Osteoporosis/Sarcopenia in Chronic Liver Disease: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor:

The prevalence of osteoporosis and sarcopenia is significantly higher in patients with liver disease than in those without liver disease and osteoporosis and sarcopenia negatively influence morbidity and mortality in liver disease.

  • osteoporosis
  • sarcopenia
  • osteosarcopenia
  • chronic liver disease

1. Introduction

Osteoporosis is characterized by low bone mass, the deterioration of bone macro-and micro-architecture, and is a common complication observed in patients with chronic liver disease [1]. The prevalence of osteoporosis in chronic liver disease patients is 10–40%, which is higher than in the general population without liver disease [1]. The presence of osteoporosis in patients with liver disease adversely affects their clinical outcomes in terms of quality of life, survival, and liver-related complications, regardless of etiology and severity [1][2]. Since sarcopenia was first proposed as the concept of age-related loss of muscle mass in 1989, numerous studies have demonstrated its molecular pathogenesis and clinical implications, especially in the context of chronic liver disease, as the liver is an important organ for carbohydrate, protein, and lipid metabolism, whose deterioration results in protein supply dysregulation and hyperammonemia, inevitably influencing skeletal muscle homeostasis [3][4][5][6][7][8][9]. Therefore, sarcopenia was observed almost half of patients with liver cirrhosis, and negatively influenced the mortality and prognosis of liver disease [3][9]. Recent evidence of the interaction between osteoporosis and sarcopenia has led to the concept of osteosarcopenia, describing the concomitant development of sarcopenia and osteoporosis [10][11]. As common musculoskeletal complications that develop with aging, sarcopenia and osteoporosis share genetic, endocrine, and mechanical risk factors, and are also closely connected both mechanically and metabolically [12][13][14][15]. Osteosarcopenia has a negative effect on the quality of life and the clinical outcome in the events of falls, disability, hospitalization, and fracture, thus contributing to a higher mortality, which has highlighted its importance as a global health concern [16][17]. However, despite the high prevalence and clinical significance of osteoporosis and sarcopenia in patients with liver disease, attention and management strategies for these musculoskeletal disorders are frequently overlooked in clinical practice for patients with liver disease. Additionally, current understanding of molecular mechanism of osteosarcopenia in terms of bone and muscle crosstalk in patients with chronic liver disease is limited.

2. Osteoporosis in Chronic Liver Disease

2.1. Prevalence and Clinical Outcomes of Osteoporosis in Chronic Liver Disease

A number of studies on osteoporosis in chronic liver disease have focused on cholestatic liver disease, including primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC), and end-stage liver cirrhosis [18]. The prevalence of osteoporosis is reported to be 20–32% in PBC and 15% in PSC, with the severity of liver disease being a risk factor for in PBC patients [19][20][21]. About 10–20% patients with PBC experienced facture, and the risk of fracture in this patient group was 2-fold higher than in the general population [22][23]. Patient with non-cirrhotic chronic hepatitis B or C exhibited lower bone mineral density (BMD) and developed osteoporosis with a prevalence of 10–30% [24][25][26][27][28][29]. Hansen et al. reported that fracture risk was higher in HCV-exposed patients [29]. In those with liver cirrhosis, the prevalence of osteoporosis was 12–28%, with advanced-stage cirrhosis and alcohol-associated liver cirrhosis being significantly associated with osteoporosis [30][31][32][33]. Interestingly, liver transplantation improved BMD, especially in patients receiving less glucocorticoid treatment, without cholestasis, and exhibiting an elevation of vitamin D and parathyroid hormones 4–6 months after liver transplantation [34][35]. With regard to non-cholestatic liver disease, a retrospective study reported that osteoporotic fractures were 2.5-fold more common in patients with non-alcoholic fatty liver disease (NAFLD), and low BMD was more prominent in those with higher disease activity, such as in cases with non-alcoholic steatohepatitis (NASH), significant fibrosis, and a high fatty liver index [36][37][38][39]. As alcohol consumption is an independent risk factor for osteoporosis, patients with considerable alcohol consumption developed osteoporosis without liver cirrhosis [40]. About 30% and 36% of alcoholic patients showed osteoporosis and vertebral fracture upon radiologic examination, respectively [41]. Importantly, abstinence increased BMD and bone formation marker osteocalcin levels in alcoholics [42]. Osteoporosis was observed in 25–34% of patients with hereditary hemochromatosis, independent of cirrhosis or hypogonadism [43][44][45]. The prevalence of osteopenia and osteoporosis was 9% and 50%, respectively, in patients with Wilson’s disease, which was significantly higher than in the healthy population [46]. In addition, Quemeneur et al. reported that half of patients with Wilson’s disease suffered peripheral fractures [47].

2.2. Molecular Mechanism of Osteoporosis in Chronic Liver Disease

The pathophysiology of chronic liver disease-associated osteoporosis is complex. Increased bone resorption is an important pathological characteristic of osteoporosis, especially in patients with end-stage cholestatic liver disease, viral hepatitis, and NAFLD. Suggested molecular mechanisms underlying bone resorption in chronic liver disease include the receptor activator of nuclear factor kappa (RANK)-RANK ligand (RANKL)-osteoprotegerin (OPG) system, upregulation of proinflammatory cytokines, such as interleukin-1 (IL-1), IL-6, and tumor necrosis factor alpha (TNF-α), as well as low levels of testosterone [1][2][48][49]. Bone remodeling is tightly regulated by osteocytes and osteoblasts through different cytokines and hormones modulating the activation, resorption, reversal, formation, and termination phase [50]. Various types of signals are involved, including mechanical strain, bone damage, as well as hormone-induced bone remodeling via macrophage colony-stimulating factor (M-CSF), RANKL, and OPG secreted by osteoblasts. RANKL and OPG are members of the TNF superfamily and are crucial for the regulation of bone resorption [51]. RANKL, through its receptor RANK, activates osteoclast formation, activation, and survival, while OPG, which is another receptor for RANKL, restrains osteoclastogenesis and inhibits bone loss by binding RANKL to prevent the RANK-RANKL cascade [51]. RANKL is a type II transmembrane protein with a C-terminal extracellular domain. This ectodomain cleaved is cleaved by matrix metalloproteinases to yield soluble RANKL (sRANKL) in the extracellular environment and both membrane-bound and sRANKL bind to RANK [52]. In chronic liver disease, an imbalance between RANKL and OPG leads to high sRANKL levels, which increases bone turnover. Thus, the ratio of OPG/sRANKL might indicate a homeostatic response for bone mass preservation [53]. Moschen et al. reported that sRANKL levels were higher in patients with liver disease than those in controls, except those in the cirrhotic subgroup, while OPG levels were found to be proportional to the severity of liver disease and highest in the cirrhotic subgroup with osteoporosis and osteopenia, resulting in a greater OPG/sRNAKL ratio in the cirrhotic subgroup with osteoporosis and osteopenia than in cirrhotic patients with normal BMD. These results suggested that high sRNAKL levels corresponded to increased bone turnover in patients with liver disease, and that OPG was also increased to compensate for negative bone turnover. Therefore, the high OPG/sRANKL ratio could be explained a response to maintain bone homeostasis in these patients [54]. In addition, RANKL/OPG gene expression, indicative of osteoblast-related osteoclastogenesis, was increased in the serum of jaundice patients [55].

In a chronic inflammation state, proinflammatory cytokines, especially IL-6, IL-1, and TNF-α, contribute to osteoclast activation and subsequent bone resorption [56]. IL-6 and IL-1 directly modulated osteoclastogenesis by enhancing osteoclast function [57]. They was also shown to indirectly promote osteoclast activity by facilitating RANKL production in osteoblasts [56][57][58][59]. Experimental studies revealed that IL-6 was associated with disrupted osteogenesis of bone marrow stem cells in osteoporosis models, and suppression of the IL-6 receptor prevented osteoclast-mediated bone resorption [60][61]. As IL-6 increases during liver injury to stimulate liver regeneration, upregulated IL-6 could affect bone remodeling in various types of liver disease [62][63]. Additionally, ethanol seems to activate osteoclasts through the induction of IL-6 and TNF-α [64][65]. Another potent inflammatory cytokine, TNF-α, is also involved in inflammatory bone resorption by stimulating RANKL expression in osteoblasts and tissue stromal cells, in turn promoting osteoclast differentiation and activity [66]. In particular, TNF-α enhanced CSF-1 receptor gene expression during the initial stage of osteoclastogenesis and subsequently stimulated osteoblast precursors, which resulted in increased osteoclast formation independent of the RANKL pathway [67]. These proinflammatory cytokines had effects on osteoporosis in viral hepatitis and NASH. Gonzalez-Calvin et al. showed that serum soluble TNF receptor p55 levels were significantly higher in patients with viral cirrhosis with osteoporosis than those without osteoporosis and positively associated with bone resorption [68]. In addition, since obesity is considered a chronic inflammatory state, these proinflammatory cytokines contribute to osteoporosis in NAFLD [69]. Indeed, low BMD is significantly associated with NASH presenting elevated alanine aminotransferase (ALT) and C-reactive protein (CRP) than in simple steatosis [37]. Another study showed that TNF-α levels are elevated in pediatric NASH [70]. Furthermore, Kim et al. reported that liver fibrosis in NAFLD is significantly correlated with low BMD, suggesting an association between aggravation of hepatic inflammation, fibrosis, and bone loss in NAFLD patients [38]. These proinflammatory cytokines have also been proposed as involved in dysbiosis-induced osteoporosis associated with chronic liver disease. Altered short-chain fatty acid (SCFA) levels and increased gut permeability (“leaky-gut syndrome”) may affect bone remodeling by regulating inflammation and immune system [2]. In addition, impaired liver function and cholestasis result in decreased 25-hydroxylation and intestinal absorption of vitamin D. Vitamin D deficiency is associated with osteoporosis in patients with liver cirrhosis [30][33]. Calcium and vitamin D deficiencies in patients with cholestatic liver disease caused secondary hyperparathyroidism, subsequently enhancing bone resorption [71]. Vitamin K is known to be involved in osteoblast apoptosis inhibition and osteoclast differentiation [72][73][74]. Therefore, decreased vitamin K levels were suggested to affect bone metabolism in chronic liver disease. Vitamin K deficiency reduced bone matrix proteins such as osteocalcin and osteonectin in patients with PBC [75][76].

Bone formation is also compromised in chronic liver disease as a result of toxic materials, sclerostin, and decreased anabolic hormones, which contribute to osteoporosis in patients with PBC, advanced stage liver cirrhosis, hereditary hemochromatosis, and Wilson disease [1][2][48]. Direct or indirect toxic compounds such as bilirubin, alcohol, iron, and copper accumulate in specific liver diseases, which can impair bone formation by inhibiting osteoblast proliferation and differentiation as well as bone mineralization by osteoblasts [45][55]. Unconjugated bilirubin decreased the survival of osteoblast, and osteoblast differentiation was significantly reduced only in jaundiced patients, except in patients with normal bilirubin levels, whereas ursodeoxycholic acid compensated for the negative effect of cholestatsis on osteoblast survival, proliferation and mineralization [55][77]. Previous studies reported that the severity of liver disease including cholestasis is significantly associated with osteoporosis in patients with PBC [19][20][23]. In hereditary hemochromatosis, lumbar spine BMD is significantly decreased in parallel to an increase of iron and alkaline phosphatase (ALP) levels [45]. In addition, bone synthesis is lower in alcoholic patients with low levels of osteocalcin, which is secreted from osteoblasts, and plays a role in calcium homeostasis, bone matrix mineralization, and osteoblastic proliferation [53]. Under physiological conditions, after bone resorption, mesenchymal stem cells and early osteoblast progenitors differentiate into osteoblasts through Wnt, bone morphogenetic protein (BMP), and fibroblast growth factor (FGF) signaling, leading to bone formation [2][78]. As a regulator of bone formation, sclerostin is produced in osteocytes and hinders osteoblast differentiation and proliferation, subsequently restricting bone formation by antagonizing Wnt signaling via binding to low-density lipoprotein receptor-related proteins (LRP) 5/6 transmembrane receptors [79][80]. As a result of sclerostin expression, the Wnt receptor is blocked and glycogen synthase kinase 3 phosphorylates β-catenin, which is involved in ubiquitination and degradation through the proteasome pathway [81]. Guanabens et al. proposed sclerostin as a crucial regulator of the Wnt/β-catenin pathway in relation to bone formation in patients with PBC [82]. A cross-sectional study revealed that sclerostin levels were significantly increased in patients with advanced cirrhosis when compared to those with early cirrhosis or healthy controls [83].

Insulin-like growth factor (IGF-1), which is secreted from hepatocytes by growth hormone (GH) has an anabolic effect on bone growth by suppressing osteoblast apoptosis and enhancing osteoblastogenesis through stabilization of the Wnt/β-catenin pathway [84][85]. In addition, IGF-1 reduced bone resorption through the OPG and RANKL system [86]. In end-stage liver disease, hepatocellular dysfunction and reduced GH receptors lead to low serum IGF-1 levels, subsequently causing osteoporosis [87]. Insulin, an important hormone associated with NAFLD, also affects bone remodeling by activating collagen synthesis and stimulating osteoblast proliferation and differentiation [88]. In addition, hypogonadism, which results from hyperestrogenism of portal hypertension in males and suppression of the hypothalamic-pituitary-gonadal axis in females, is frequently observed in hemochromatosis, liver cirrhosis, and alcoholics liver disease [1][89]. Since testosterone directly modulates osteoblasts and osteocytes via the androgen receptor to stimulate trabecular bone formation and prevent its loss, low testosterone level owing to hypogonadism in male enhances osteoclast function and induces bone turnover [53][90]. Though estrogen levels are increased in patient with liver cirrhosis due to increased peripheral conversion of androgen to estrogen, altered estrogen metabolism in liver cirrhosis contributes to a decrease in degradation of estrogen metabolites [91]. Because the bone protective effect of these estrogens is weak, it is not enough to overcome post-menopausal osteoporosis in women and liver-disease related osteoporosis in men [92][93].

3. Sarcopenia in Chronic Liver Disease

3.1. Definition of Sarcopenia

Sarcopenia, a condition characterized by the loss of skeletal muscle mass and strength, has been explored as a prognostic predictor for various diseases [94]. However, consensus criteria for the diagnosis of sarcopenia have not yet been established, and different definitions have been proposed by several groups [95][96]. The European Working Group on Sarcopenia in Older People (EWGSOP) first proposed the definition of sarcopenia in 2010, with muscle mass being a cardinal requirement for sarcopenia diagnosis [97]. The working group categorized sarcopenia into two categories. Primary sarcopenia referred to age-related muscle mass deterioration, while secondary sarcopenia was defined as caused by factors other than aging, such as inflammatory processes, disease-related disabilities, and inadequate energy or protein intake. Given the growing body of scientific data on sarcopenia, the EWGSOP recently updated diagnostic criteria so that muscle strength is the principal determinant for diagnosis with strict cut-off values (hand grip strength <27 kg for men and <16 kg for women) due to its significant correlation with clinical outcomes when compared to muscle mass [95]. According to the revised criteria, sarcopenia is defined as low muscle strength in parallel to low muscle mass or decreased muscle function. In contrast, the Asian Working Group for Sarcopenia (AWGS) regarded low muscle mass as the cardinal criterion for the diagnosis of sarcopenia. Therefore, AWGS defined sarcopenia as low muscle mass with low muscle strength or low physical performance [96]. Further, both EWGSOP2 and AWGS recognize entirely impaired muscle mass, muscle strength, and muscle function as diagnostic criteria for severe sarcopenia [95][96]. On the other hand, Clark et al. proposed in 2008 the concept of dynapenia, a state of age-associated decline in muscle strength which focused on other physiologic factors except for muscle mass loss in 2008 [98]. Therefore, dynapenia is similar to the revised EWGSOP definition of sarcoepnia in that decrease of skeletal muscle mass was not always necessary for the diagnosis of dynapenia, but sarcopenia is recognized to develop earlier in life, unlike dynapenia.

3.2. Prevalence and Clinical Outcomes of Sarcopenia in Chronic Liver Disease

Sarcopenia results from the accelerated loss of muscle mass and function, which in turn contributes to adverse clinical outcomes including fracture, frailty, and mortality. Although the prevalence of sarcopenia depends on the definition used, which is based on, for example, muscle mass cutoff points as well as other factors, the condition has been rigorously studied in the context of various chronic liver diseases [94]. With regard to non-alcoholic liver disease, previous studies reported a significant relationship between sarcopenia, sarcopenic obesity, and NAFLD, with this association being later described as independent of obesity and insulin resistance [99][100][101][102][103].

In addition, a longitudinal cohort study revealed that increased skeletal mass during the study period had a beneficial effect against the development of NAFLD or improved existing NAFLD at baseline [104]. Muscle mass loss was observed in 40–70% of liver cirrhosis patients, and most studies reported negative influences of sarcopenia on mortality and liver cirrhosis complications, such as hepatic encephalopathy [105][106][107][108][109][110][111]. The degree of sarcopenia was associated with Child-Pugh score in patients with cirrhosis, and sarcopenia was a predictor of survival independently or in combination with the model for end-stage liver disease (MELD) score, especially in patients with low scores (<15) [112][113][114]. Unfortunately, after liver transplantation, sarcopenia did not improve and even worsened because of immunosuppressive drugs such as steroids, calcineurin inhibitors, and mammalian target of rapamycin (mTOR) inhibitors [6]. Sarcopenia also had significant impacts on the development of diabetes mellitus, the risk of infection, the length of hospitalization, and mortality in patients who underwent liver transplantation [115][116][117][118][119][120][121][122]. Further, sarcopenia was shown to independently predict mortality, overall survival, and recurrence-free survival in patients with hepatocellular carcinoma [123][124][125].

3.3. Molecular Mechanism of Sarcopenia in Chronic Liver Disease

In normal physiology, skeletal muscle protein turnover is maintained as a balance between protein synthesis and breakdown. mTOR is an essential modulator of translational control, majorly involved in protein synthesis [126]. Exercise, branched chain amino acids (BCAAs), as well as various hormones, including testosterone, insulin, and IGF-1, activate the mTOR pathway in muscle cells through protein kinase B, which subsequently triggers several intracellular pathways for muscle protein synthesis, including p70 ribosomal S6 kinase 1 (S6K1) and eukaryotic initiation factor 4E binding protein (4E-BP1) [126]. In addition, the proliferation of muscle satellite cells, which are muscle fiber precursors, is critical for muscle growth and is activated in response to IGF-1 and BCAAs through protein kinase B [126][127].

In contrast, myostatin, a transforming growth factor beta (TGF-β) superfamily member produced in muscle, inhibits protein synthesis by maintaining satellite cells in a quiescent state, activating SMAD family transcription factors 2 and 3 (SMAD 2/3), and stimulating proteolysis via forkhead box O transcription factors (FoxOs) associated with the ubiquitin-proteasome pathway (UPP) and autophagy [128][129]. Impaired mitochondrial function, insulin resistance, and reactive oxygen species (ROS) also stimulate autophagy.

This entry is adapted from the peer-reviewed paper 10.3390/ijms22052604

References

  1. Guañabens, N.; Parés, A. Osteoporosis in chronic liver disease. Liver Int. 2018, 38, 776–785.
  2. Jeong, H.M.; Kim, D.J. Bone Diseases in Patients with Chronic Liver Disease. Int. J. Mol. Sci. 2019, 20, 4270.
  3. Ebadi, M.; Bhanji, R.A.; Mazurak, V.C.; Montano-Loza, A.J. Sarcopenia in cirrhosis: From pathogenesis to interventions. J. Gastroenterol. 2019, 54, 845–859.
  4. Meyer, F.; Bannert, K.; Wiese, M.; Esau, S.; Sautter, L.F.; Ehlers, L.; Aghdassi, A.A.; Metges, C.C.; Garbe, L.-A.; Jaster, R.; et al. Molecular Mechanism Contributing to Malnutrition and Sarcopenia in Patients with Liver Cirrhosis. Int. J. Mol. Sci. 2020, 21, 5357.
  5. Fernández-Mincone, T.; Contreras-Briceño, F.; Espinosa-Ramírez, M.; García-Valdés, P.; López-Fuenzalida, A.; Riquelme, A.; Arab, J.P.; Cabrera, D.; Arrese, M.; Barrera, F. Nonalcoholic fatty liver disease and sarcopenia: Pathophysiological connections and therapeutic implications. Expert Rev. Gastroenterol. Hepatol. 2020, 14, 1–17.
  6. Jindal, A.; Jagdish, R.K. Sarcopenia: Ammonia metabolism and hepatic encephalopathy. Clin. Mol. Hepatol. 2019, 25, 270–279.
  7. Carey, E.J.; Lai, J.C.; Sonnenday, C.; Tapper, E.B.; Tandon, P.; Duarte-Rojo, A.; Dunn, M.A.; Tsien, C.; Kallwitz, E.R.; Ng, V.; et al. A North American Expert Opinion Statement on Sarcopenia in Liver Transplantation. Hepatology 2019, 70, 1816–1829.
  8. Hsu, C.-S.; Kao, J.-H. Sarcopenia and chronic liver diseases. Expert Rev. Gastroenterol. Hepatol. 2018, 12, 1229–1244.
  9. Dasarathy, S.; Merli, M. Sarcopenia from mechanism to diagnosis and treatment in liver disease. J. Hepatol. 2016, 65, 1232–1244.
  10. Kirk, B.; Zanker, J.; Duque, G. Osteosarcopenia: Epidemiology, diagnosis, and treatment—Facts and numbers. J. Cachex-Sarcopenia Muscle 2020, 11, 609–618.
  11. Kirk, B.; Al Saedi, A.; Duque, G. Osteosarcopenia: A case of geroscience. Aging Med. 2019, 2, 147–156.
  12. Bonewald, L. Use it or lose it to age: A review of bone and muscle communication. Bone 2019, 120, 212–218.
  13. Picca, A.; Calvani, R.; Manes-Gravina, E.; Spaziani, L.; Landi, F.; Bernabei, R.; Marzetti, E. Bone-Muscle Crosstalk: Unraveling New Therapeutic Targets for Osteoporosis. Curr. Pharm. Des. 2018, 23, 6256–6263.
  14. Hirschfeld, H.P.; Kinsella, R.; Duque, G. Osteosarcopenia: Where bone, muscle, and fat collide. Osteoporos. Int. 2017, 28, 2781–2790.
  15. Kawao, N.; Kaji, H. Interactions Between Muscle Tissues and Bone Metabolism. J. Cell. Biochem. 2015, 116, 687–695.
  16. Wang, Y.-J.; Wang, Y.; Zhan, J.-K.; Tang, Z.-Y.; He, J.-Y.; Tan, P.; Deng, H.-Q.; Huang, W.; Liu, Y.-S. Sarco-Osteoporosis: Prevalence and Association with Frailty in Chinese Community-Dwelling Older Adults. Int. J. Endocrinol. 2015, 2015, 482940.
  17. Yoo, J.-I.; Kim, H.; Ha, Y.-C.; Kwon, H.-B.; Koo, K.-H. Osteosarcopenia in Patients with Hip Fracture Is Related with High Mortality. J. Korean Med. Sci. 2018, 33, e27.
  18. Collier, J. Bone disorders in chronic liver disease. Hepatology 2007, 46, 1271–1278.
  19. Menon, K.; Angulo, P.; Weston, S.; Dickson, E.; Lindor, K.D. Bone disease in primary biliary cirrhosis: Independent indicators and rate of progression. J. Hepatol. 2001, 35, 316–323.
  20. Guañabens, N.; Parés, A.; Ros, I.; Caballería, L.; Pons, F.; Vidal, S.; Monegal, A.; Peris, P.; Rodés, J. Severity of cholestasis and advanced histological stage but not menopausal status are the major risk factors for osteoporosis in primary biliary cirrhosis. J. Hepatol. 2005, 42, 573–577.
  21. Angulo, P.; Grandison, G.A.; Fong, D.G.; Keach, J.C.; Lindor, K.D.; Bjornsson, E.; Koch, A. Bone Disease in Patients with Primary Sclerosing Cholangitis. Gastroenterology 2011, 140, 180–188.
  22. Solaymani—Dodaran, M.; Card, T.R.; Aithal, G.P.; West, J. Fracture Risk in People with Primary Biliary Cirrhosis: A Population-Based Cohort Study. Gastroenterology 2006, 131, 1752–1757.
  23. Guañabens, N.; Cerdá, D.; Monegal, A.; Pons, F.; Caballería, L.; Peris, P.; Parés, A. Low Bone Mass and Severity of Cholestasis Affect Fracture Risk in Patients with Primary Biliary Cirrhosis. Gastroenterology 2010, 138, 2348–2356.
  24. Orsini, L.G.S.; Pinheiro, M.M.; Castro, C.H.M.; Silva, A.E.B.; Szejnfeld, V.L. Bone Mineral Density Measurements, Bone Markers and Serum Vitamin D Concentrations in Men with Chronic Non-Cirrhotic Untreated Hepatitis C. PLoS ONE 2013, 8, e81652.
  25. Lai, J.C.; Shoback, D.M.; Zipperstein, J.; Lizaola, B.; Tseng, S.; Terrault, N.A. Bone Mineral Density, Bone Turnover, and Systemic Inflammation in Non-cirrhotics with Chronic Hepatitis C. Dig. Dis. Sci. 2015, 60, 1813–1819.
  26. Huang, Z.; Wei, H.; Cheng, C.; Yang, S.; Wang, J.; Liu, X. Low bone mineral density in chronic hepatitis B virus infection: A case-control study. Pak. J. Med. Sci. 2017, 33, 457–461.
  27. Wei, M.T.; Le, A.K.; Chang, M.S.; Hsu, H.; Nguyen, P.; Zhang, J.Q.; Wong, C.; Wong, C.; Cheung, R.; Nguyen, M.H. Antiviral therapy and the development of osteopenia/osteoporosis among Asians with chronic hepatitis B. J. Med. Virol. 2019, 91, 1288–1294.
  28. Schiefke, I.; Fach, A.; Wiedmann, M.; Aretin, A.V.; Schenker, E.; Borte, G.; Wiese, M.; Moessner, J. Reduced bone mineral density and altered bone turnover markers in patients with non-cirrhotic chronic hepatitis B or C infection. World J. Gastroenterol. 2005, 11, 1843–1847.
  29. Hansen, A.-B.E.; Omland, L.H.; Krarup, H.; Obel, N. Fracture risk in hepatitis C virus infected persons: Results from the DANVIR cohort study. J. Hepatol. 2014, 61, 15–21.
  30. Goubraim, R.; Kabbaj, N.; Salihoun, M.; Chaoui, Z.; Nya, M.; Amrani, N. Metabolic Bone Disease in Viral Cirrhosis: A Prospective Study. ISRN Hepatol. 2013, 2013, 276563.
  31. Sokhi, R.P.; Anantharaju, A.; Kondaveeti, R.; Creech, S.D.; Islam, K.K.; Van Thiel, D.H. Bone mineral density among cirrhotic patients awaiting liver transplantation. Liver Transplant. 2004, 10, 648–653.
  32. Zheng, J.-P.; Miao, H.-X.; Zheng, S.-W.; Liu, W.-L.; Chen, C.-Q.; Zhong, H.-B.; Li, S.-F.; Fang, Y.-P.; Sun, C.-H. Risk factors for osteoporosis in liver cirrhosis patients measured by transient elastography. Medicine 2018, 97, e10645.
  33. Monegal, A.; Navasa, M.; Guañabens, N.; Peris, P.; Pons, F.; De Osaba, M.J.M.; Rimola, A.; Rodés, J.; Muñoz-Gómez, J. Osteoporosis and Bone Mineral Metabolism Disorders in Cirrhotic Patients Referred for Orthotopic Liver Transplantation. Calcif. Tissue Int. 1997, 60, 148–154.
  34. Monegal, A.; Navasa, M.; Guañabens, N.; Peris, P.; Pons, F.; De Osaba, M.J.M.; Ordi, J.; Rimola, A.; Rodés, J.; Muñoz-Gómez, J. Bone Disease after Liver Transplantation: A Long-Term Prospective Study of Bone Mass Changes, Hormonal Status and Histomorphometric Characteristics. Osteoporos. Int. 2001, 12, 484–492.
  35. Guichelaar, M.M.J.; Kendall, R.; Malinchoc, M.; Hay, J.E. Bone mineral density before and after OLT: Long-term follow-up and predictive factors. Liver Transplant. 2006, 12, 1390–1402.
  36. Li, M.; Xu, Y.; Xu, M.; Ma, L.; Wang, T.; Liu, Y.; Dai, M.; Chen, Y.; Lu, J.; Liu, J.; et al. Association between Nonalcoholic Fatty Liver Disease (NAFLD) and Osteoporotic Fracture in Middle-Aged and Elderly Chinese. J. Clin. Endocrinol. Metab. 2012, 97, 2033–2038.
  37. Purnak, T.; Beyazit, Y.; Ozaslan, E.; Efe, C.; Hayretci, M.; Ozaslan, A.P.E. The evaluation of bone mineral density in patients with nonalcoholic fatty liver disease. Wien. Klin. Wochenschr. 2012, 124, 526–531.
  38. Kim, G.; Kim, K.J.; Rhee, Y.; Lim, S.-K. Significant liver fibrosis assessed using liver transient elastography is independently associated with low bone mineral density in patients with non-alcoholic fatty liver disease. PLoS ONE 2017, 12, e0182202.
  39. Ahn, S.H.; Seo, D.H.; Kim, S.H.; Nam, M.-S.; Hong, S. The relationship between fatty liver index and bone mineral density in Koreans: KNHANES 2010–2011. Osteoporos. Int. 2017, 29, 181–190.
  40. Malik, P.; Gasser, R.W.; Kemmler, G.; Moncayo, R.; Finkenstedt, G.; Kurz, M.; Fleischhacker, W.W. Low Bone Mineral Density and Impaired Bone Metabolism in Young Alcoholic Patients without Liver Cirrhosis: A Cross-Sectional Study. Alcohol. Clin. Exp. Res. 2009, 33, 375–381.
  41. Peris, P.; Guañabens, N.; Parés, A.; Pons, F.; Del Rio, L.; Monegal, A.; Surís, X.; Caballería, J.; Rodés, J.; Muñoz-Gómez, J. Vertebral fractures and osteopenia in chronic alcoholic patients. Calcif. Tissue Int. 1995, 57, 111–114.
  42. Peris, P.; Pares, A.; Guanabens, N.; Del Rio, L.; Pons, F.; De Osaba, M.J.M.; Monegal, A.; Caballería, J.; Rodés, J.; Muñoz-Gómez, J. Bone mass improves in alcoholics after 2 years of abstinence. J. Bone Miner. Res. 1994, 9, 1607–1612.
  43. Sinigaglia, L.; Fargion, S.; Fracanzani, A.L.; Binelli, L.; Battafarano, N.; Varenna, M.; Piperno, A.; Fiorelli, G. Bone and joint involvement in genetic hemochromatosis: Role of cirrhosis and iron overload. J. Rheumatol. 1997, 24, 1809–1813.
  44. Guggenbuhl, P.; Deugnier, Y.; Boisdet, J.F.; Rolland, Y.; Perdriger, A.; Pawlotsky, Y.; Chalès, G. Bone mineral density in men with genetic hemochromatosis and HFE gene mutation. Osteoporos. Int. 2005, 16, 1809–1814.
  45. Valenti, L.; Varenna, M.; Fracanzani, A.L.; Rossi, V.; Fargion, S.; Sinigaglia, L. Association between iron overload and osteoporosis in patients with hereditary hemochromatosis. Osteoporos. Int. 2009, 20, 549–555.
  46. Weiss, K.H.; Van De Moortele, M.; Gotthardt, D.N.; Pfeiffenberger, J.; Seessle, J.; Ullrich, E.; Gielen, E.; Borghs, H.; Adriaens, E.; Stremmel, W.; et al. Bone demineralisation in a large cohort of Wilson disease patients. J. Inherit. Metab. Dis. 2015, 38, 949–956.
  47. Quemeneur, A.-S.; Trocello, J.-M.; Ea, H.-K.; Ostertag, A.; Leyendecker, A.; Duclos-Vallee, J.-C.; De Vernejoul, M.-C.; Woimant, F.; Lioté, F. Bone status and fractures in 85 adults with Wilson’s disease. Osteoporos. Int. 2014, 25, 2573–2580.
  48. Danford, C.J.; Trivedi, H.D.; Bonder, A. Bone Health in Patients with Liver Diseases. J. Clin. Densitom. 2020, 23, 212–222.
  49. Danford, C.J.; Trivedi, H.D.; Papamichael, K.; Tapper, E.B.; Bonder, A. Osteoporosis in primary biliary cholangitis. World J. Gastroenterol. 2018, 24, 3513–3520.
  50. Raggatt, L.J.; Partridge, N.C. Cellular and Molecular Mechanisms of Bone Remodeling. J. Biol. Chem. 2010, 285, 25103–25108.
  51. Boyce, B.F.; Xing, L. Functions of RANKL/RANK/OPG in bone modeling and remodeling. Arch. Biochem. Biophys. 2008, 473, 139–146.
  52. Ono, T.; Hayashi, M.; Sasaki, F.; Nakashima, T. RANKL biology: Bone metabolism, the immune system, and beyond. Inflamm. Regen. 2020, 40, 1–16.
  53. Santos, L.A.A.; Romeiro, F.G. Diagnosis and Management of Cirrhosis-Related Osteoporosis. BioMed Res. Int. 2016, 2016, 1423462.
  54. Moschen, A.R.; Kaser, A.; Stadlmann, S.; Millonig, G.; Kaser, S.; Mühllechner, P.; Habior, A.; Graziadei, I.; Vogel, W.; Tilg, H. The RANKL/OPG system and bone mineral density in patients with chronic liver disease. J. Hepatol. 2005, 43, 973–983.
  55. Ruiz-Gaspà, S.; Martinez-Ferrer, A.; Guañabens, N.; Dubreuil, M.; Peris, P.; Enjuanes, A.; De Osaba, M.J.M.; Alvarez, L.; Monegal, A.; Combalia, A.; et al. Effects of bilirubin and sera from jaundiced patients on osteoblasts: Contribution to the development of osteoporosis in liver diseases. Hepatology 2011, 54, 2104–2113.
  56. Blaschke, M.; Koepp, R.; Cortis, J.; Komrakova, M.; Schieker, M.; Hempel, U.; Siggelkow, H. IL-6, IL-1β, and TNF-α only in combination influence the osteoporotic phenotype in Crohn’s patients via bone formation and bone resorption. Adv. Clin. Exp. Med. 2018, 27, 45–56.
  57. Lorenzo, J.; Horowitz, M.; Choi, Y. Osteoimmunology: Interactions of the Bone and Immune System. Endocr. Rev. 2008, 29, 403–440.
  58. Wu, Q.; Zhou, X.; Huang, D.; Ji, Y.; Kang, F. IL-6 Enhances Osteocyte-Mediated Osteoclastogenesis by Promoting JAK2 and RANKL Activity In Vitro. Cell. Physiol. Biochem. 2017, 41, 1360–1369.
  59. Nakchbandi, I.A.; Mitnick, M.A.; Lang, R.; Gundberg, C.; Kinder, B.; Insogna, K. Circulating Levels of Interleukin-6 Soluble Receptor Predict Rates of Bone Loss in Patients with Primary Hyperparathyroidism. J. Clin. Endocrinol. Metab. 2002, 87, 4946–4951.
  60. Li, X.; Zhou, Z.-Y.; Zhang, Y.-Y.; Yang, H.-L. IL-6 Contributes to the Defective Osteogenesis of Bone Marrow Stromal Cells from the Vertebral Body of the Glucocorticoid-Induced Osteoporotic Mouse. PLoS ONE 2016, 11, e0154677.
  61. Axmann, R.; Böhm, C.; Krönke, G.; Zwerina, J.; Smolen, J.; Schett, G. Inhibition of interleukin-6 receptor directly blocks osteoclast formation in vitro and in vivo. Arthritis Rheum. 2009, 60, 2747–2756.
  62. Norris, C.A.; He, M.; Kang, L.-I.; Ding, M.Q.; Radder, J.E.; Haynes, M.M.; Yang, Y.; Paranjpe, S.; Bowen, W.C.; Orr, A.; et al. Synthesis of IL-6 by Hepatocytes Is a Normal Response to Common Hepatic Stimuli. PLoS ONE 2014, 9, e96053.
  63. Shimada, M.; Matsumata, T.; Taketomi, A.; Shirabe, K.; Yamamoto, K.; Takenaka, K.; Sugimachi, K. The role of interleukin-6, interleukin-16, tumor necrosis factor-alpha and endotoxin in hepatic resection. Hepatogastroenterology 1995, 42, 691–697.
  64. Guarino, M.; Loperto, I.; Camera, S.; Cossiga, V.; Di Somma, C.; Colao, A.; Caporaso, N.; Morisco, F. Osteoporosis across chronic liver disease. Osteoporos. Int. 2016, 27, 1967–1977.
  65. Handzlik-Orlik, G.; Holecki, M.; Wilczyński, K.; Duława, J. Osteoporosis in liver disease: Pathogenesis and management. Ther. Adv. Endocrinol. Metab. 2016, 7, 128–135.
  66. Zhao, B.; Grimes, S.N.; Li, S.; Hu, X.; Ivashkiv, L.B. TNF-induced osteoclastogenesis and inflammatory bone resorption are inhibited by transcription factor RBP-J. J. Exp. Med. 2012, 209, 319–334.
  67. Boyce, B.E.; Li, P.; Yao, Z.; Zhang, Q.; Badell, I.R.; Schwarz, E.M.; O’Keefe, R.J.; Xing, L. TNF.ALPHA. and pathologic bone resorption. Keio J. Med. 2005, 54, 127–131.
  68. González-Calvin, J.L.; Gallego-Rojo, F.; Fernández-Pérez, R.; Casado-Caballero, F.; Ruiz-Escolano, E.; Olivares, E.G. Osteoporosis, Mineral Metabolism, and Serum Soluble Tumor Necrosis Factor Receptor p55 in Viral Cirrhosis. J. Clin. Endocrinol. Metab. 2004, 89, 4325–4330.
  69. Filip, R.; Radzki, R.P.; Bieńko, M. Novel insights into the relationship between nonalcoholic fatty liver disease and osteoporosis. Clin. Interv. Aging 2018, 13, 1879–1891.
  70. Manco, M.; Marcellini, M.; Giannone, G.; Nobili, V. Correlation of Serum TNF-α Levels and Histologic Liver Injury Scores in Pediatric Nonalcoholic Fatty Liver Disease. Am. J. Clin. Pathol. 2007, 127, 954–960.
  71. McCaughan, G.; Feller, R. Osteoporosis in Chronic Liver Disease: Pathogenesis, Risk Factors, and Management. Dig. Dis. 1994, 12, 223–231.
  72. Koshihara, Y.; Hoshi, K.; Okawara, R.; Ishibashi, H.; Yamamoto, S. Vitamin K stimulates osteoblastogenesis and inhibits osteoclastogenesis in human bone marrow cell culture. J. Endocrinol. 2003, 176, 339–348.
  73. Hiruma, Y.; Nakahama, K.-I.; Fujita, H.; Morita, I. Vitamin K2 and geranylgeraniol, its side chain component, inhibited osteoclast formation in a different manner. Biochem. Biophys. Res. Commun. 2004, 314, 24–30.
  74. Cockayne, S.; Adamson, J.; Lanham-New, S.; Shearer, M.J.; Gilbody, S.; Torgerson, D.J. Vitamin K and the prevention of fractures: Systematic review and meta-analysis of randomized controlled trials. Arch. Intern. Med. 2006, 166, 1256–1261.
  75. Kowdley, K.V.; Emond, M.J.; Sadowski, J.A.; Kaplan, M.M. Plasma vitamin K1 level is decreased in primary biliary cirrhosis. Am. J. Gastroenterol. 1997, 92, 2059–2061.
  76. Nishiguchi, S.; Shimoi, S.; Kurooka, H.; Tamori, A.; Habu, D.; Takeda, T.; Ochi, H. Randomized pilot trial of vitamin K2 for bone loss in patients with primary biliary cirrhosis. J. Hepatol. 2001, 35, 543–545.
  77. Ruiz-Gaspà, S.; Dubreuil, M.; Guañabens, N.; Combalia, A.; Peris, P.; Monegal, A.; Parés, A. Ursodeoxycholic acid decreases bilirubin-induced osteoblast apoptosis. Eur. J. Clin. Investig. 2014, 44, 1206–1214.
  78. Ukon, Y.; Makino, T.; Kodama, J.; Tsukazaki, H.; Tateiwa, D.; Yoshikawa, H.; Kaito, T. Molecular-Based Treatment Strategies for Osteoporosis: A Literature Review. Int. J. Mol. Sci. 2019, 20, 2557.
  79. Delgado-Calle, J.; Sato, A.Y.; Bellido, T. Role and mechanism of action of sclerostin in bone. Bone 2017, 96, 29–37.
  80. Reid, I.R. Targeting Sclerostin in Postmenopausal Osteoporosis: Focus on Romosozumab and Blosozumab. BioDrugs 2017, 31, 289–297.
  81. Li, X.; Zhang, Y.; Kang, H.; Liu, W.; Liu, P.; Zhang, J.; Harris, S.E.; Wu, D. Sclerostin Binds to LRP5/6 and Antagonizes Canonical Wnt Signaling. J. Biol. Chem. 2005, 280, 19883–19887.
  82. Guañabens, N.; Gifre, L.; Miquel, R.; Peris, P.; Monegal, A.; Dubrueil, M.; Arias, A.; Parés, A.; Ruiz-Gaspà, S. Sclerostin Expression in Bile Ducts of Patients with Chronic Cholestasis May Influence the Bone Disease in Primary Biliary Cirrhosis. J. Bone Miner. Res. 2016, 31, 1725–1733.
  83. Rhee, Y.; Kim, W.J.; Han, K.J.; Kil Lim, S.; Kim, S.H. Effect of liver dysfunction on circulating sclerostin. J. Bone Miner. Metab. 2013, 32, 545–549.
  84. Locatelli, V.; Bianchi, V.E. Effect of GH/IGF-1 on Bone Metabolism and Osteoporsosis. Int. J. Endocrinol. 2014, 2014, 235060.
  85. Kobayashi, Y.; Uehara, S.; Udagawa, N.; Takahashi, N. Regulation of bone metabolism by Wnt signals. J. Biochem. 2016, 159, 387–392.
  86. Guerra-Menéndez, L.; Sádaba, M.C.; Puche, J.E.; Lavandera, J.L.; de Castro, L.F.; de Gortázar, A.R.; Castilla-Cortázar, I. IGF-I increases markers of osteoblastic activity and reduces bone resorption via osteoprotegerin and RANK-ligand. J. Trans. Med. 2013, 11, 271.
  87. De La Garza, R.G.; Morales-Garza, L.A.; Martin-Estal, I.; Castilla-Cortazar, I. Insulin-Like Growth Factor-1 Deficiency and Cirrhosis Establishment. J. Clin. Med. Res. 2017, 9, 233–247.
  88. Thrailkill, K.M.; Lumpkin, C.K.; Bunn, R.C.; Kemp, S.F.; Fowlkes, J.L. Is insulin an anabolic agent in bone? Dissecting the diabetic bone for clues. Am. J. Physiol. Metab. 2005, 289, E735–E745.
  89. Neong, S.F.; Billington, E.O.; Congly, S.E. Sexual Dysfunction and Sex Hormone Abnormalities in Patients with Cirrhosis: Review of Pathogenesis and Management. Hepatology 2018, 69, 2683–2695.
  90. Golds, G.; Houdek, D.; Arnason, T. Male Hypogonadism and Osteoporosis: The Effects, Clinical Consequences, and Treatment of Testosterone Deficiency in Bone Health. Int. J. Endocrinol. 2017, 2017, 4602129.
  91. Sarkar, M.; Lai, J.C.; Sawinski, D.; Zeigler, T.E.; Cedars, M.; Forde, K.A. Sex hormone levels by presence and severity of cirrhosis in women with chronic hepatitis C virus infection. J. Viral Hepat. 2018, 26, 258–262.
  92. Nakchbandi, I.A. Osteoporosis and fractures in liver disease: Relevance, pathogenesis and therapeutic implications. World J. Gastroenterol. 2014, 20, 9427–9438.
  93. Zumoff, B.; Fishman, J.; Gallagher, T.F.; Hellman, L. Estradiol metabolism in cirrhosis. J. Clin. Investig. 1968, 47, 20–25.
  94. Cruz-Jentoft, A.J.; Sayer, A.A. Sarcopenia. Lancet 2019, 393, 2636–2646.
  95. Cruz-Jentoft, A.J.; Bahat, G.; Bauer, J.; Boirie, Y.; Bruyère, O.; Cederholm, T.; Cooper, C.; Landi, F.; Rolland, Y.; Sayer, A.A.; et al. Sarcopenia: Revised European consensus on definition and diagnosis. Age Ageing 2019, 48, 16–31.
  96. Chen, L.-K.; Woo, J.; Assantachai, P.; Auyeung, T.-W.; Chou, M.-Y.; Iijima, K.; Jang, H.C.; Kang, L.; Kim, M.; Kim, S.; et al. Asian Working Group for Sarcopenia: 2019 Consensus Update on Sarcopenia Diagnosis and Treatment. J. Am. Med. Dir. Assoc. 2020, 21, 300–307.e2.
  97. Cruz-Jentoft, A.J.; Baeyens, J.P.; Bauer, J.M.; Boirie, Y.; Cederholm, T.; Landi, F.; Martin, F.C.; Michel, J.-P.; Rolland, Y.; Schneider, S.M.; et al. Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing 2010, 39, 412–423.
  98. Clark, B.C.; Manini, T.M. Sarcopenia =/= dynapenia. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2008, 63, 829–834.
  99. Lee, Y.-H.; Jung, K.S.; Kim, S.U.; Yoon, H.-J.; Yun, Y.J.; Lee, B.-W.; Kang, E.S.; Han, K.-H.; Lee, H.C.; Cha, B.-S. Sarcopaenia is associated with NAFLD independently of obesity and insulin resistance: Nationwide surveys (KNHANES 2008–2011). J. Hepatol. 2015, 63, 486–493.
  100. Hong, H.C.; Hwang, S.Y.; Choi, H.Y.; Yoo, H.J.; Seo, J.A.; Kim, S.G.; Kim, N.H.; Baik, S.H.; Choi, D.S.; Choi, K.M. Relationship between sarcopenia and nonalcoholic fatty liver disease: The Korean Sarcopenic Obesity Study. Hepatology 2014, 59, 1772–1778.
  101. Carias, S.; Castellanos, A.L.; Vilchez, V.; Nair, R.; Cruz, A.C.D.; Watkins, J.; Barrett, T.; Trushar, P.; Esser, K.; Gedaly, R. Nonalcoholic steatohepatitis is strongly associated with sarcopenic obesity in patients with cirrhosis undergoing liver transplant evaluation. J. Gastroenterol. Hepatol. 2016, 31, 628–633.
  102. Koo, B.K.; Kim, D.; Joo, S.K.; Kim, J.H.; Chang, M.S.; Kim, B.G.; Lee, K.L.; Kim, W. Sarcopenia is an independent risk factor for non-alcoholic steatohepatitis and significant fibrosis. J. Hepatol. 2017, 66, 123–131.
  103. Gan, D.; Wang, L.; Jia, M.; Ru, Y.; Ma, Y.; Zheng, W.; Zhao, X.; Yang, F.; Wang, T.; Mu, Y.; et al. Low muscle mass and low muscle strength associate with nonalcoholic fatty liver disease. Clin. Nutr. 2020, 39, 1124–1130.
  104. Kim, G.; Lee, S.; Lee, Y.; Jun, J.E.; Ahn, J.; Bae, J.C.; Jin, S.; Hur, K.Y.; Jee, J.H.; Lee, M.; et al. Relationship Between Relative Skeletal Muscle Mass and Nonalcoholic Fatty Liver Disease: A 7-Year Longitudinal Study. Hepatology 2018, 68, 1755–1768.
  105. Tandon, P.; Ney, M.; Irwin, I.; Ma, M.M.; Gramlich, L.; Bain, V.G.; Esfandiari, N.; Baracos, V.; Montano-Loza, A.J.; Myers, R.P. Severe muscle depletion in patients on the liver transplant wait list: Its prevalence and independent prognostic value. Liver Transplant. 2012, 18, 1209–1216.
  106. Montano-Loza, A.J.; Meza-Junco, J.; Prado, C.M.; Lieffers, J.R.; Baracos, V.E.; Bain, V.G.; Sawyer, M.B. Muscle Wasting Is Associated with Mortality in Patients with Cirrhosis. Clin. Gastroenterol. Hepatol. 2012, 10, 166–173.e1.
  107. Merli, M.; Giusto, M.; Lucidi, C.; Giannelli, V.; Pentassuglio, I.; Di Gregorio, V.; Lattanzi, B.; Riggio, O. Muscle depletion increases the risk of overt and minimal hepatic encephalopathy: Results of a prospective study. Metab. Brain Dis. 2013, 28, 281–284.
  108. Kim, T.Y.; Kim, M.Y.; Sohn, J.H.; Kim, S.M.; Ryu, J.A.; Lim, S.; Kim, Y. Sarcopenia as a Useful Predictor for Long-Term Mortality in Cirrhotic Patients with Ascites. J. Korean Med. Sci. 2014, 29, 1253–1259.
  109. Hanai, T.; Shiraki, M.; Nishimura, K.; Ohnishi, S.; Imai, K.; Suetsugu, A.; Takai, K.; Shimizu, M.; Moriwaki, H. Sarcopenia impairs prognosis of patients with liver cirrhosis. Nutrient 2015, 31, 193–199.
  110. Hanai, T.; Shiraki, M.; Ohnishi, S.; Miyazaki, T.; Ideta, T.; Kochi, T.; Imai, K.; Suetsugu, A.; Takai, K.; Moriwaki, H.; et al. Rapid skeletal muscle wasting predicts worse survival in patients with liver cirrhosis. Hepatol. Res. 2016, 46, 743–751.
  111. Nardelli, S.; Lattanzi, B.; Torrisi, S.; Greco, F.; Farcomeni, A.; Gioia, S.; Merli, M.; Riggio, O. Sarcopenia Is Risk Factor for Development of Hepatic Encephalopathy after Transjugular Intrahepatic Portosystemic Shunt Placement. Clin. Gastroenterol. Hepatol. 2017, 15, 934–936.
  112. Durand, F.; Buyse, S.; Francoz, C.; Laouénan, C.; Bruno, O.; Belghiti, J.; Moreau, R.; Vilgrain, V.; Valla, D. Prognostic value of muscle atrophy in cirrhosis using psoas muscle thickness on computed tomography. J. Hepatol. 2014, 60, 1151–1157.
  113. Montano-Loza, A.J.; Duarte-Rojo, A.; Meza-Junco, J.; Baracos, V.E.; Sawyer, M.B.; Pang, J.X.Q.; Beaumont, C.; Esfandiari, N.; Myers, R.P. Inclusion of Sarcopenia Within MELD (MELD-Sarcopenia) and the Prediction of Mortality in Patients with Cirrhosis. Clin. Transl. Gastroenterol. 2015, 6, e102.
  114. Kang, S.H.; Jeong, W.K.; Baik, S.K.; Cha, S.H.; Kim, M.Y. Impact of sarcopenia on prognostic value of cirrhosis: Going beyond the hepatic venous pressure gradient and MELD score. J. Cachex-Sarcopenia Muscle 2018, 9, 860–870.
  115. Englesbe, M.J.; Patel, S.P.; He, K.; Lynch, R.J.; Schaubel, D.E.; Harbaugh, C.; Holcombe, S.A.; Wang, S.C.; Segev, D.L.; Sonnenday, C.J. Sarcopenia and Mortality after Liver Transplantation. J. Am. Coll. Surg. 2010, 211, 271–278.
  116. Kaido, T.; Ogawa, K.; Fujimoto, Y.; Ogura, Y.; Hata, K.; Ito, T.; Tomiyama, K.; Yagi, S.; Mori, A.; Uemoto, S. Impact of Sarcopenia on Survival in Patients Undergoing Living Donor Liver Transplantation. Arab. Archaeol. Epigr. 2013, 13, 1549–1556.
  117. Krell, R.W.; Kaul, D.R.; Martin, A.R.; Englesbe, M.J.; Sonnenday, C.J.; Cai, S.; Malani, P.N. Association between sarcopenia and the risk of serious infection among adults undergoing liver transplantation. Liver Transplant. 2013, 19, 1396–1402.
  118. Tsien, C.; Garber, A.; Narayanan, A.; Shah, S.N.; Barnes, D.; Eghtesad, B.; Fung, J.; McCullough, A.J.; Dasarathy, S. Post-liver transplantation sarcopenia in cirrhosis: A prospective evaluation. J. Gastroenterol. Hepatol. 2014, 29, 1250–1257.
  119. Montano-Loza, A.J.; Meza-Junco, J.; Baracos, V.E.; Prado, C.M.M.; Ma, M.; Meeberg, G.; Beaumont, C.; Tandon, P.; Esfandiari, N.; Sawyer, M.B.; et al. Severe muscle depletion predicts postoperative length of stay but is not associated with survival after liver transplantation. Liver Transplant. 2014, 20, 640–648.
  120. Masuda, T.; Shirabe, K.; Ikegami, T.; Harimoto, N.; Yoshizumi, T.; Soejima, Y.; Uchiyama, H.; Ikeda, T.; Baba, H.; Maehara, Y. Sarcopenia is a prognostic factor in living donor liver transplantation. Liver Transplant. 2014, 20, 401–407.
  121. Hamaguchi, Y.; Kaido, T.; Okumura, S.; Fujimoto, Y.; Ogawa, K.; Mori, A.; Hammad, A.; Tamai, Y.; Inagaki, N.; Uemoto, S. Impact of quality as well as quantity of skeletal muscle on outcomes after liver transplantation. Liver Transplant. 2014, 20, 1413–1419.
  122. Kalafateli, M.; Mantzoukis, K.; Yau, Y.C.; Mohammad, A.O.; Arora, S.; Rodrigues, S.; De Vos, M.; Papadimitriou, K.; Thorburn, D.; O’Beirne, J.; et al. Malnutrition and sarcopenia predict post-liver transplantation outcomes independently of the Model for End-stage Liver Disease score. J. Cachex-Sarcopenia Muscle 2017, 8, 113–121.
  123. Meza-Junco, J.; Montano-Loza, A.J.; Baracos, V.E.; Prado, C.M.; Bain, V.G.; Beaumont, C.; Esfandiari, N.; Lieffers, J.R.; Sawyer, M.B. Sarcopenia as a Prognostic Index of Nutritional Status in Concurrent Cirrhosis and Hepatocellular Carcinoma. J. Clin. Gastroenterol. 2013, 47, 861–870.
  124. Harimoto, N.; Shirabe, K.; Yamashita, Y.-I.; Ikegami, T.; Yoshizumi, T.; Soejima, Y.; Ikeda, T.; Maehara, Y.; Nishie, A.; Yamanaka, T. Sarcopenia as a predictor of prognosis in patients following hepatectomy for hepatocellular carcinoma. BJS 2013, 100, 1523–1530.
  125. Fujiwara, N.; Nakagawa, H.; Kudo, Y.; Tateishi, R.; Taguri, M.; Watadani, T.; Nakagomi, R.; Kondo, M.; Nakatsuka, T.; Minami, T.; et al. Sarcopenia, intramuscular fat deposition, and visceral adiposity independently predict the outcomes of hepatocellular carcinoma. J. Hepatol. 2015, 63, 131–140.
  126. Drummond, M.J.; Dreyer, H.C.; Fry, C.S.; Glynn, E.L.; Rasmussen, B.B. Nutritional and contractile regulation of human skeletal muscle protein synthesis and mTORC1 signaling. J. Appl. Physiol. 2009, 106, 1374–1384.
  127. Nishikawa, H.; Enomoto, H.; Ishii, A.; Iwata, Y.; Miyamoto, Y.; Ishii, N.; Yuri, Y.; Hasegawa, K.; Nakano, C.; Nishimura, T.; et al. Elevated serum myostatin level is associated with worse survival in patients with liver cirrhosis. J. Cachex-Sarcopenia Muscle 2017, 8, 915–925.
  128. Han, H.; Zhou, X.; Mitch, W.E.; Goldberg, A.L. Myostatin/activin pathway antagonism: Molecular basis and therapeutic potential. Int. J. Biochem. Cell Biol. 2013, 45, 2333–2347.
  129. Garikipati, D.K.; Rodgers, B.D. Myostatin inhibits myosatellite cell proliferation and consequently activates differentiation: Evidence for endocrine-regulated transcript processing. J. Endocrinol. 2012, 215, 177–187.
More
This entry is offline, you can click here to edit this entry!
Video Production Service