries [21]. Intriguingly, 30% of cancer patients die due to cachexia, while more than 50% of patients die with cachexia being present [22]. The healthcare costs were reported around 18.5 billion dollar in the US in 2000 [23] and it was estimated for Europe that sarcopenia leads to increases in hospitalization costs between 34% and 58% [24].

These estimations are complicated by the fact that there is still no consensus on how to diagnose sarcopenia.

Diagnosis

In contrast to osteoporosis, which can routinely be defined by declining bone mineral density since 1994 [25], diagnosis of sarcopenia and muscle wasting remains challenging. Generally, muscle mass, strength and physical performance are evaluated to detect sarcopenia. Methods include computed tomography, magnetic resonance imaging, the assessment of handgrip strength and gait speed as recently reviewed by Choi [26]. According to the International Working Group on Sarcopenia (IWGS), the loss of muscle protein mass and function [27] can be further defined as a low relative appendicular skeletal muscle mass (RASM, lean mass divided by height squared [kg/m2]) [28-30]. In contrast, the criteria defined by the European Working Group on Sarcopenia in Older People (EWGSOP) include a decline in muscle mass plus either low muscle strength or low physical performance [31]. The latter takes handgrip strength or grip strength as parameters of muscle strength, which are interestingly long-term predictors of mortality [6,32].

Models of muscle wasting

In aging, cancer, inflammation and hormone alterations are major contributing factors of muscle wasting. Accordingly, several strategies were described to investigate the mechanisms of muscle wasting in mice based on these three aspects. Generally, the loss of muscle mass was observed in xenograft and genetic mouse models as well as after chemical treatments (Figure 1).

osteoporosis-and-physical-activity-aging-related

Figure 1: The interplay of aging-related factors in the development of sarcopenia and potential therapeutic approaches.

The progress of aging is frequently accompanied by the development of cancer, inflammatory diseases, and hormonal alterations. These aspects contribute to the increased inflammatory signaling, however, also changes in myostatin levels can be observed during aging. In inflammatory processes, NF-κB is commonly activated which leads to elevated synthesis of the E3 ligase MuRF1. In addition, FOXO levels are elevated which induce the E3 ligase Atrogin1/MAFbx. Consequently, the ubiquitin-proteasome system is deregulated resulting in high levels of protein degradation. Moreover, aging is frequently accompanied by low levels of testosterone, estrogen, GH, and IGF-1, which normally inhibits AKT/PI3K signaling. Accordingly, the decrement of GH and IGF-1 leads to potent AKT/ PI3K activity inducing FOXO. Similarly, myostatin was described as a further factor inducing FOXO, for instance by being involved into the phosphorylation of SMAD2/3 and thereby facilitating their binding to SMAD4. Therefore, cancer, inflammatory signaling, hormonal changes, myostatin, and deregulations in the ubiquitin-proteasome system can contribute to the development of sarcopenia which is characterized by a decrease in muscle mass, strength and physical performance. Several mouse models reflect symptoms of this disease and various potential therapeutic measures were successfully tested in these experimental animals. So far, no potent drug to reduce the burden of sarcopenia was generated for clinical practice.

Cancer

Xenograft models describe the transplantation of cells, such as cancer cell lines, subcutaneously into the flanks of immunodeficient mice, thereby frequently developing large tumors until muscle wasting becomes apparent. For instance, in Lewis lung carcinoma (LLC) mice, LLC cells are transplanted subcutaneously [33]. Studies showed that on day 21 and 25 after inoculation, fat mass and lean mass were significantly reduced [34]. In this model, the skeletal muscle-isoform of gp130 (skm-gp130) seems to regulate muscle mass signaling crucially through STAT3 and p38 [35]. In another xenograft model, the Murine adenocarcinoma 16 (MAC16) mouse, UCP2 and -3-expression both seem to protract muscle wasting, whether inflammatory cytokines seem not to interfere [36]. Twenty days after injecting MAC16 colon adenocarcinoma cells, lean body mass was significantly reduced, independent from the cumulative food intake [37]. Another wellestablished model is the C26 colorectal carcinoma model [38], in which a C26-tumor fragment is subcutaneously transplanted. After fifteen days, mice lost approximately 3g; after another one week, the animals lost 12g in comparison to control groups, especially by wasting of adipose and muscle tissue [39,40]. Another xenograft model for muscle wasting is the Solid Ehrlich carcinoma in which Ehrlich-Lettre ascites (EAC) carcinoma tumor cells are injected subcutaneously. 28 days after injection, handgrip strength and body weight were significantly reduced, whereas food intake was not altered [41,42]. Finally, the Yoshida AH-130–model is generated by transplantation of AH-130 ascites hepatoma cells [43]. Through activation of the myostatin-system, the ubiquitin proteolytic system is activated via Atrogin-1, MuRF-1 and E214k. Muscle loss is mainly detected in M. gastrocnemius after seven days. While xenograft transplantations reflect a relatively simple approach which can lead to the desired outcome of muscle loss, they do not reflect the human situation properly, for instance, due to the immunodeficiency [13].

This problem can be overcome by using genetic mouse models in which frequently certain mutations or knockouts are induced to study the role of a specific gene in vivo . The adenomatous polyposis coli (APC) multiple intestinal neoplasia (ApcMin/+) model, for example, is characterized by a mutation in the APC tumor-suppressor. The heterozygous Min-mutation leads to the formation of numerous adenomas, forming throughout the intestine and consequently causing muscle wasting and fatigue. Regularly, the animals die at an age of four to six months. In these mice, lean muscle mass and single muscle mass decreases significantly between week 13 and 22 [44,45]. Intriguingly, in this model weight loss seems to be accompanied by an increase in IL-6 levels, thereby especially affecting M. gastrocnemius [46-48].

Furthermore, pancreatic adenocarcinomas often correlate with a cachectic phenotype [32,33] which indicates the potential of the common LSL-KRASG12D/+; LSL-TRP53R172H/+; Pdx-1-Cre (KPC) mouse model for studying muscle wasting, especially in M. quadriceps femoris. In these KPC-mice, pancreatic adenocarcinoma develops with median survival of five months [13]. Underlying molecular mechanisms involve increased levels of Atrogin-1 and MuRF1 [49-52].

Inflammation

As previously mentioned, inflammatory signaling is highly associated with sarcopenia. Mechanistically, increased tumor necrosis factor (TNF)-α levels lead to inflammation via nuclear factor κB (NF- κB) activation and finally result in muscle wasting. For example, the SP-C/TNF model is characterized by increased TNF-α level in lung and serum due to the lung-specific surfactant protein c-promoter (SFTPC). Consequently, pulmonary inflammation and thus wasting and impaired muscle regeneration were observed in these animals [33,34], preferably in male mice. Similarly, the increase of the TNF-like weak inducer of apoptosis (TWEAK) protein exacerbates overall muscle atrophy, whereas transgenic ablation decreases muscle loss. This effect was primarily observed in M. soleus which showed reduced fiber size after 6 months and mostly increased fast-type fibers. As expected, these mice were characterized by an increased abundance of the E3 ligase MuRF1, however, MAFbx levels remained unchanged [35,36]. Moreover, the muscle-specific expression of IκB kinase (IKK) in MIKK mice is required for ubiquitination and degradation of IκBα [37], which activates NF-κB. Under the control of the muscle creatine kinase promoter, constitutively active IKKβ via its phosphorylation of S177 and S181 leads to muscle wasting by impaired skeletal muscle mass. Mostly, number of fibers is normal, but fiber diameter, area and function are significantly reduced [38,39]. Other genetic models such as muscle creatine kinase-Cre mice on a Cre-LoxP system have been described, that make it possible to knockout specific proteins like TRAF6 [40,41] or TGFβR-II [42] in the muscle, leading to prevented cancer cachexia in an experimental model [41]. Besides these genetic models for inflammation, there are also chemical models to induce inflammatory signaling. Colitis induced by chemical treatments reflects the human situation in inflammatory bowel diseases (IBDs) and can serve as model for wasting [49]. For instance, it was demonstrated that dextran sodium sulfate (DSS)- and trinitrobenzene sulphonic acid (TNBS)-mediated colitis deregulated the expression of smooth muscle contractile proteins in vivo , thereby impairing muscle function [43]. In addition, muscles of TNBS-treated mice were characterized by increased Atrogin-1 and MuRF1 mRNA levels and elevated protein degradation rates [44].

Hormones

In contrast to cancer- and inflammation-related experimental animals, mouse models for hormonal deficiency and resulting effects on muscle mass are rare. For instance, due to its major function in muscle development and function, mice with a global reduction of insulin-like growth factor (IGF)-1 were generated. Unfortunately, these mice were characterized by impaired tissue development and subsequent postnatal lethality [45]. It was possible to overcome this problem by the generation of mice with a muscle-specific IGF-1R/IR (insulin receptor) knockout. These animals showed a severe decrease in muscle mass [46]. In addition, one of the main contributing component of sarcopenia is the growth hormone receptor (GHR). It could be demonstrated that GHR loss in mice resulted in a lower muscle mass in M. soleus and M. tibialis anterior [47] and, therefore, GHR knockout can serve as a model of age-related sarcopenia. Finally, the reduction of estrogen levels via ovariectomy was shown to be a valuable model for sarcopenia [48], especially when housing animals in an enriched environment [49].

Finally, the administration of Angiotensin II via osmotic minipumps over two weeks leads to a weight-reduction of 56% after one week and 41% after two weeks. Mechanistically, reactive oxygen species and IGF-1 lead to apoptosis and protein degradation and therefore muscle wasting [50,51], which reveals the Angiotensin IIInfusion as an uncommon model for muscle wasting.

While mouse models do have several limitations, such as the high effort to generate them, heterogeneous knockout efficiencies, and a more rapid muscle loss compared to humans, they give us the opportunity to dissect molecular pathways.

Pathogenesis

Based on these models, four main pathways involved in the development of muscle wasting have been described: Inflammation, ubiquitin proteasome-mediated degradation, hormonal alterations and malignancies. Molecular key players in these pathways were defined and reflect potential therapeutic targets.

Inflammation

Inflammatory signaling, one of the key mechanisms underlying muscle loss, frequently involves the activation of the NF-κB via TNF, finally resulting in the production of pro-inflammatory cytokines including interleukins [52]. For instance, when investigating sarcopenic obesity, it was demonstrated that inflammatory signaling is frequently induced in adipose tissue in which adipocytes and infiltrating macrophages secrete pro-inflammatory cytokines. In fact, high levels of C-reactive protein (CRP), IL-6 and TNF-α [53] were associated with decreased muscle mass and strength in patients. Interestingly, TNF-α secretion was shown to promote apoptotic signaling with age. Since muscle fibers are multinucleated, apoptosis does not necessarily interfere with fiber integrity, however, it was found to contribute to sarcopenia [54,55]. Moreover, inflammatory bowel diseases were associated with sarcopenia. In fact, besides IBD-related malnutrition which is associated with weight loss, inflammatory signaling was shown to significantly contribute to sarcopenia. Generally, IBDs such as Crohn’s disease (CD) and ulcerative colitis (UC) are characterized by activated NF-κB signaling [56] and up to 60% of Crohn’s disease (CD) patients were affected by severe muscle loss [57]. Accordingly, treating CD patients with the TNF-α inhibitor Infliximab resulted in an increase in muscle volume and strength, as well as in a decrement in IL-6 levels [58]. Similarly, COPD is associated with increased NF-κB activation [59] and 15% of affected individuals develop sarcopenia [12]. Furthermore, COPD patients were more likely to present SO resulting in a severe systemic inflammation and reduced exercise capacity compared to individuals without COPD [60].

Ubiquitin proteasome-mediated degradation

Besides inflammatory signaling, the homeostasis between protein synthesis and degradation is not maintained in sarcopenia. In addition to its function in cytokine production, activated NF-κB can translocate into the nucleus to mediate the induction of Muscle RING-Finger Protein-1 (MuRF1). MuRF1 is an E3 ligase which conjugates protein substrates with an ubiquitin molecule, thereby potentially targeting its substrates for proteasomal degradation. Interestingly, MuRF1 activation interferes with the sarcomere integrity by degrading the myosin heavy chain and further filament components [61,62]. Similarly, the E3 ligase Atrogin-1/Muscle Atrophy F-box (MAFbx) is induced upon constitutive Forkhead box O (FOXO) 3 activation [63]. In addition, myostatin, a protein exclusively expressed in smooth muscle cells, was shown to increase FOXO levels by assisting during the phosphorylation of the transcription factor SMAD2/3 and thereby facilitating its binding to SMAD4 as reviewed in detail elsewhere [64]. Consistently, MuRF1 mRNA levels were increased in limb muscles of cachectic COPD patients [65] and MAFBx in smokers [66]. Interestingly, the deletion of MuRF1 and MAFbx in vivo could prevent muscle loss [67] and treatment of myoblasts with IGF-1 in vitro was able to inhibit the expression of MuRF1 and Atrogin-1 [68]. Moreover, the E3 ligase tumor necrosis factor α receptor adaptor protein 6 (TRAF6) is upregulated in skeletal muscle wasting through the activation of JNK1/2 [41]. Consequently, the downstream signal molecules MAFBx and MuRF1 were upregulated [69]. Interestingly, upon cytokine stimulation TRAF6 was shown to be involved in the activation of NF-κB and Protein Kinase B(PKB/AKT)/ Phosphoinositide 3-kinase (PI3K) signaling [70,71]. In addition, it was described to be critical for homeostasis of satellite stem cell function and therefore myofiber regeneration [72]. Accordingly, inhibition of TRAF6 resulted in the preservation of cancer cachexia [41]. Interestingly, TRAF6 loss exacerbates DSS-induced colitis in mice [73].

Hormonal alterations

Aging is accompanied by changed levels of hormone production. For instance, secretion of growth hormone (GH) and IGF-1 decline with age and, interestingly, this phenomenon is associated with sarcopenia [74]. Generally, binding of IGF-1 to its receptor leads to the activation of the AKT/PI3K pathway. Decreased AKT/PI3K signaling results in impaired phosphorylation of FOXO members resulting in the nuclear translocation of this transcription factor family. Nuclear localization and constitutive activation of FOXO3 induced transcription of Atrogin/MAFbx and thereby severe muscle atrophy [63].

Besides the effect on GH and IGF-1, the decline in testosterone and estrogen levels were correlated with aging and sarcopenia [75]. Interestingly, testosterone supplementation increases IGF-1 expression in vitro [76] and exerts a positive effect on muscle mass and strength in older men [77]. In contrast, the effect of estrogen was highly dependent on the route of administration since IGF-1 levels increased after transdermal and decreased after oral treatment [78].

Malignancies

Muscle wasting was described as comorbidity in several human malignancies, including colorectal, pancreatic, and lung cancer [79]. Similar to sarcopenic obesity and hormonal alterations, malignancies frequently develop in elderly individuals. In fact, 50% of all cancer diagnoses and 70% of cancer-related deaths are attributed to an age ≥ 65 years [80]. As recently reviewed by Gordon and colleagues, inflammatory responses, including active NF-κB signaling, significantly contributes to cancer-related cachexia. For instance, lung cancer patients are characterized by highly elevated NF-κB activity in limb muscles [81] and in a mouse model for lung cancer, IL-6 secretion modulated muscle mass via inducing FOXO3, Signal transducer and activator of transcription 3 (STAT3), and the kinase p38 [82]. Moreover, myostatin was shown to be secreted by C26 colon cancer cells in vitro and to upregulate MuRF1 and Atrogin-1 levels [83]. The increased abundance of MuRF1 and Atrogin-1, thus, deregulated ubiquitin proteasome‐mediated degradation could reflect another mechanism associated with cancer cachexia. Accordingly, myostatin inhibition increased skeletal muscle mass and strength in vivo [84].

Therapy

Therapeutic options in sarcopenia consist of physical training [27], sufficient nutrition and pharmacotherapy [85-87]. Pharmacotherapy is based on the highly heterogeneous underlying molecular pathways associated with muscle wasting. Thus, possible therapeutic options aim at different targets.

Since inflammation is one of the key players mediating the development of sarcopenia, several studies sought to inhibit underlying pathways in order to prevent muscle loss. For instance, the inhibition of TNF-α, an activator of NF-κB signaling, using Thalidomide was able to reduce the severity of muscle loss in rats with biliary cirrhosis [88]. However, as described in a Cochrane review, the effects of Thalidomide in the treatment of cancer-associated cachexia are not completely clear. Therefore, its suitability for clinical practice still needs to be elucidated [89]. As mentioned earlier, treating IBD patients with Infliximab, a potent anti-TNF-α agent, resulted in an increase in muscle mass and strength [58]. Similarly, administration of IL-6-neutralizing antibodies could restore tumor-induced cachexia in mice [90]. Moreover, inhibition of cyclooxygenases (COX), mediators of inflammatory responses, represents a potential therapeutic strategy [91]. Interestingly, we recently demonstrated the beneficial effect of the lipoxygenase inhibitor baicalein on muscles [48]. Generally, this agent is a potent inhibitor of inflammation by interfering with COX-2 gene expression [92]. During our study we discovered that baicalein treatment increased muscle fiber area and diameter and led to elevated number of capillaries per fiber in non-ovariectomized as well as ovariectomized rats. Together, a protective effect of baicalein on muscle cells was suggested due to the stimulation of angiogenesis in skeletal muscle and by reducing the muscle loss mediated by decreased estrogen levels [48]. Studies like this indicate the potential of antiinflammatory agents in the treatment of sarcopenia

Furthermore, the success of drugs interfering with hormone deregulation was reported. Hormone supplementation is FDAapproved for several syndromes associated with muscle wasting. However, while testosterone therapy was able to increase muscle strength in several studies, GH supplementation failed to induce changes in muscle function [93] but could increase muscle mass [94,95]. Likewise on the hormone level, encouraging effects of vitamin D on muscle strength [96] in vitamin D-deficient individuals [97,98] have been discovered.

Another apparent target is myostatin, which inhibits protein synthesis and, accordingly, systemical administration of myostatin in vivo resulted in severe muscle loss. Antibodies targeting myostatin were generated and in vivo data show increased muscle mass and grip strength in mice [84]. Furthermore, myostatin-inhibitors such as propeptide-Fc (GDF8 propeptide-FC) increased muscle mass in vivo [99]. The glycoprotein Follistatin inhibits myostatin and therefore leads to muscle hypertrophy in vivo [100,101]. Similarly, soluble myostatin receptors reduced muscle atrophy [102] and siRNA-mediated myostatin silencing showed promising results in mouse models [103]. Similarly, adeno-associated virus serotype 8 (AAV8) vectors can be used to transfect myostatin propeptides and subsequently raise muscle growth [69].

Another potential target is the E3 ligase TRAF6 which is involved in protein degradation via the ubiquitin-proteasome system. Interestingly, anti-TRAF6 siRNA administered in a mouse model rescued muscle atrophy in vivo [69]. Taken together, several molecular players involved in the development of sarcopenia were defined and inhibition of these factors was shown to have a beneficial effect on muscle mass and function in several in vivo studies. However, while several promising drugs were generated, the ultimate objective to use a therapeutic agent in clinical practice to lower sarcopenia burden in patients still requires further investigations.

Conclusion

Sarcopenia reflects a major health problem by affecting the individuals’ quality of life as well as by causing a significant economic burden for society. Over the last decade, several molecular players involved in the development of this disease were defined in mouse models as well as in patient samples. A number of in vivo studies indicated promising therapeutic strategies, however, no suitable drug was generated for clinical practice so far. Thus, further extensive research is required in this field.

Genetic mouse models offer attractive experimental setups since they reflect the human situation more closely than xenograft models. In future projects, molecular players of muscle loss could be identified by next generation sequencing using sarcopenia patient-derived muscle samples. Subsequently, the function of potential mediators of this medical condition could be defined by performing muscle-specific knockouts in mice. Finally, methods to rescue these phenotypes could include treatment with anti-inflammatory drugs, specific smallmolecule inhibitors or physical training.

Finally, inflammation seems to be one of the major mechanisms involved in muscle loss and, importantly, inflammatory signaling accompanies a variety of further pathological conditions including cancer, IBDs and COPD. Therefore, muscular targeting of antiinflammatory agents would be highly promising for future studies.

References

  1. Rosenberg IH (1997) Sarcopenia: Origins and clinical relevance. J Nutr 127: 990S-991S.
  2. Thomas T, Milagros JT, Gregorio V (2016) Muscle fibers secrete FGFBP1 to slow degeneration of neuromuscular synapses during aging and progression of ALS. Journal of neuroscience 37: 70-82
  3. He H, Liu Y, Tian Q, Papasian CJ, Hu T, et al. (2016) Relationship of sarcopenia and body composition with osteoporosis. Osteoporos Int. 27: 473-482.
  4. Johnell O, Kanis JA (2006) An estimate of the worldwide prevalence and disability associated with osteoporotic fractures. Osteoporos Int 17: 1726-1733.
  5. Hernlund E, Svedbom A, Ivergård M, Compston J, Cooper C (2013) Osteoporosis in the European Union: Medical management, epidemiology and economic burden. A report prepared in collaboration with the International Osteoporosis Foundation (IOF) and the European Federation of Pharmaceutical Industry Associations (EFPIA). Arch Osteoporos 8:136.
  6. Janssen I, Heymsfield SB, Ross R (2002) Low relative skeletal muscle mass (sarcopenia) in older persons is associated with functional impairment and physical disability. J Am Geriatr Soc 50: 889-896.
  7. Villareal DT, Apovian CM, Kushner RF, Klein S (2005) Obesity in older adults: Technical review and position statement of the American Society for Nutrition and NAASO, The Obesity Society. Am J Clin Nutr 82: 923-934.
  8. Flegal KM, Carroll MD, Kuczmarski RJ, Johnson CL (1998) Overweight and obesity in the United States: Prevalence and trends, 1960-1994. Int J Obes Relat Metab Disord 22: 39-47.
  9. Gallagher D, Visser M, De Meersman RE, Sepúlveda D, Baumgartner RN, et al. (1997) Appendicular skeletal muscle mass: Effects of age, gender and ethnicity. J Appl Physiol 83: 229-239.
  10. Baumgartner RN, Stauber PM, McHugh D, Koehler KM, Garry PJ (1995) Cross-sectional age differences in body composition in persons 60+ years of age. J Gerontol A Biol Sci Med Sci 50: M307-M316.
  11. Schrager MA, Metter EJ, Simonsick E, Ble A, Bandinelli S, et al. (1985) Sarcopenic obesity and inflammation in the InCHIANTI study. J Appl Physiol 102: 919-925.
  12. Jones SE, Maddocks M, Kon SS, Canavan JL, Nolan CM, et al. (2015) Sarcopenia in COPD: prevalence, clinical correlates and response to pulmonary rehabilitation. Thorax 70: 213-218.
  13. Holecek M (2012) Muscle wasting in animal models of severe illness. Int J Exp Pathol 93: 157-171.
  14. Johns N, Stephens NA, Fearon KC (2013) Muscle wasting in cancer. Int J Biochem Cell Biol 45: 2215-2129.
  15. Bindels LB, Delzenne NM (2013) Muscle wasting: The gut microbiota as a new therapeutic target? Int J Biochem Cell Biol 45: 2186-2190.
  16. Kalafateli M, Konstantakis C, Thomopoulos K, Triantos C (2015) Impact of muscle wasting on survival in patients with liver cirrhosis. World J Gastroenterol 21: 7357-7361.
  17. Cohen S, Nathan JA, Goldberg AL (2015) Muscle wasting in disease: Molecular mechanisms and promising therapies Nat Rev Drug Discov 14: 58-74.
  18. Romanick M, Thompson LV, Brown-Borg HM (2013) Murine models of atrophy, cachexia and sarcopenia in skeletal muscle. Biochim Biophys Acta 1832: 1410-1420.
  19. Morley JE, Thomas DR, Wilson MM (2006) Cachexia: Pathophysiology and clinical relevance. Am J Clin Nutr 83: 735-743.
  20. Dodson S, Baracos VE, Jatoi A, Evans WJ, Cella D, et al. (2011) Muscle wasting in cancer cachexia: Clinical implications, diagnosis, and emerging treatment strategies. Annu Rev Med 62: 265-279.
  21. Hall DT, Ma JF, Marco SD, Gallouzi IE (2011) Inducible nitric oxide synthase (iNOS) in muscle wasting syndrome, sarcopenia and cachexia. Aging 3: 702-715.
  22. von Haehling S, Anker SD (2010) Cachexia as a major underestimated and unmet medical need: Facts and numbers. J Cachexia Sarcopenia Muscle 1: 1-5.
  23. Janssen I, Shepard DS, Katzmarzyk PT, Roubenoff R (2004) The healthcare costs of sarcopenia in the United States. J Am Geriatr Soc 52: 80-85.
  24. Sousa AS, Guerra RS, Fonseca I, Pichel F, Ferreira S, et al. (2016) Financial impact of sarcopenia on hospitalization costs. Eur J Clin Nutr 70: 1046-1051.
  25. Assessment of fracture risk and its application to screening for postmenopausal osteoporosis. Report of a WHO study group. World Health Organ Tech Rep Ser 843: 1-129.
  26. Choi KM (2013) Sarcopenia and sarcopenic obesity. Endocrinol Metab 28: 86-89.
  27. Rolland Y, Czerwinski S, Abellan Van Kan G, Morley JE, Cesari M et al. (2008) Sarcopenia: Its assessment, etiology, pathogenesis, consequences and future perspectives. J Nutr Health Aging 12: 433-450.
  28. Cooper C, Dere W, Evans W, Kanis JA, Rizzoli R, et al. (2012) Frailty and sarcopenia: Definitions and outcome parameters. Osteoporos Int 23:1839-1848.
  29. Fielding RA, Vellas B, Evans WJ, Bhasin S, Morley JE, et al. (2011) Sarcopenia: An undiagnosed condition in older adults. Current consensus definition: prevalence, etiology, and consequences. International working group on sarcopenia. J Am Med Dir Assoc 12: 249-256.
  30. Newman AB, Kupelian V, Visser M, Simonsick E, Goodpaster B, et al. (2003) Sarcopenia: Alternative definitions and associations with lower extremity function. J Am Geriatr Soc 51: 1602-1609.
  31. Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, et al. (2010) Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing 39: 412-423.
  32. Gale CR, Martyn CN, Cooper C, Sayer AA (2007) Grip strength, body composition, and mortality. Int J Epidemiol 36: 228-235.
  33. Langen RC, Schols AM, Kelders MC, van der Velden JL, Wouters EF et al. (2006) Muscle wasting and impaired muscle regeneration in a murine model of chronic pulmonary inflammation. Am J Respir Cell Mol Biol 35: 689-696.
  34. Tang K, Murano G, Wagner H, Nogueira L, Wagner PD, et al. (1985) Impaired exercise capacity and skeletal muscle function in a mouse model of pulmonary inflammation. J Appl Physiol 114:1340-1350.
  35. Mittal A, Bhatnagar S, Kumar A, Lach-Trifilieff E, Wauters S, et al. (2010) The TWEAK-Fn14 system is a critical regulator of denervation-induced skeletal muscle atrophy in mice. J Cell Biol 188: 833-849.
  36. Bhatnagar S, Kumar A (2012) The TWEAK-Fn14 system: breaking the silence of cytokine-induced skeletal muscle wasting. Curr Mol Med 12: 3-13.
  37. Yaron A, Hatzubai A, Davis M, Lavon I, Amit S, et al. (1988) Identification of the receptor component of the IkappaBalpha-ubiquitin ligase. Nature 396: 590-594.
  38. Cai D, Frantz JD, Tawa NE Jr, Melendez PA, Oh BC, JR et al. (2004) IKKbeta/NF-kappaB activation causes severe muscle wasting in mice. Cell 119: 285-298.
  39. Bodine SC, Baehr LM (2014) Skeletal muscle atrophy and the E3 ubiquitin ligases MuRF1 and MAFbx/atrogin-1. Am J Physiol Endocrinol Metab 307: E469-E484.
  40. Kobayashi T, Walsh PT, Walsh MC, Speirs KM, Chiffoleau E, et al., (2003) TRAF6 is a critical factor for dendritic cell maturation and development. Immunity 19: 353-363.
  41. Paul PK, Gupta SK, Bhatnagar S, Panguluri SK, Darnay BG, et al. (2010) Targeted ablation of TRAF6 inhibits skeletal muscle wasting in mice. J Cell Biol 191: 1395-1411.
  42. Wang X, Wang J, Wang L, Wang HM, Guan YF, et al. (2012) Construction of the skeletal muscle-specific TbetaR II knockout mice. Zhongguo Ying Yong Sheng Li Xue Za Zhi 28: 284-287.
  43. Kobayashi T, Walsh PT, Walsh MC, Speirs KM, Chiffoleau E, et al. (2003) Changes in the expression of smooth muscle contractile proteins in TNBS- and DSS-induced colitis in mice. Immunity 19: 353-363.
  44. Frances P, Katia M, Maithili N, Leo F, Margaret LS, et al. (2010) Skeletal muscle catabolism in trinitrobenzene sulfonic acid-induced murine colitis. Metabolism 59:1680-1690.
  45. Liu JL, Yakar S, LeRoith D (2000) Conditional knockout of mouse insulin-like growth factor-1 gene using the Cre/loxP system. Proc Soc Exp Biol Med 223: 344-351.
  46. O'Neill BT, Lauritzen HP, Hirshman MF, Smyth G, Goodyear LJ et al. (2015) Differential role of Insulin/IGF-1 receptor signaling in muscle growth and glucose homeostasis. Cell Rep 11: 1220-1235.
  47. Athanassia S, Mickaël O, Cécile K, Ram KM, John JK, et al. (2006) Growth hormone promotes skeletal muscle cell fusion independent of insulin-like growth factor 1 up-regulation. Proc Natl Acad Sci USA 103: 7315-7320.
  48. Saul D, Kling JH, Kosinsky RL, Hoffmann DB, Komrakova M, et al. (2016) Effect of the Lipoxygenase Inhibitor Baicalein on Muscles in Ovariectomized Rats. J Nutr Metab 2016: 3703216.
  49. Fonseca H, Powers SK, Gonçalves D, Santos A, Mota MP, et al. (2012) Physical inactivity is a major contributor to ovariectomy-induced sarcopenia. Int J Sports Med 33: 268-278.
  50. Delafontaine P, Yoshida T (2016) The renin-angiotensin system and the biology of skeletal muscle: mechanisms of muscle wasting in chronic disease states. Trans Am Clin Climatol Assoc 127: 245-258.
  51. Yoshida T, Tabony AM, Galvez S, Mitch WE, Higashi Y, et al. (2013) Molecular mechanisms and signaling pathways of angiotensin II-induced muscle wasting: potential therapeutic targets for cardiac cachexia. Int J Biochem Cell Biol 45: 2322-2332.
  52. Lawrence T (2009) The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect Biol 1: a001651.
  53. Visser M, Pahor M, Taaffe DR, Goodpaster BH, Simonsick EM, et al. (2002) Relationship of interleukin-6 and tumor necrosis factor-alpha with muscle mass and muscle strength in elderly men and women: The Health ABC Study. J Gerontol A Biol Sci Med Sci 57: M326-M332.
  54. Pollack M, Phaneuf S, Dirks A, Leeuwenburgh C (2002) The role of apoptosis in the normal aging brain, skeletal muscle, and heart. Ann N Y Acad Sci 959: 93-107.
  55. Dirks-Naylor AJ, Lennon-Edwards S (2011) Cellular and molecular mechanisms of apoptosis in age-related muscle atrophy. Curr Aging Sci 4: 269-278.
  56. Schreiber S, Nikolaus S, Hampe J (1998) Activation of nuclear factor kappa B inflammatory bowel disease. Gut 42: 477-484.
  57. Schneider SM, Al-Jaouni R, Filippi J, Wiroth JB, Zeanandin G, et al. (2008) Sarcopenia is prevalent in patients with Crohn's disease in clinical remission. Inflamm Bowel Dis 14:1562-1568.
  58. Subramaniam K, Fallon K, Ruut T, Lane D, McKay R, et al. (2015) Infliximab reverses inflammatory muscle wasting (sarcopenia) in Crohn's disease. Aliment Pharmacol Ther 41: 419-428.
  59. Fermoselle C, Rabinovich R, Ausín P, Puig-Vilanova E, Coronell C, et al. (2012) Does oxidative stress modulate limb muscle atrophy in severe COPD patients? Eur Respir J 40: 851-862.
  60. Joppa P, Tkacova R, Franssen FM, Hanson C, Rennard SI, et al. (2016) Sarcopenic obesity, functional outcomes, and systemic inflammation in patients with chronic obstructive pulmonary disease. J Am Med Dir Assoc 17: 712-718.
  61. Clarke BA, Drujan D, Willis MS, Murphy LO, Corpina RA, et al. (2007) The E3 Ligase MuRF1 degrades myosin heavy chain protein in dexamethasone-treated skeletal muscle. Cell Metab 6: 376-385.
  62. Shenhav C, Jeffrey JB, Steven PG, David JG, David MV, et al. (2009) During muscle atrophy, thick, but not thin, filament components are degraded by MuRF1-dependent ubiquitylation. JCB 185: 1083-1095.
  63. Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, et al. (2004) Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 117: 399-412.
  64. Elkina Y, von Haehling S, Anker SD, Springer J (2011) The role of myostatin in muscle wasting: an overview. J Cachexia Sarcopenia Muscle 2: 143-151.
  65. Doucet M, Russell AP, Léger B, Debigaré R, Joanisse DR, et al. (2007) Muscle atrophy and hypertrophy signaling in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med 176: 261-269.
  66. Petersen AM, Magkos F, Atherton P, Selby A, Smith K, et al. (2007) Smoking impairs muscle protein synthesis and increases the expression of myostatin and MAFbx in muscle Am J Physiol Endocrinol Metab 293: E843-E848.
  67. Bodine SC, Latres E, Baumhueter S, Lai VK, Nunez L, et al. (2001) Identification of ubiquitin ligases required for skeletal muscle atrophy. Science 294: 1704-1708.
  68. Sacheck JM, Ohtsuka A, McLary SC, Goldberg AL (2004) IGF-I stimulates muscle growth by suppressing protein breakdown and expression of atrophy-related ubiquitin ligases, atrogin-1 and MuRF1. Am J Physiol Endocrinol Metab 287: E591-601.
  69. Sun H, Gong Y, Qiu J, Chen Y, Ding F, et al. (2014) TRAF6 inhibition rescues dexamethasone-induced muscle atrophy. Int J Mol Sci 15: 11126-11141.
  70. Dickson KM, Bhakar AL, Barker PA (2004) TRAF6-dependent NF-kB transcriptional activity during mouse development. Dev Dyn 231: 122-127.
  71. Yoon K, Jung EJ, Lee SY (2008) TRAF6-mediated regulation of the PI3 kinase (PI3K)-Akt-GSK3beta cascade is required for TNF-induced cell survival. Biochem Biophys Res Commun 371:118-121
  72. Hindi SM, Kumar A (2016) TRAF6 regulates satellite stem cell self-renewal and function during regenerative myogenesis. J Clin Invest 126: 151-168.
  73. Vlantis K, Polykratis A, Welz PS, van Loo G, Pasparakis M, et al. (2016) TLR-independent anti-inflammatory function of intestinal epithelial TRAF6 signalling prevents DSS-induced colitis in mice. Gut 65: 935-943.
  74. Junnila RK, List EO, Berryman DE, Murrey JW, Kopchick JJ (2013) The GH/IGF-1 axis in ageing and longevity. Nat Rev Endocrinol 9: 366-376.
  75. Chahal HS, Drake WM (2007) The endocrine system and ageing. J Pathol 211: 173-180.
  76. Sculthorpe N, Solomon AM, Sinanan AC, Bouloux PM, Grace F, et al. (2012) Androgens affect myogenesis in vitro and increase local IGF-1 expression. Med Sci Sports Exerc 44: 610-615.
  77. Sattler FR, Castaneda-Sceppa C, Binder EF, Schroeder ET, Wang Y, et al. (2009) Testosterone and growth hormone improve body composition and muscle performance in older men. J Clin Endocrinol Metab 94: 1991-2001.
  78. Ho KK, Weissberger AJ (1992) Impact of short-term estrogen administration on growth hormone secretion and action: Distinct route-dependent effects on connective and bone tissue metabolism. J Bone Miner Res 7: 821-827.
  79. Kathleen MF, John MB, Shravanthi RG, Richard M, Chiun-FC (2009) Estimation of Cachexia among Cancer Patients Based on Four Definitions. Journal of oncology 2009: 7.
  80. Mazzola P, Radhi S, Mirandola L, Annoni G, Jenkins M, et al. (2012) Aging, cancer, and cancer vaccines. Immun Ageing 9: 4.
  81. Op den Kamp CM, Langen RC, Snepvangers FJ, de Theije CC, Schellekens JM, et al. (2013) Nuclear transcription factor kappa B activation and protein turnover adaptations in skeletal muscle of patients with progressive stages of lung cancer cachexia. Am J Clin Nutr 98: 738-748.
  82. Puppa MJ, Gao S, Narsale AA, Carson JA (2014) Skeletal muscle glycoprotein 130's role in Lewis lung carcinoma-induced cachexia. FASEB J 28: 998-1009.
  83. Lokireddy S, Wijesoma IW, Bonala S, Wei M, Sze SK (2012) Myostatin is a novel tumoral factor that induces cancer cachexia. Biochem J 446: 23-36.
  84. Whittemore LA, Song K, Li X, Aghajanian J, Davies M, et al. (2003) Inhibition of myostatin in adult mice increases skeletal muscle mass and strength. Biochem Biophys Res Commun 300: 965-971.
  85. Cruz-Jentoft AJ, Landi F, Schneider SM, Zúñiga C, Arai H, et al. (2014) Prevalence of and interventions for sarcopenia in ageing adults: A systematic review. Report of the international sarcopenia initiative (EWGSOP and IWGS). Age Ageing 43: 748-759.
  86. Bokshan SL, DePasse JM, Daniels AH (2016) Sarcopenia in orthopedic surgery. Orthopedics 39: e295-300.
  87. Morley JE (2016) Pharmacologic options for the treatment of sarcopenia. Calcif Tissue Int 98: 319-333.
  88. Li TH, Lee PC, Lee KC, Hsieh YC, Tsai CY, et al. (2016) Down-regulation of common NFkappaB-iNOS pathway by chronic thalidomide treatment improves hepatopulmonary syndrome and muscle wasting in rats with biliary cirrhosis. Sci Rep 6: 39405.
  89. Reid J, Mills M, Cantwell M, Cardwell CR, Murray LJ, et al. (2012) Thalidomide for managing cancer cachexia. Cochrane Database Syst Rev: CD008664.
  90. Zaki MH, Nemeth JA, Trikha (2004) M CNTO 328, a monoclonal antibody to IL-6, inhibits human tumor-induced cachexia in nude mice. Int J Cancer 111: 592-595.
  91. Trappe TA, Liu SZ (1985) Effects of prostaglandins and COX-inhibiting drugs on skeletal muscle adaptations to exercise. J Appl Physiol 115: 909-919.
  92. Woo KJ, Lim JH, Suh SI, Kwon YK, Shin SW, et al. (2006) Differential inhibitory effects of baicalein and baicalin on LPS-induced cyclooxygenase-2 expression through inhibition of C/EBPbeta DNA-binding activity. Immunobiology 211: 359-368.
  93. Giannoulis MG, Martin FC, Nair KS, Umpleby AM, Sonksen P (2012) Hormone replacement therapy and physical function in healthy older men. Time to talk hormones? Endocr Rev 33: 314-77.
  94. Amitani M, Asakawa A, Amitani H, Inui A (2013) Control of food intake and muscle wasting in cachexia. Int J Biochem Cell Biol 45: 2179-2185.
  95. Reano S, Graziani A, Filigheddu N (2014) Acylated and unacylated ghrelin administration to blunt muscle wasting. Curr Opin Clin Nutr Metab Care 17: 236-240.
  96. Muir SW, Montero-Odasso M (2011) Effect of vitamin D supplementation on muscle strength, gait and balance in older adults: A systematic review and meta-analysis. J Am Geriatr Soc 59: 2291-300.
  97. Stockton KA, Mengersen K, Paratz JD, Kandiah D, Bennell KL et al. (2011) Effect of vitamin D supplementation on muscle strength: A systematic review and meta-analysis. Osteoporos Int 22: 859-871.
  98. Ceglia L, Niramitmahapanya S, da Silva Morais M, Rivas DA, Harris SS, et al. (2013) A randomized study on the effect of vitamin D3 supplementation on skeletal muscle morphology and vitamin D receptor concentration in older women. J Clin Endocrinol Metab 98: E1927-E1935.
  99. Arounleut P, Bialek P, Liang LF, Upadhyay S, Fulzele S, et al. (2013) A myostatin inhibitor (propeptide-Fc) increases muscle mass and muscle fiber size in aged mice but does not increase bone density or bone strength. Exp Gerontol 48: 898-904.
  100. Gilson H, Schakman O, Kalista S, Lause P, Tsuchida K, et al. (2009) Follistatin induces muscle hypertrophy through satellite cell proliferation and inhibition of both myostatin and activin. Am J Physiol Endocrinol Metab 297: E157-164.
  101. Zhu J, Li Y, Lu A, Gharaibeh B, Ma J, et al. (2011) Follistatin improves skeletal muscle healing after injury and disease through an interaction with muscle regeneration, angiogenesis and fibrosis. Am J Pathol 179: 915-930.
  102. Ohsawa Y, Hagiwara H, Nakatani M, Yasue A, Moriyama K, et al. (2006) Muscular atrophy of caveolin-3-deficient mice is rescued by myostatin inhibition. J Clin Invest 116: 2924-2934.
  103. Kawakami E, Kinouchi N, Adachi T, Ohsawa Y, Ishimaru N, et al. (2011) Atelocollagen-mediated systemic administration of myostatin-targeting siRNA improves muscular atrophy in caveolin-3-deficient mice. Dev Growth Differ 53: 48-54.
Citation: Saul D, Schilling AF, Kosinsky RL (2017) Why Age Matters: Inflammation, Cancer and Hormones in the Development of Sarcopenia. J Osteopor Phys Act 5:191.

Copyright: © 2017 Saul D, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top