Skip Navigation
Skip to contents

Diabetes Metab J : Diabetes & Metabolism Journal

Search
OPEN ACCESS

Articles

Page Path
HOME > Diabetes Metab J > Volume 39(1); 2015 > Article
Review
Pathophysiology Hepatokines as a Link between Obesity and Cardiovascular Diseases
Hye Jin Yoo, Kyung Mook Choi
Diabetes & Metabolism Journal 2015;39(1):10-15.
DOI: https://doi.org/10.4093/dmj.2015.39.1.10
Published online: February 16, 2015
  • 4,384 Views
  • 51 Download
  • 67 Web of Science
  • 68 Crossref
  • 69 Scopus

Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea.

Corresponding author: Kyung Mook Choi. Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, 148 Gurodong-ro, Guro-gu, Seoul 152-703, Korea. medica7@gmail.com

Copyright © 2015 Korean Diabetes Association

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • Non-alcoholic fatty liver disease, which is considered a hepatic manifestation of metabolic syndrome, independently increases the risks of developing cardiovascular disease (CVD) and type 2 diabetes mellitus. Recent emerging evidence suggests that a group of predominantly liver-derived proteins called hepatokines directly affect the progression of atherosclerosis by modulating endothelial dysfunction and infiltration of inflammatory cells into vessel walls. Here, we summarize the role of the representative hepatokines fibroblast growth factor 21, fetuin-A, and selenoprotein P in the progression of CVD.
Non-alcoholic fatty liver disease (NAFLD) is a spectrum of progressive liver diseases, including simple steatosis, non-alcoholic steatohepatitis, fibrosis, and eventually cirrhosis, in the absence of excessive alcohol consumption [1] and has been considered a hepatic manifestation of metabolic syndrome. NAFLD is a marker of pathologic ectopic fat accumulation combined with a low-grade chronic inflammatory state [2]. The liver is a pivotal visceral organ with respect to fat accumulation. Accumulation of lipid droplets decreases the efficacy of insulin signaling. Hepatic lipids also induce endoplasmic reticulum stress, leading to activation of c-Jun N-terminal kinases and nuclear factor κB (NF-κB), two major regulators of inflammatory pathways that can inhibit phosphorylation of insulin receptor substraste-1 [3]. Recent evidence suggests that NAFLD is an emerging risk factor for cardiovascular disease (CVD) independent of traditional CVD risk factors, implying that NAFLD might be directly involved in CVD pathogenesis. Here, we will summarize the relationship between NAFLD and CVD, focusing on the causative roles of liver-derived hepatokines in the progression of CVD (Fig. 1).
A recent comprehensive systematic review of 27 studies showed that NAFLD is associated with subclinical atherosclerosis independent of traditional risk factors and metabolic syndrome and across a wide range of ethnicities [4]. In that review, 14 among a total of 16 studies that examined the association of NAFLD with carotid intima media thickness (CIMT) demonstrated a significant increase in the mean CIMT in individuals with NAFLD compared to healthy people, even after adjusting for other confounding variables [4]. Using multislice computed tomography, Akabame et al. [5] showed that liver fat was significantly related to the presence of vulnerable plaques with a lipid core. Furthermore, Villanova et al. [6] reported that brachial artery flow-mediated dilatation (FMD), representing endothelial dysfunction, was reduced by 48% in subjects with NAFLD and that NAFLD predicted a reduced FMD (odds ratio [OR], 6.7; 95% confidence interval [CI], 1.26 to 36.1) after adjusting for age, sex, body mass index, and insulin resistance.
Several large cross-sectional and prospective studies showed that NAFLD was associated with an increased prevalence of CVD and the incidence of cardiovascular (CV) events irrespective of other CVD risk factors. In 2,839 patients with type 2 diabetes mellitus (DM), those with NAFLD had a higher prevalence of coronary, cerebrovascular, and peripheral vascular disease than their counterparts without NAFLD, independent of conventional CVD risk factors, medication history, and DM-related variables [7]. In addition, during a median 7.3 years of follow-up, an elevated baseline level of serum γ-glutamyl transferase, a surrogate marker for hepatic steatosis, was significantly associated with increased risk of all-cause and CVD mortality in men, even after adjusting for other metabolic risk factors [8]. Therefore, these studies suggested that assessment of NAFLD might be a helpful approach to more accurate and early CVD risk stratification.
The underlying mechanism by which NAFLD increases the risk of CVD has not been clarified; moreover, whether NAFLD is an independent risk factor for CVD or simply a risk marker that coexists in people at increased risk of CVD is still controversial. The potential mechanism by which NAFLD increases CVD risk is based on the development of inflamed visceral adipose tissue [9], which is the main source of an elevated flux of free fatty acids (FFAs) into the portal vein for direct transport to the liver and subsequent hepatic fat accumulation [10]. Therefore, NAFLD can be considered a sensitive marker of pathological dysfunction of visceral adipose tissue that is more relevant to CV outcome than simply adipose tissue mass.
On the other hand, hepatic steatosis itself leads to intrahepatic inflammation through activation of NF-κB pathways that exacerbate insulin resistance both locally in the liver and systemically. The liver of subjects with NAFLD might release a variety of proatherogenic, proinflammatory, and diabetogenic mediators such as high-sensitivity C-reactive protein (hsCRP), fibrinogen, and plasminogen activator inhibitor-1 (PAI-1), which have important roles in the development of CVD [1]. hsCRP, which is primarily produced by the liver and is a marker of inflammation, was an independent predictor of CV events in several large studies [11]. Similarly, fibrinogen and PAI-1 also originate from hepatic tissue and are activators of the coagulation system thus enhancing atherothrombosis [2], suggesting that an increase in liver-secreted factors in NAFLD plays an important role in the pathogenesis of systemic inflammation and atherosclerosis. Therefore, the liver functions as an inducer of systemic inflammation as well as a target organ of various inflammatory reactions that occur within dysfunctional adipose tissue.
Recent evidence suggests that a group of predominantly liver-derived proteins called hepatokines directly affect glucose and lipid metabolism, similar to previously described adipokines and myokines [12]. At the present time, fetuin-A, fibroblast growth factor 21 (FGF-21), and selenoprotein P (SeP) are considered representative hepatokines; however, there have been very limited studies exploring the direct function of these hepatokines on the development of CVD.
FGF-21 is a 181-amino acid peptide hormone that is primarily secreted by the liver and acts as a potent metabolic regulator [13]. Expression of the human FGF-21 gene is mediated by peroxisome proliferator-activated receptor-α (PPAR-α) during starvation [14] and regulated by PPAR-γ after feeding [15]. Circulating FFAs, a characteristic feature of fasting, and different kinds of stress such as hepatic injury, chemical insult, and disease, stimulate secretion of FGF-21 into the circulation [16]. Administration of FGF-21 to animal models and humans has been shown to decrease body weight and levels of blood triglycerides and low density lipoprotein (LDL) cholesterol, and to improve insulin sensitivity. Mashili et al. [17] reported that FGF-21 treatment induced basal and insulin-stimulated glucose uptake in human skeletal muscle cells through upregulation of glucose transporter-1. Very recently, a randomized phase 1 clinical trial showed that treatment with LY2405319, an analog of FGF-21, produced significant improvements in dyslipidemia of obese human subjects with type 2 DM [18]. FGF-21 stimulates lipolysis in the diabetic db/db mouse and obese humans, and individuals with a cluster of metabolic disorders have increased serum FGF-21 levels [19] to compensate for the abnormal metabolic status.
Early studies on FGF-21 focused on its role as a metabolic hormone during fasting or starvation. More recent studies have indicated a possible role of FGF-21 in CVD. Planavila et al. [20] demonstrated that FGF-21 knockout mice exhibited increased cardiac mass and impaired cardiac function, which could be ameliorated by treatment with FGF-21, suggesting its protective role against hypertrophic insults. Furthermore, FGF-21 infusion into a rat heart significantly recovered cardiac function following myocardial infarction [21]. In cultured rodent cardiac microvascular endothelial cells, FGF-21 expression was upregulated when the cells were incubated with oxidized LDL, indicating that FGF-21 might be secreted by endothelial cells in response to stress and that elevated levels may be a signal of endothelial cell injury [22]. In accordance with these animal studies, elevated serum FGF-21 levels are associated with carotid atherosclerosis in humans, independent of established CVD risk factors [23]. Multivariate logistic regression analysis also identified serum FGF-21 level as one of the independent factors of coronary artery disease occurrence (OR, 2.98; 95% CI, 1.014 to 8.786; P<0.05) [24]. Furthermore, we previously reported that brachial-ankle pulse wave velocity reflecting arterial stiffness had a significant positive correlation with circulating FGF-21 levels [25]. Therefore, FGF-21 may be an attractive target for the diagnosis and treatment of obesity and related diseases, including CVD.
Fetuin-A is a 64-kDa phosphorylated glycoprotein that is primarily synthesized by hepatocytes [26]. Fetuin-A is a natural inhibitor of the insulin receptor tyrosine kinase, leading to insulin resistance in rodents [27]. Apart from its direct effects on the insulin receptor, fetuin-A promotes insulin resistance by propagating a proinflammatory state. Fetuin-A treatment aggravates proinflammatory cytokine expression while reducing adiponectin expression in both adipocytes and monocytes [28]. Furthermore, incubation of HepG2 cells or rat hepatocytes with palmitate stimulates binding of NF-κB to the fetuin-A promoter, thereby augmenting fetuin-A synthesis and secretion [29]. We previously reported that palmitate-induced fetuin-A stimulated triacylglycerol accumulation in hepatocytes and that adiponectin inhibited palmitate-induced hepatic fetuin-A expression through the adenosine monophosphate-activated protein kinase (AMPK) pathway [30]. These results suggest that fetuin-A might directly cause insulin resistance and modulate inflammatory reactions, leading to various metabolic disturbances. Consistent with these findings, many epidemiologic studies have observed elevated levels of circulating fetuin-A in obesity and related metabolic diseases including type 2 DM, metabolic syndrome, and NAFLD [31,32,33]. Our previous study showed a significant decrease in circulating fetuin-A levels after 12 weeks of caloric restriction that was accompanied by improvements in visceral fat area, blood pressure, lipid profiles, and glucose levels [34]. In the European Prospective Investigation into Cancer and Nutrition-Potsdam study, plasma fetuin-A levels were positively associated with the incidence of diabetes after adjustment for sex, body mass index, waist circumference, and lifestyle risk factors during 7 years of follow-up [35].
However, the relationship between circulating fetuin-A and CVD risk appears to be more complicated. Fetuin-A can bind with Ca2+, inhibiting ectopic calcification [36]. In studies examining patients with chronic kidney diseases, fetuin-A level is inversely associated with calcification scores, CV events, and CV mortality [37]. Nondiabetics with a higher fetuin-A level have decreased risks of incident CVD and CVD-related mortality, whereas type 2 diabetics with higher fetuin-A levels have increased risks of incident CVD and CVD-related mortality [38,39]. Possible mechanisms by which fetuin-A can promote atherosclerosis in CVD patients are through induction of insulin resistance and increased expression of cytokines in monocytes that participate in the inflammation. Siegel-Axel et al. [40] demonstrated that fetuin-A inhibited the proliferation of perivascular fat cells and increased the expression of proinflammatory cytokines including interlukin-8, interlukin-6, monocyte chemotactic protein-1, and PAI-1 in these cells. Further studies should be performed to determine the exact function of fetuin-A in CVD according to the diverse underlying patient conditions.
SeP, a 42-kDa glycoprotein, is produced in the liver and secreted into plasma [41]. SeP was recently identified as a hepatokine associated with insulin resistance in humans through serial analysis of gene expression [42]. Administration of SeP to mice decreased insulin signaling and glucose metabolism in both liver and skeletal muscle, whereas SeP-deficient mice showed enhanced insulin signaling and improved glucose tolerance [42]. In our previous studies, patients with type 2 DM and those with NAFLD had higher serum SeP levels than healthy controls [43,44]. Furthermore, we found that salsalate and adiponectin ameliorated palmitate-induced insulin resistance in hepatocytes by inhibition of SeP through the AMPK-Forkhead box protein O1α (FOXO1α) pathway [45], suggesting that SeP acting via the AMPK-FOXO1α-dependent pathway might be a novel mechanism mediating the antidiabetic effects of salsalate and adiponectin.
However, there have been very few studies on the relationship of SeP with CVD. We first reported that circulating SeP level has an independent association with carotid intima-media thickness even after adjustment for other confounding factors [44]. Very recently, Ishikura et al. [46] showed that physiological concentrations of SeP inhibited vascular endothelial growth factor-stimulated cell proliferation, tubule formation, and migration in human umbilical vein endothelial cells, leading to impaired angiogenesis and delay of wound closure in mice overexpressing SeP. Further studies are needed to explore the direct relationship between SeP with CVD and the underlying mechanism.
Hepatokines that are mainly secreted from the liver are known to directly affect glucose and lipid metabolism. There is accumulating evidence that various hepatokines can modulate inflammatory processes that in turn mediate atherosclerotic process. Therefore, identification of novel hepatokines might allow the development of new strategies to diagnose and treat NAFLD-related metabolic disturbances, including CVD.
Acknowledgements
This research was supported by the Basic Science Research Program through the National Research Foundation of Korea funded by the Ministry of Education, Science and Technology (2012006363, KMC and 2012R1A1A1005257, HJY), by the Brain Korea 21 Project of the Ministry of Education and Human Resources Development, Republic of Korea (KMC and SHB), and by a grant from Korea University (KMC).

No potential conflict of interest relevant to this article was reported.

  • 1. Anstee QM, Targher G, Day CP. Progression of NAFLD to diabetes mellitus, cardiovascular disease or cirrhosis. Nat Rev Gastroenterol Hepatol 2013;10:330-344. ArticlePubMedPDF
  • 2. Bhatia LS, Curzen NP, Calder PC, Byrne CD. Non-alcoholic fatty liver disease: a new and important cardiovascular risk factor? Eur Heart J 2012;33:1190-1200. ArticlePubMed
  • 3. Byrne CD, Targher G. Ectopic fat, insulin resistance, and nonalcoholic fatty liver disease: implications for cardiovascular disease. Arterioscler Thromb Vasc Biol 2014;34:1155-1161. PubMed
  • 4. Oni ET, Agatston AS, Blaha MJ, Fialkow J, Cury R, Sposito A, Erbel R, Blankstein R, Feldman T, Al-Mallah MH, Santos RD, Budoff MJ, Nasir K. A systematic review: burden and severity of subclinical cardiovascular disease among those with nonalcoholic fatty liver; should we care? Atherosclerosis 2013;230:258-267. ArticlePubMed
  • 5. Akabame S, Hamaguchi M, Tomiyasu K, Tanaka M, Kobayashi-Takenaka Y, Nakano K, Oda Y, Yoshikawa T. Evaluation of vulnerable coronary plaques and non-alcoholic fatty liver disease (NAFLD) by 64-detector multislice computed tomography (MSCT). Circ J 2008;72:618-625. ArticlePubMed
  • 6. Villanova N, Moscatiello S, Ramilli S, Bugianesi E, Magalotti D, Vanni E, Zoli M, Marchesini G. Endothelial dysfunction and cardiovascular risk profile in nonalcoholic fatty liver disease. Hepatology 2005;42:473-480. ArticlePubMed
  • 7. Targher G, Bertolini L, Padovani R, Rodella S, Tessari R, Zenari L, Day C, Arcaro G. Prevalence of nonalcoholic fatty liver disease and its association with cardiovascular disease among type 2 diabetic patients. Diabetes Care 2007;30:1212-1218. ArticlePubMedPDF
  • 8. Haring R, Wallaschofski H, Nauck M, Dorr M, Baumeister SE, Volzke H. Ultrasonographic hepatic steatosis increases prediction of mortality risk from elevated serum gamma-glutamyl transpeptidase levels. Hepatology 2009;50:1403-1411. ArticlePubMed
  • 9. Badman MK, Flier JS. The adipocyte as an active participant in energy balance and metabolism. Gastroenterology 2007;132:2103-2115. ArticlePubMed
  • 10. Bjorntorp P. "Portal" adipose tissue as a generator of risk factors for cardiovascular disease and diabetes. Arteriosclerosis 1990;10:493-496. ArticlePubMed
  • 11. Lavie CJ, Milani RV, Verma A, O'Keefe JH. C-reactive protein and cardiovascular diseases: is it ready for primetime? Am J Med Sci 2009;338:486-492. ArticlePubMed
  • 12. Stefan N, Haring HU. The role of hepatokines in metabolism. Nat Rev Endocrinol 2013;9:144-152. ArticlePubMedPDF
  • 13. Dushay J, Chui PC, Gopalakrishnan GS, Varela-Rey M, Crawley M, Fisher FM, Badman MK, Martinez-Chantar ML, Maratos-Flier E. Increased fibroblast growth factor 21 in obesity and nonalcoholic fatty liver disease. Gastroenterology 2010;139:456-463. ArticlePubMed
  • 14. Galman C, Lundasen T, Kharitonenkov A, Bina HA, Eriksson M, Hafstrom I, Dahlin M, Amark P, Angelin B, Rudling M. The circulating metabolic regulator FGF21 is induced by prolonged fasting and PPARalpha activation in man. Cell Metab 2008;8:169-174. PubMed
  • 15. Dutchak PA, Katafuchi T, Bookout AL, Choi JH, Yu RT, Mangelsdorf DJ, Kliewer SA. Fibroblast growth factor-21 regulates PPARgamma activity and the antidiabetic actions of thiazolidinediones. Cell 2012;148:556-567. ArticlePubMedPMC
  • 16. Yu H, Xia F, Lam KS, Wang Y, Bao Y, Zhang J, Gu Y, Zhou P, Lu J, Jia W, Xu A. Circadian rhythm of circulating fibroblast growth factor 21 is related to diurnal changes in fatty acids in humans. Clin Chem 2011;57:691-700. ArticlePubMedPDF
  • 17. Mashili FL, Austin RL, Deshmukh AS, Fritz T, Caidahl K, Bergdahl K, Zierath JR, Chibalin AV, Moller DE, Kharitonenkov A, Krook A. Direct effects of FGF21 on glucose uptake in human skeletal muscle: implications for type 2 diabetes and obesity. Diabetes Metab Res Rev 2011;27:286-297. ArticlePubMed
  • 18. Gaich G, Chien JY, Fu H, Glass LC, Deeg MA, Holland WL, Kharitonenkov A, Bumol T, Schilske HK, Moller DE. The effects of LY2405319, an FGF21 analog, in obese human subjects with type 2 diabetes. Cell Metab 2013;18:333-340. ArticlePubMed
  • 19. Dostalova I, Haluzikova D, Haluzik M. Fibroblast growth factor 21: a novel metabolic regulator with potential therapeutic properties in obesity/type 2 diabetes mellitus. Physiol Res 2009;58:1-7. PubMed
  • 20. Planavila A, Redondo I, Hondares E, Vinciguerra M, Munts C, Iglesias R, Gabrielli LA, Sitges M, Giralt M, van Bilsen M, Villarroya F. Fibroblast growth factor 21 protects against cardiac hypertrophy in mice. Nat Commun 2013;4:2019ArticlePubMedPDF
  • 21. Patel V, Adya R, Chen J, Ramanjaneya M, Bari MF, Bhudia SK, Hillhouse EW, Tan BK, Randeva HS. Novel insights into the cardio-protective effects of FGF21 in lean and obese rat hearts. PLoS One 2014;9:e87102ArticlePubMedPMC
  • 22. Lu Y, Liu JH, Zhang LK, Du J, Zeng XJ, Hao G, Huang J, Zhao DH, Wang GZ, Zhang YC. Fibroblast growth factor 21 as a possible endogenous factor inhibits apoptosis in cardiac endothelial cells. Chin Med J (Engl) 2010;123:3417-3421. PubMed
  • 23. Chow WS, Xu A, Woo YC, Tso AW, Cheung SC, Fong CH, Tse HF, Chau MT, Cheung BM, Lam KS. Serum fibroblast growth factor-21 levels are associated with carotid atherosclerosis independent of established cardiovascular risk factors. Arterioscler Thromb Vasc Biol 2013;33:2454-2459. ArticlePubMed
  • 24. Shen Y, Ma X, Zhou J, Pan X, Hao Y, Zhou M, Lu Z, Gao M, Bao Y, Jia W. Additive relationship between serum fibroblast growth factor 21 level and coronary artery disease. Cardiovasc Diabetol 2013;12:124ArticlePubMedPMCPDF
  • 25. Yang SJ, Hong HC, Choi HY, Yoo HJ, Cho GJ, Hwang TG, Baik SH, Choi DS, Kim SM, Choi KM. Effects of a three-month combined exercise programme on fibroblast growth factor 21 and fetuin-A levels and arterial stiffness in obese women. Clin Endocrinol (Oxf) 2011;75:464-469. ArticlePubMedPDF
  • 26. Nangami GN, Watson K, Parker-Johnson K, Okereke KO, Sakwe A, Thompson P, Frimpong N, Ochieng J. Fetuin-A (alpha2HS-glycoprotein) is a serum chemo-attractant that also promotes invasion of tumor cells through Matrigel. Biochem Biophys Res Commun 2013;438:660-665. PubMed
  • 27. Denecke B, Graber S, Schafer C, Heiss A, Woltje M, Jahnen-Dechent W. Tissue distribution and activity testing suggest a similar but not identical function of fetuin-B and fetuin-A. Biochem J 2003;376(Pt 1):135-145. ArticlePubMedPMCPDF
  • 28. Hennige AM, Staiger H, Wicke C, Machicao F, Fritsche A, Haring HU, Stefan N. Fetuin-A induces cytokine expression and suppresses adiponectin production. PLoS One 2008;3:e1765ArticlePubMedPMC
  • 29. Dasgupta S, Bhattacharya S, Biswas A, Majumdar SS, Mukhopadhyay S, Ray S, Bhattacharya S. NF-kappaB mediates lipid-induced fetuin-A expression in hepatocytes that impairs adipocyte function effecting insulin resistance. Biochem J 2010;429:451-462. PubMed
  • 30. Jung TW, Youn BS, Choi HY, Lee SY, Hong HC, Yang SJ, Yoo HJ, Kim BH, Baik SH, Choi KM. Salsalate and adiponectin ameliorate hepatic steatosis by inhibition of the hepatokine fetuin-A. Biochem Pharmacol 2013;86:960-969. ArticlePubMed
  • 31. Ou HY, Yang YC, Wu HT, Wu JS, Lu FH, Chang CJ. Increased fetuin-A concentrations in impaired glucose tolerance with or without nonalcoholic fatty liver disease, but not impaired fasting glucose. J Clin Endocrinol Metab 2012;97:4717-4723. ArticlePubMed
  • 32. Ix JH, Wassel CL, Kanaya AM, Vittinghoff E, Johnson KC, Koster A, Cauley JA, Harris TB, Cummings SR, Shlipak MG. Health ABC Study. Fetuin-A and incident diabetes mellitus in older persons. JAMA 2008;300:182-188. ArticlePubMedPMC
  • 33. Ix JH, Shlipak MG, Brandenburg VM, Ali S, Ketteler M, Whooley MA. Association between human fetuin-A and the metabolic syndrome: data from the Heart and Soul Study. Circulation 2006;113:1760-1767. ArticlePubMedPMC
  • 34. Choi KM, Han KA, Ahn HJ, Lee SY, Hwang SY, Kim BH, Hong HC, Choi HY, Yang SJ, Yoo HJ, Baik SH, Choi DS, Min KW. The effects of caloric restriction on fetuin-A and cardiovascular risk factors in rats and humans: a randomized controlled trial. Clin Endocrinol (Oxf) 2013;79:356-363. ArticlePubMed
  • 35. Stefan N, Fritsche A, Weikert C, Boeing H, Joost HG, Haring HU, Schulze MB. Plasma fetuin-A levels and the risk of type 2 diabetes. Diabetes 2008;57:2762-2767. ArticlePubMedPMCPDF
  • 36. Mori K, Emoto M, Inaba M. Fetuin-A and the cardiovascular system. Adv Clin Chem 2012;56:175-195. ArticlePubMed
  • 37. Evrard S, Delanaye P, Kamel S, Cristol JP, Cavalier E. SFBC/SN joined working group on vascular calcifications. Vascular calcification: from pathophysiology to biomarkers. Clin Chim Acta 2015;438:401-414. ArticlePubMed
  • 38. Jensen MK, Bartz TM, Mukamal KJ, Djousse L, Kizer JR, Tracy RP, Zieman SJ, Rimm EB, Siscovick DS, Shlipak M, Ix JH. Fetuin-A, type 2 diabetes, and risk of cardiovascular disease in older adults: the cardiovascular health study. Diabetes Care 2013;36:1222-1228. PubMedPMC
  • 39. Laughlin GA, Cummins KM, Wassel CL, Daniels LB, Ix JH. The association of fetuin-A with cardiovascular disease mortality in older community-dwelling adults: the Rancho Bernardo study. J Am Coll Cardiol 2012;59:1688-1696. PubMedPMC
  • 40. Siegel-Axel DI, Ullrich S, Stefan N, Rittig K, Gerst F, Klingler C, Schmidt U, Schreiner B, Randrianarisoa E, Schaller HE, Stock UA, Weigert C, Konigsrainer A, Haring HU. Fetuin-A influences vascular cell growth and production of proinflammatory and angiogenic proteins by human perivascular fat cells. Diabetologia 2014;57:1057-1066. ArticlePubMedPDF
  • 41. Burk RF, Hill KE. Selenoprotein P: an extracellular protein with unique physical characteristics and a role in selenium homeostasis. Annu Rev Nutr 2005;25:215-235. ArticlePubMed
  • 42. Misu H, Takamura T, Takayama H, Hayashi H, Matsuzawa-Nagata N, Kurita S, Ishikura K, Ando H, Takeshita Y, Ota T, Sakurai M, Yamashita T, Mizukoshi E, Yamashita T, Honda M, Miyamoto K, Kubota T, Kubota N, Kadowaki T, Kim HJ, Lee IK, Minokoshi Y, Saito Y, Takahashi K, Yamada Y, Takakura N, Kaneko S. A liver-derived secretory protein, selenoprotein P, causes insulin resistance. Cell Metab 2010;12:483-495. ArticlePubMed
  • 43. Choi HY, Hwang SY, Lee CH, Hong HC, Yang SJ, Yoo HJ, Seo JA, Kim SG, Kim NH, Baik SH, Choi DS, Choi KM. Increased selenoprotein p levels in subjects with visceral obesity and nonalcoholic Fatty liver disease. Diabetes Metab J 2013;37:63-71. ArticlePubMedPMC
  • 44. Yang SJ, Hwang SY, Choi HY, Yoo HJ, Seo JA, Kim SG, Kim NH, Baik SH, Choi DS, Choi KM. Serum selenoprotein P levels in patients with type 2 diabetes and prediabetes: implications for insulin resistance, inflammation, and atherosclerosis. J Clin Endocrinol Metab 2011;96:E1325-E1329. ArticlePubMed
  • 45. Jung TW, Choi HY, Lee SY, Hong HC, Yang SJ, Yoo HJ, Youn BS, Baik SH, Choi KM. Salsalate and adiponectin improve palmitate-induced insulin resistance via inhibition of selenoprotein P through the AMPK-FOXO1alpha pathway. PLoS One 2013;8:e66529ArticlePubMedPMC
  • 46. Ishikura K, Misu H, Kumazaki M, Takayama H, Matsuzawa-Nagata N, Tajima N, Chikamoto K, Lan F, Ando H, Ota T, Sakurai M, Takeshita Y, Kato K, Fujimura A, Miyamoto K, Saito Y, Kameo S, Okamoto Y, Takuwa Y, Takahashi K, Kidoya H, Takakura N, Kaneko S, Takamura T. Selenoprotein P as a diabetes-associated hepatokine that impairs angiogenesis by inducing VEGF resistance in vascular endothelial cells. Diabetologia 2014;57:1968-1976. ArticlePubMedPDF
Fig. 1

Underlying mechanism connecting non-alcoholic fatty liver disease (NAFLD) to cardiovascular disease (CVD). LDL, low density lipoprotein; HDL, high density lipoprotein; hsCRP, high-sensitivity C-reactive protein; PAI-1, plasminogen activator inhibitor-1; FGF-21, fibroblast growth factor 21.

dmj-39-10-g001.jpg

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • The liver-heart axis in patients with severe obesity: The association between liver fibrosis and chronic myocardial injury may be explained by shared risk factors of cardiovascular disease
      J. Young, K.A. Seeberg, K.M. Aakre, H. Borgeraas, N. Nordstrand, T. Wisløff, J. Hjelmesæth, T. Omland, J.K. Hertel
      Clinical Biochemistry.2024; 123: 110688.     CrossRef
    • From Beats to Metabolism: the Heart at the Core of Interorgan Metabolic Cross Talk
      Rafael Romero-Becera, Ayelén M. Santamans, Alba C. Arcones, Guadalupe Sabio
      Physiology.2024; 39(2): 98.     CrossRef
    • Exerkines and cardiometabolic benefits of exercise: from bench to clinic
      Leigang Jin, Candela Diaz-Canestro, Yu Wang, Michael Andrew Tse, Aimin Xu
      EMBO Molecular Medicine.2024;[Epub]     CrossRef
    • Mechanisms underlying the bidirectional association between nonalcoholic fatty liver disease and hypertension
      Hironori Nakagami
      Hypertension Research.2023; 46(2): 539.     CrossRef
    • Effects of treatment of non-alcoholic fatty liver disease on heart failure with preserved ejection fraction
      Zifeng Yang, Ruifeng Tian, Xiao-Jing Zhang, Jingjing Cai, Zhi-Gang She, Hongliang Li
      Frontiers in Cardiovascular Medicine.2023;[Epub]     CrossRef
    • “Alphabet” Selenoproteins: Implications in Pathology
      Carmen Beatrice Dogaru, Carmen Duță, Corina Muscurel, Irina Stoian
      International Journal of Molecular Sciences.2023; 24(20): 15344.     CrossRef
    • Overview of the Association between Non-Alcoholic Fatty Liver Disease and Hypertension
      Niki S. Kakouri, Costas G. Thomopoulos, Eirini P. Siafi, Angeliki E. Valatsou, Kyriakos S. Dimitriadis, Iliana P. Mani, Sotirios P. Patsilinakos, Dimitrios M. Tousoulis, Konstantinos P. Tsioufis
      Cardiology Discovery.2023;[Epub]     CrossRef
    • Reversal of nonalcoholic fatty liver disease reduces the risk of cardiovascular disease among Korean
      Yun Hwan Oh, Seogsong Jeong, Sun Jae Park, Joseph C Ahn, Sang Min Park
      Medicine.2023; 102(44): e35804.     CrossRef
    • Change of cardiovascular risk associated serologic biomarkers after gastric bypass: A comparison of diabetic and non-diabetic Asian patients
      Jih-Hua Wei, Ming-Hsien Lee, Wei-Jei Lee, Shu-Chun Chen, Owaid M. Almalki, Jung-Chien Chen, Chun-Chi Wu, Yi-Chih Lee
      Asian Journal of Surgery.2022; 45(11): 2253.     CrossRef
    • The effect of 12 weeks of training in water on serum levels of SIRT1 and FGF-21, glycemic index, and lipid profile in patients with type 2 diabetes
      Bahram Jamali Gharakhanlou, Solmaz Babaei Bonab
      International Journal of Diabetes in Developing Countries.2022; 42(4): 727.     CrossRef
    • Obesity is an important determinant of severity in newly defined metabolic dysfunction-associated fatty liver disease
      Ji Hye Huh, Kwang Joon Kim, Seung Up Kim, Bong-Soo Cha, Byung-Wan Lee
      Hepatobiliary & Pancreatic Diseases International.2022; 21(3): 241.     CrossRef
    • Hepatic PTEN Signaling Regulates Systemic Metabolic Homeostasis through Hepatokines-Mediated Liver-to-Peripheral Organs Crosstalk
      Flavien Berthou, Cyril Sobolewski, Daniel Abegg, Margot Fournier, Christine Maeder, Dobrochna Dolicka, Marta Correia de Sousa, Alexander Adibekian, Michelangelo Foti
      International Journal of Molecular Sciences.2022; 23(7): 3959.     CrossRef
    • AMP activated kinase negatively regulates hepatic Fetuin-A via p38 MAPK-C/EBPβ/E3 Ubiquitin Ligase Signaling pathway
      Vishal Kothari, Jeganathan Ramesh Babu, Suresh T. Mathews, Regis Moreau
      PLOS ONE.2022; 17(5): e0266472.     CrossRef
    • Modern aspects of pathogenesis of comorbidity of non-alcoholic fatty liver disease and hypertension in the presence or absence of chronic kidney disease.
      K.O. Prosolenko, К.A. Lapshyna, V.V. Ryabuha
      Shidnoevropejskij zurnal vnutrisnoi ta simejnoi medicini.2022; 2022(1): 55.     CrossRef
    • A coagulation factor moonlights in the heart
      Dan Tong, Joseph A. Hill
      Science.2022; 377(6613): 1382.     CrossRef
    • Fetuin-A and Its Association with Anthropometric, Atherogenic, and Biochemical Parameters and Indices among Women with Polycystic Ovary Syndrome
      Karolina Kulik-Kupka, Marzena Jabczyk, Justyna Nowak, Paweł Jagielski, Bartosz Hudzik, Barbara Zubelewicz-Szkodzińska
      Nutrients.2022; 14(19): 4034.     CrossRef
    • Mitochondrial Dysfunction is a Key Pathway that Links Saturated Fat Intake to the Development and Progression of NAFLD
      Ruth C. R. Meex, Ellen E. Blaak
      Molecular Nutrition & Food Research.2021;[Epub]     CrossRef
    • New Insights Into the Comorbidity of Coronary Heart Disease and Depression
      Yeshun Wu, Bin Zhu, Zijun Chen, Jiahao Duan, Ailin Luo, Ling Yang, Chun Yang
      Current Problems in Cardiology.2021; 46(3): 100413.     CrossRef
    • Liver hepatokines and peroxisomes as therapeutic targets for cardiovascular diseases
      Kerui Huang, Hua Bai
      Future Cardiology.2021; 17(4): 535.     CrossRef
    • Hepatocardiac or Cardiohepatic Interaction: From Traditional Chinese Medicine to Western Medicine
      Yaxing Zhang, Xian-Ming Fang, Michał Tomczyk
      Evidence-Based Complementary and Alternative Medicine.2021; 2021: 1.     CrossRef
    • Nonalcoholic Fatty Liver Disease (NAFLD). Mitochondria as Players and Targets of Therapies?
      Agostino Di Ciaula, Salvatore Passarella, Harshitha Shanmugam, Marica Noviello, Leonilde Bonfrate, David Q.-H. Wang, Piero Portincasa
      International Journal of Molecular Sciences.2021; 22(10): 5375.     CrossRef
    • Investigating Fetuin-A and Paraoxonase-1 Activity as Markers in Polycystic Ovary Syndrome Based on Body Mass Index: A Prospective Case-Control Study
      Tugba Gurbuz, Sebnem Alanya Tosun, Aysegul Cebi, Oya Gokmen, Murat Usta
      Cureus.2021;[Epub]     CrossRef
    • Extent and features of liver steatosis in vitro pave the way to endothelial dysfunction without physical cell-to-cell contact
      Francesca Baldini, Mohamad Khalil, Nadia Serale, Adriana Voci, Piero Portincasa, Laura Vergani
      Nutrition, Metabolism and Cardiovascular Diseases.2021; 31(12): 3522.     CrossRef
    • Association of Fetuin-B with Subclinical Atherosclerosis in Obese Chinese Adults
      Zhibin Li, Chunmei He, Yongwen Liu, Dongmei Wang, Mingzhu Lin, Changqin Liu, Xiulin Shi, Zheng Chen, Xuejun Li, Shuyu Yang, Weihua Li
      Journal of Atherosclerosis and Thrombosis.2020; 27(5): 418.     CrossRef
    • Serum Fetuin-A levels are increased and associated with insulin resistance in women with polycystic ovary syndrome
      Sha Liu, Wenjing Hu, Yirui He, Ling Li, Hua Liu, Lin Gao, Gangyi Yang, Xin Liao
      BMC Endocrine Disorders.2020;[Epub]     CrossRef
    • The intrinsic and extrinsic elements regulating inflammation
      M. Mollaei, A. Abbasi, Z.M. Hassan, N. Pakravan
      Life Sciences.2020; 260: 118258.     CrossRef
    • A Close Relationship between Non-Alcoholic Fatty Liver Disease Marker and New-Onset Hypertension in Healthy Korean Adults
      Jae-Hyung Roh, Jae-Hyeong Park, Hanbyul Lee, Yong-Hoon Yoon, Minsu Kim, Yong-Giun Kim, Gyung-Min Park, Jae-Hwan Lee, In-Whan Seong
      Korean Circulation Journal.2020; 50(8): 695.     CrossRef
    • Elevated blood pressure, cardiometabolic risk and target organ damage in youth with overweight and obesity
      Procolo Di Bonito, Lucia Pacifico, Maria Rosaria Licenziati, Claudio Maffeis, Anita Morandi, Melania Manco, Emanuele Miraglia del Giudice, Anna Di Sessa, Giuseppina Campana, Nicola Moio, Marco Giorgio Baroni, Claudio Chiesa, Giovanni De Simone, Giuliana V
      Nutrition, Metabolism and Cardiovascular Diseases.2020; 30(10): 1840.     CrossRef
    • Liver governs adipose remodelling via extracellular vesicles in response to lipid overload
      Yue Zhao, Meng-Fei Zhao, Shan Jiang, Jing Wu, Jia Liu, Xian-Wen Yuan, Di Shen, Jing-Zi Zhang, Nan Zhou, Jian He, Lei Fang, Xi-Tai Sun, Bin Xue, Chao-Jun Li
      Nature Communications.2020;[Epub]     CrossRef
    • Metabolic Inflammation—A Role for Hepatic Inflammatory Pathways as Drivers of Comorbidities in Nonalcoholic Fatty Liver Disease?
      Nadine Gehrke, Jörn M. Schattenberg
      Gastroenterology.2020; 158(7): 1929.     CrossRef
    • Effect of Moderate Aerobic Exercise on Serum Levels of FGF21 and Fetuin A in Women with Type 2 Diabetes
      Exir Vizvari, Parvin farzanegi, Hajar Abbas Zade
      Medical Laboratory Journal.2020; 14(6): 17.     CrossRef
    • Fibroblast growth factor 21: A role in cardiometabolic disorders and cardiovascular risk prediction?
      Niki Katsiki, Christos Mantzoros
      Metabolism.2019; 93: iii.     CrossRef
    • Current and Future Nutritional Strategies to Modulate Inflammatory Dynamics in Metabolic Disorders
      Willem van den Brink, Jolanda van Bilsen, Kanita Salic, Femke P. M. Hoevenaars, Lars Verschuren, Robert Kleemann, Jildau Bouwman, Gabriele V. Ronnett, Ben van Ommen, Suzan Wopereis
      Frontiers in Nutrition.2019;[Epub]     CrossRef
    • Fetuin-A is also an adipokine
      Ishwarlal Jialal, Roma Pahwa
      Lipids in Health and Disease.2019;[Epub]     CrossRef
    • Fibroblast growth factor 21 in lipid metabolism and non-alcoholic fatty liver disease
      Xin Su, Yi Kong, Daoquan Peng
      Clinica Chimica Acta.2019; 498: 30.     CrossRef
    • Inter-organ cross-talk in metabolic syndrome
      Christina Priest, Peter Tontonoz
      Nature Metabolism.2019; 1(12): 1177.     CrossRef
    • The persistence of fatty liver has a differential impact on the development of diabetes: The Kangbuk Samsung Health Study
      Ji Cheol Bae, Ji Min Han, Jung Hwan Cho, Hyemi Kwon, Se Eun Park, Cheol-Young Park, Won-Young Lee, Ki-Won Oh, Sam Kwon, Sung-Woo Park, Eun Jung Rhee
      Diabetes Research and Clinical Practice.2018; 135: 1.     CrossRef
    • Hypertension, diabetes, atherosclerosis and NASH: Cause or consequence?
      Amedeo Lonardo, Fabio Nascimbeni, Alessandro Mantovani, Giovanni Targher
      Journal of Hepatology.2018; 68(2): 335.     CrossRef
    • Frequency and Antibiotic Resistance Pattern of Diarrheagenic Escherichia coli (DEC) Strains Isolated from Children Aged Less Than 10 Years
      Shahram Shahraki Zahedani, Nasrin sayadzai
      Medical Laboratory Journal .2018; 12(2): 7.     CrossRef
    • Fetuin-A levels are increased in the adipose tissue of diabetic obese humans but not in circulation
      Abdelkrim Khadir, Sina Kavalakatt, Dhanya Madhu, Maha Hammad, Sriraman Devarajan, Jaakko Tuomilehto, Ali Tiss
      Lipids in Health and Disease.2018;[Epub]     CrossRef
    • Implication of liver enzymes on incident cardiovascular diseases and mortality: A nationwide population-based cohort study
      Kyung Mook Choi, Kyungdo Han, Sanghyun Park, Hye Soo Chung, Nam Hoon Kim, Hye Jin Yoo, Ji-A Seo, Sin Gon Kim, Nan Hee Kim, Sei Hyun Baik, Yong Gyu Park, Seon Mee Kim
      Scientific Reports.2018;[Epub]     CrossRef
    • Obesity and Male Infertility: Role of Fatty Acids in the Modulation of Sperm Energetic Metabolism
      Alessandra Ferramosca, Mariangela Di Giacomo, Natalina Moscatelli, Vincenzo Zara
      European Journal of Lipid Science and Technology.2018;[Epub]     CrossRef
    • The association between circulating fetuin-A levels and type 2 diabetes mellitus risk: systematic review and meta-analysis of observational studies
      F. Roshanzamir, M. Miraghajani, M. H. Rouhani, M. Mansourian, R. Ghiasvand, S. M. Safavi
      Journal of Endocrinological Investigation.2018; 41(1): 33.     CrossRef
    • Clinical and Body Compositional Factors Associated with Metabolic Syndrome in Obese Koreans: A Cross-Sectional Study
      Yoo Mee Kim, Sunghoon Kim, Se Hwa Kim, Young Jun Won
      Metabolic Syndrome and Related Disorders.2018; 16(6): 290.     CrossRef
    • Association of non-alcoholic steatohepatitis with subclinical myocardial dysfunction in non-cirrhotic patients
      Yong-ho Lee, Kwang Joon Kim, Myung eun Yoo, Gyuri Kim, Hye-jin Yoon, Kwanhyeong Jo, Jong-Chan Youn, Mijin Yun, Jun Yong Park, Chi Young Shim, Byung-Wan Lee, Seok-Min Kang, Jong-Won Ha, Bong-Soo Cha, Eun Seok Kang
      Journal of Hepatology.2018; 68(4): 764.     CrossRef
    • Non-alcoholic fatty liver disease and hypertension: coprevalent or correlated?
      Dimitrios Oikonomou, Georgios Georgiopoulos, Vassiliki Katsi, Chris Kourek, Constantinos Tsioufis, Alexendra Alexopoulou, Evaggelia Koutli, Dimitrios Tousoulis
      European Journal of Gastroenterology & Hepatology.2018; 30(9): 979.     CrossRef
    • Implication of Nonalcoholic Fatty Liver Disease, Metabolic Syndrome, and Subclinical Inflammation on Mild Renal Insufficiency
      Ga Eun Nam, Soon Young Hwang, Hye Soo Chung, Ju Hee Choi, Hyun Jung Lee, Nam Hoon Kim, Hye Jin Yoo, Ji-A Seo, Sin Gon Kim, Nan Hee Kim, Sei Hyun Baik, Kyung Mook Choi
      International Journal of Endocrinology.2018; 2018: 1.     CrossRef
    • Serum levels of fetuin-A are negatively associated with log transformation levels of thyroid-stimulating hormone in patients with hyperthyroidism or euthyroidism
      Fen-Yu Tseng, Yen-Ting Chen, Yu-Chiao Chi, Pei-Lung Chen, Wei-Shiung Yang
      Medicine.2018; 97(46): e13254.     CrossRef
    • Relationship of Circulating Fetuin-A Levels with Body Size and Metabolic Phenotypes
      Hye Soo Chung, Hyun Jung Lee, Soon Young Hwang, Ju-Hee Choi, Hye Jin Yoo, Ji A. Seo, Sin Gon Kim, Nan Hee Kim, Dong Seop Choi, Sei Hyun Baik, Kyung Mook Choi
      International Journal of Endocrinology.2018; 2018: 1.     CrossRef
    • Lobeglitazone, a Novel Thiazolidinedione, Improves Non-Alcoholic Fatty Liver Disease in Type 2 Diabetes: Its Efficacy and Predictive Factors Related to Responsiveness
      Yong-ho Lee, Jae Hyeon Kim, So Ra Kim, Heung Yong Jin, Eun-Jung Rhee, Young Min Cho, Byung-Wan Lee
      Journal of Korean Medical Science.2017; 32(1): 60.     CrossRef
    • Dietary fatty acids influence sperm quality and function
      A. Ferramosca, N. Moscatelli, M. Di Giacomo, V. Zara
      Andrology.2017; 5(3): 423.     CrossRef
    • Obesity is more closely related with hepatic steatosis and fibrosis measured by transient elastography than metabolic health status
      Ji Hye Huh, Kwang Joon Kim, Seung Up Kim, Seung Hwan Han, Kwang-Hyub Han, Bong-Soo Cha, Choon Hee Chung, Byung-Wan Lee
      Metabolism.2017; 66: 23.     CrossRef
    • Hepatokines: linking nonalcoholic fatty liver disease and insulin resistance
      Ruth C. R. Meex, Matthew J. Watt
      Nature Reviews Endocrinology.2017; 13(9): 509.     CrossRef
    • Association of leukocyte cell-derived chemotaxin 2 (LECT2) with NAFLD, metabolic syndrome, and atherosclerosis
      Hye Jin Yoo, Soon Young Hwang, Ju-Hee Choi, Hyun Jung Lee, Hye Soo Chung, Ji-A Seo, Sin Gon Kim, Nan Hee Kim, Sei Hyun Baik, Dong Seop Choi, Kyung Mook Choi, Pavel Strnad
      PLOS ONE.2017; 12(4): e0174717.     CrossRef
    • T2DiACoD: A Gene Atlas of Type 2 Diabetes Mellitus Associated Complex Disorders
      Jyoti Rani, Inna Mittal, Atreyi Pramanik, Namita Singh, Namita Dube, Smriti Sharma, Bhanwar Lal Puniya, Muthukurussi Varieth Raghunandanan, Ahmed Mobeen, Srinivasan Ramachandran
      Scientific Reports.2017;[Epub]     CrossRef
    • The fatty liver index as a predictor of incident chronic kidney disease in a 10-year prospective cohort study
      Ji Hye Huh, Jang Young Kim, Eunhee Choi, Jae Seok Kim, Yoosoo Chang, Ki-Chul Sung, Tatsuo Shimosawa
      PLOS ONE.2017; 12(7): e0180951.     CrossRef
    • A high‐fat diet negatively affects rat sperm mitochondrial respiration
      A. Ferramosca, A. Conte, N. Moscatelli, V. Zara
      Andrology.2016; 4(3): 520.     CrossRef
    • The Impact of Organokines on Insulin Resistance, Inflammation, and Atherosclerosis
      Kyung Mook Choi
      Endocrinology and Metabolism.2016; 31(1): 1.     CrossRef
    • Metformin Restores Parkin-Mediated Mitophagy, Suppressed by Cytosolic p53
      Young Song, Woo Lee, Yong-ho Lee, Eun Kang, Bong-Soo Cha, Byung-Wan Lee
      International Journal of Molecular Sciences.2016; 17(1): 122.     CrossRef
    • Pathogenesis of nonalcoholic steatohepatitis
      Wensheng Liu, Robert D. Baker, Tavleen Bhatia, Lixin Zhu, Susan S. Baker
      Cellular and Molecular Life Sciences.2016; 73(10): 1969.     CrossRef
    • The ratio of skeletal muscle mass to visceral fat area is a main determinant linking circulating irisin to metabolic phenotype
      You-Cheol Hwang, Won Seon Jeon, Cheol-Young Park, Byung-Soo Youn
      Cardiovascular Diabetology.2016;[Epub]     CrossRef
    • Sirtuin 3 (SIRT3) Regulates α-Smooth Muscle Actin (α-SMA) Production through the Succinate Dehydrogenase-G Protein-coupled Receptor 91 (GPR91) Pathway in Hepatic Stellate Cells
      Ying Hui Li, Dae Hee Choi, Eun Hye Lee, Su Ryeon Seo, Seungkoo Lee, Eun-Hee Cho
      Journal of Biological Chemistry.2016; 291(19): 10277.     CrossRef
    • Statins Increase Mitochondrial and Peroxisomal Fatty Acid Oxidation in the Liver and Prevent Non-Alcoholic Steatohepatitis in Mice
      Han-Sol Park, Jung Eun Jang, Myoung Seok Ko, Sung Hoon Woo, Bum Joong Kim, Hyun Sik Kim, Hye Sun Park, In-Sun Park, Eun Hee Koh, Ki-Up Lee
      Diabetes & Metabolism Journal.2016; 40(5): 376.     CrossRef
    • Hormetic and regulatory effects of lipid peroxidation mediators in pancreatic beta cells
      Giuseppe Maulucci, Bareket Daniel, Ofir Cohen, Yossef Avrahami, Shlomo Sasson
      Molecular Aspects of Medicine.2016; 49: 49.     CrossRef
    • Extrahepatic Complications of Nonalcoholic Fatty Liver Disease
      Kristina R. Chacko, John Reinus
      Clinics in Liver Disease.2016; 20(2): 387.     CrossRef
    • Use of a Diabetes Self-Assessment Score to Predict Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis
      Gyuri Kim, Yong-ho Lee, Young Min Park, Jungghi Kim, Heesuk Kim, Byung-Wan Lee, Eun Seok Kang, Bong-Soo Cha, Hyun Chul Lee, Dae Jung Kim
      Medicine.2015; 94(27): e1103.     CrossRef
    • A Prospective Study of Fatty Liver Index and Incident Hypertension: The KoGES-ARIRANG Study
      Ji Hye Huh, Song Vogue Ahn, Sang Baek Koh, Eunhee Choi, Jang Young Kim, Ki-Chul Sung, Eung Ju Kim, Jeong Bae Park, Vincent Wong
      PLOS ONE.2015; 10(11): e0143560.     CrossRef
    • Endocrine causes of nonalcoholic fatty liver disease
      Laura Marino
      World Journal of Gastroenterology.2015; 21(39): 11053.     CrossRef

    • PubReader PubReader
    • Cite
      CITE
      export Copy
      Close
      Download Citation
      Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

      Format:
      • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
      • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
      Include:
      • Citation for the content below
      Hepatokines as a Link between Obesity and Cardiovascular Diseases
      Diabetes Metab J. 2015;39(1):10-15.   Published online February 16, 2015
      Close
    • XML DownloadXML Download
    Figure

    Diabetes Metab J : Diabetes & Metabolism Journal