Skip to main content
Top
Published in: Current Gastroenterology Reports 1/2012

01-02-2012 | Liver (BR Bacon, Section Editor)

Iron Metabolism in Nonalcoholic Fatty Liver Disease

Authors: James E. Nelson, Heather Klintworth, Kris V. Kowdley

Published in: Current Gastroenterology Reports | Issue 1/2012

Login to get access

Abstract

Non-Alcoholic Fatty Liver Disease (NAFLD) is a common worldwide clinical and major public health problem affecting both adults and children in developed nations. Increased hepatic iron stores are observed in about one-third of adult NAFLD patients. Iron deposition may occur in parenchymal and/or non-parenchymal cells of the reticuloendothelial system (RES). Similar patterns of iron deposition have been associated with increased severity of other chronic liver diseases including HCV infection and dysmetabolic iron overload, suggesting there may be a common mechanism for hepatic iron deposition in these diseases. In NAFLD, iron may potentiate the onset and progression of disease by increasing oxidative stress and altering insulin signaling and lipid metabolism. The impact of iron in these processes may depend upon the sub-cellular location of iron deposition in hepatocytes or RES cells. Iron depletion therapy has shown efficacy at reducing serum aminotransferase levels and improving insulin sensitivity in subjects with NAFLD.
Literature
1.
go back to reference Cohen JC, Horton JD, Hobbs HH. Human fatty liver disease: old questions and new insights. Science. 2011;332:1519–23.PubMedCrossRef Cohen JC, Horton JD, Hobbs HH. Human fatty liver disease: old questions and new insights. Science. 2011;332:1519–23.PubMedCrossRef
2.
3.
go back to reference George DK, Goldwurm S, MacDonald GA, et al. Increased hepatic iron concentration in nonalcoholic steatohepatitis is associated with increased fibrosis. Gastroenterology. 1998;114:311–8.PubMedCrossRef George DK, Goldwurm S, MacDonald GA, et al. Increased hepatic iron concentration in nonalcoholic steatohepatitis is associated with increased fibrosis. Gastroenterology. 1998;114:311–8.PubMedCrossRef
4.
go back to reference Bacon BR, Farahvash MJ, Janney CG, Neuschwander-Tetri BA. Nonalcoholic steatohepatitis: an expanded clinical entity. Gastroenterology. 1994;107:1103–9.PubMed Bacon BR, Farahvash MJ, Janney CG, Neuschwander-Tetri BA. Nonalcoholic steatohepatitis: an expanded clinical entity. Gastroenterology. 1994;107:1103–9.PubMed
5.
go back to reference Sumida Y, Nakashima T, Yoh T, et al. Serum thioredoxin levels as a predictor of steatohepatitis in patients with nonalcoholic fatty liver disease. J Hepatol. 2003;38:32–8.PubMedCrossRef Sumida Y, Nakashima T, Yoh T, et al. Serum thioredoxin levels as a predictor of steatohepatitis in patients with nonalcoholic fatty liver disease. J Hepatol. 2003;38:32–8.PubMedCrossRef
6.
go back to reference Akin K, Beyler AR, Kaya M, Erden E. The importance of iron and copper accumulation in the pathogenesis of non-alcoholic steatohepatitis. Turk J Gastroenterol. 2003;14:228–33.PubMed Akin K, Beyler AR, Kaya M, Erden E. The importance of iron and copper accumulation in the pathogenesis of non-alcoholic steatohepatitis. Turk J Gastroenterol. 2003;14:228–33.PubMed
7.
go back to reference Bugianesi E, Manzini P, D’Antico S, et al. Relative contribution of iron burden, HFE mutations, and insulin resistance to fibrosis in nonalcoholic fatty liver. Hepatology. 2004;39:179–87.PubMedCrossRef Bugianesi E, Manzini P, D’Antico S, et al. Relative contribution of iron burden, HFE mutations, and insulin resistance to fibrosis in nonalcoholic fatty liver. Hepatology. 2004;39:179–87.PubMedCrossRef
8.
go back to reference Chitturi S, Weltman M, Farrell GC, et al. HFE mutations, hepatic iron, and fibrosis: ethnic-specific association of NASH with C282Y but not with fibrotic severity. Hepatology. 2002;36:142–9.PubMedCrossRef Chitturi S, Weltman M, Farrell GC, et al. HFE mutations, hepatic iron, and fibrosis: ethnic-specific association of NASH with C282Y but not with fibrotic severity. Hepatology. 2002;36:142–9.PubMedCrossRef
9.
go back to reference Younossi ZM, Gramlich T, Bacon BR, et al. Hepatic iron and nonalcoholic fatty liver disease. Hepatology. 1999;30:847–50.PubMedCrossRef Younossi ZM, Gramlich T, Bacon BR, et al. Hepatic iron and nonalcoholic fatty liver disease. Hepatology. 1999;30:847–50.PubMedCrossRef
10.
go back to reference Zamin Jr I, Mattos AA, Mattos AZ, et al. Prevalence of the hemochromatosis gene mutation in patients with nonalcoholic steatohepatitis and correlation with degree of liver fibrosis. Arq Gastroenterol. 2006;43:224–8.PubMedCrossRef Zamin Jr I, Mattos AA, Mattos AZ, et al. Prevalence of the hemochromatosis gene mutation in patients with nonalcoholic steatohepatitis and correlation with degree of liver fibrosis. Arq Gastroenterol. 2006;43:224–8.PubMedCrossRef
11.
go back to reference Deugnier Y, Turlin B. Pathology of hepatic iron overload. World J Gastroenterol. 2007;13:4755–60.PubMed Deugnier Y, Turlin B. Pathology of hepatic iron overload. World J Gastroenterol. 2007;13:4755–60.PubMed
13.
go back to reference •• Nelson JE, Wilson L, Brunt EM, et al.: Relationship between the pattern of hepatic iron deposition and histological severity in nonalcoholic fatty liver disease. Hepatology 2011, 53:448–457. This study performed on 849 subjects enrolled in the US NASH Clinical Research Network, established that one third of NAFLD subjects have hepatic iron deposits and that the pathogenic effects of iron depend upon the cellular location within the liver. PubMedCrossRef •• Nelson JE, Wilson L, Brunt EM, et al.: Relationship between the pattern of hepatic iron deposition and histological severity in nonalcoholic fatty liver disease. Hepatology 2011, 53:448–457. This study performed on 849 subjects enrolled in the US NASH Clinical Research Network, established that one third of NAFLD subjects have hepatic iron deposits and that the pathogenic effects of iron depend upon the cellular location within the liver. PubMedCrossRef
14.
go back to reference Sorrentino P, D’Angelo S, Ferbo U, et al. Liver iron excess in patients with hepatocellular carcinoma developed on non-alcoholic steato-hepatitis. J Hepatol. 2009;50:351–7.PubMedCrossRef Sorrentino P, D’Angelo S, Ferbo U, et al. Liver iron excess in patients with hepatocellular carcinoma developed on non-alcoholic steato-hepatitis. J Hepatol. 2009;50:351–7.PubMedCrossRef
15.
go back to reference •• Valenti L, Fracanzani AL, Bugianesi E, et al.: HFE genotype, parenchymal iron accumulation, and liver fibrosis in patients with nonalcoholic fatty liver disease. Gastroenterology 2010, 138:905–912. The second largest study to date investigating the effects of iron and HFE mutations in NAFLD, reported that hepatocellular iron, but not HFE mutations, contribute to increased fibosis in a large Italian cohort. PubMedCrossRef •• Valenti L, Fracanzani AL, Bugianesi E, et al.: HFE genotype, parenchymal iron accumulation, and liver fibrosis in patients with nonalcoholic fatty liver disease. Gastroenterology 2010, 138:905–912. The second largest study to date investigating the effects of iron and HFE mutations in NAFLD, reported that hepatocellular iron, but not HFE mutations, contribute to increased fibosis in a large Italian cohort. PubMedCrossRef
16.
go back to reference Kowdley KV. The role of iron in nonalcoholic fatty liver disease: the story continues. Gastroenterology. 2010;138:817–9.PubMedCrossRef Kowdley KV. The role of iron in nonalcoholic fatty liver disease: the story continues. Gastroenterology. 2010;138:817–9.PubMedCrossRef
17.
go back to reference Ganz T. Hepcidin–a regulator of intestinal iron absorption and iron recycling by macrophages. Best Pract Res Clin Haematol. 2005;18:171–82.PubMedCrossRef Ganz T. Hepcidin–a regulator of intestinal iron absorption and iron recycling by macrophages. Best Pract Res Clin Haematol. 2005;18:171–82.PubMedCrossRef
18.
go back to reference Xiong S, She H, Tsukamoto H. Signaling role of iron in NF-kappa B activation in hepatic macrophages. Comp Hepatol. 2004;3 Suppl 1:S36.PubMedCrossRef Xiong S, She H, Tsukamoto H. Signaling role of iron in NF-kappa B activation in hepatic macrophages. Comp Hepatol. 2004;3 Suppl 1:S36.PubMedCrossRef
19.
go back to reference Tsukamoto H. Iron regulation of hepatic macrophage TNFalpha expression. Free Radic Biol Med. 2002;32:309–13.PubMedCrossRef Tsukamoto H. Iron regulation of hepatic macrophage TNFalpha expression. Free Radic Biol Med. 2002;32:309–13.PubMedCrossRef
20.
go back to reference Xiong S, She H, Sung CK, Tsukamoto H. Iron-dependent activation of NF-kappaB in Kupffer cells: a priming mechanism for alcoholic liver disease. Alcohol. 2003;30:107–13.PubMedCrossRef Xiong S, She H, Sung CK, Tsukamoto H. Iron-dependent activation of NF-kappaB in Kupffer cells: a priming mechanism for alcoholic liver disease. Alcohol. 2003;30:107–13.PubMedCrossRef
21.
go back to reference De Domenico I, Ward DM, Langelier C, et al. The molecular mechanism of hepcidin-mediated ferroportin down-regulation. Mol Biol Cell. 2007;18:2569–78.PubMedCrossRef De Domenico I, Ward DM, Langelier C, et al. The molecular mechanism of hepcidin-mediated ferroportin down-regulation. Mol Biol Cell. 2007;18:2569–78.PubMedCrossRef
22.
go back to reference •• Nemeth E, Tuttle MS, Powelson J, et al.: Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 2004, 306:2090–2093. This landmark study established how hepcidin controls iron homeostasis. PubMedCrossRef •• Nemeth E, Tuttle MS, Powelson J, et al.: Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 2004, 306:2090–2093. This landmark study established how hepcidin controls iron homeostasis. PubMedCrossRef
23.
go back to reference Wang RH, Li C, Xu X, et al. A role of SMAD4 in iron metabolism through the positive regulation of hepcidin expression. Cell Metab. 2005;2:399–409.PubMedCrossRef Wang RH, Li C, Xu X, et al. A role of SMAD4 in iron metabolism through the positive regulation of hepcidin expression. Cell Metab. 2005;2:399–409.PubMedCrossRef
24.
go back to reference Gao J, Chen J, Kramer M, et al. Interaction of the hereditary hemochromatosis protein HFE with transferrin receptor 2 is required for transferrin-induced hepcidin expression. Cell Metab. 2009;9:217–27.PubMedCrossRef Gao J, Chen J, Kramer M, et al. Interaction of the hereditary hemochromatosis protein HFE with transferrin receptor 2 is required for transferrin-induced hepcidin expression. Cell Metab. 2009;9:217–27.PubMedCrossRef
25.
go back to reference Goswami T, Andrews NC. Hereditary hemochromatosis protein, HFE, interaction with transferrin receptor 2 suggests a molecular mechanism for mammalian iron sensing. J Biol Chem. 2006;281:28494–8.PubMedCrossRef Goswami T, Andrews NC. Hereditary hemochromatosis protein, HFE, interaction with transferrin receptor 2 suggests a molecular mechanism for mammalian iron sensing. J Biol Chem. 2006;281:28494–8.PubMedCrossRef
26.
go back to reference Schmidt PJ, Toran PT, Giannetti AM, et al. The transferrin receptor modulates Hfe-dependent regulation of hepcidin expression. Cell Metab. 2008;7:205–14.PubMedCrossRef Schmidt PJ, Toran PT, Giannetti AM, et al. The transferrin receptor modulates Hfe-dependent regulation of hepcidin expression. Cell Metab. 2008;7:205–14.PubMedCrossRef
27.
go back to reference Lee P, Peng H, Gelbart T, et al. Regulation of hepcidin transcription by interleukin-1 and interleukin-6. Proc Natl Acad Sci USA. 2005;102:1906–10.PubMedCrossRef Lee P, Peng H, Gelbart T, et al. Regulation of hepcidin transcription by interleukin-1 and interleukin-6. Proc Natl Acad Sci USA. 2005;102:1906–10.PubMedCrossRef
28.
go back to reference Wrighting DM, Andrews NC. Interleukin-6 induces hepcidin expression through STAT3. Blood. 2006;108:3204–9.PubMedCrossRef Wrighting DM, Andrews NC. Interleukin-6 induces hepcidin expression through STAT3. Blood. 2006;108:3204–9.PubMedCrossRef
29.
go back to reference Choi SO, Cho YS, Kim HL, Park JW. ROS mediate the hypoxic repression of the hepcidin gene by inhibiting C/EBPalpha and STAT-3. Biochem Biophys Res Commun. 2007;356:312–7.PubMedCrossRef Choi SO, Cho YS, Kim HL, Park JW. ROS mediate the hypoxic repression of the hepcidin gene by inhibiting C/EBPalpha and STAT-3. Biochem Biophys Res Commun. 2007;356:312–7.PubMedCrossRef
30.
go back to reference Pinto JP, Ribeiro S, Pontes H, et al. Erythropoietin mediates hepcidin expression in hepatocytes through EPOR signaling and regulation of C/EBPalpha. Blood. 2008;111:5727–33.PubMedCrossRef Pinto JP, Ribeiro S, Pontes H, et al. Erythropoietin mediates hepcidin expression in hepatocytes through EPOR signaling and regulation of C/EBPalpha. Blood. 2008;111:5727–33.PubMedCrossRef
31.
go back to reference Harrison-Findik DD, Schafer D, Klein E, et al. Alcohol metabolism-mediated oxidative stress down-regulates hepcidin transcription and leads to increased duodenal iron transporter expression. J Biol Chem. 2006;281:22974–82.PubMedCrossRef Harrison-Findik DD, Schafer D, Klein E, et al. Alcohol metabolism-mediated oxidative stress down-regulates hepcidin transcription and leads to increased duodenal iron transporter expression. J Biol Chem. 2006;281:22974–82.PubMedCrossRef
32.
go back to reference Oliveira SJ, Pinto JP, Picarote G, et al. ER stress-inducible factor CHOP affects the expression of hepcidin by modulating C/EBPalpha activity. PLoS One. 2009;4:e6618.PubMedCrossRef Oliveira SJ, Pinto JP, Picarote G, et al. ER stress-inducible factor CHOP affects the expression of hepcidin by modulating C/EBPalpha activity. PLoS One. 2009;4:e6618.PubMedCrossRef
33.
go back to reference Aigner E, Theurl I, Theurl M, et al. Pathways underlying iron accumulation in human nonalcoholic fatty liver disease. Am J Clin Nutr. 2008;87:1374–83.PubMed Aigner E, Theurl I, Theurl M, et al. Pathways underlying iron accumulation in human nonalcoholic fatty liver disease. Am J Clin Nutr. 2008;87:1374–83.PubMed
34.
go back to reference Barisani D, Pelucchi S, Mariani R, et al. Hepcidin and iron-related gene expression in subjects with Dysmetabolic Hepatic Iron Overload. J Hepatol. 2008;49:123–33.PubMedCrossRef Barisani D, Pelucchi S, Mariani R, et al. Hepcidin and iron-related gene expression in subjects with Dysmetabolic Hepatic Iron Overload. J Hepatol. 2008;49:123–33.PubMedCrossRef
35.
go back to reference Bekri S, Gual P, Anty R, et al. Increased adipose tissue expression of hepcidin in severe obesity is independent from diabetes and NASH. Gastroenterology. 2006;131:788–96.PubMedCrossRef Bekri S, Gual P, Anty R, et al. Increased adipose tissue expression of hepcidin in severe obesity is independent from diabetes and NASH. Gastroenterology. 2006;131:788–96.PubMedCrossRef
36.
go back to reference Senates E, Yilmaz Y, Colak Y, et al.: Serum Levels of Hepcidin in Patients with Biopsy-Proven Nonalcoholic Fatty Liver Disease. Metab Syndr Relat Disord 2011;9:287–290. Senates E, Yilmaz Y, Colak Y, et al.: Serum Levels of Hepcidin in Patients with Biopsy-Proven Nonalcoholic Fatty Liver Disease. Metab Syndr Relat Disord 2011;9:287–290.
37.
go back to reference Floreani A, Navaglia F, Rizzotto ER, et al. Mass spectrometry measurement of plasma hepcidin for the prediction of iron overload. Clin Chem Lab Med. 2011;49:197–206.PubMedCrossRef Floreani A, Navaglia F, Rizzotto ER, et al. Mass spectrometry measurement of plasma hepcidin for the prediction of iron overload. Clin Chem Lab Med. 2011;49:197–206.PubMedCrossRef
38.
go back to reference Pietrangelo A. Metals, oxidative stress, and hepatic fibrogenesis. Semin Liver Dis. 1996;16:13–30.PubMedCrossRef Pietrangelo A. Metals, oxidative stress, and hepatic fibrogenesis. Semin Liver Dis. 1996;16:13–30.PubMedCrossRef
39.
go back to reference Nakashima T, Sumida Y, Furutani M, et al. Elevation of serum thioredoxin levels in patients with nonalcoholic steatohepatitis. Hepatol Res. 2005;33:135–7.PubMed Nakashima T, Sumida Y, Furutani M, et al. Elevation of serum thioredoxin levels in patients with nonalcoholic steatohepatitis. Hepatol Res. 2005;33:135–7.PubMed
40.
go back to reference Malaguarnera L, Madeddu R, Palio E, et al. Heme oxygenase-1 levels and oxidative stress-related parameters in non-alcoholic fatty liver disease patients. J Hepatol. 2005;42:585–91.PubMedCrossRef Malaguarnera L, Madeddu R, Palio E, et al. Heme oxygenase-1 levels and oxidative stress-related parameters in non-alcoholic fatty liver disease patients. J Hepatol. 2005;42:585–91.PubMedCrossRef
41.
go back to reference MacDonald GA, Bridle KR, Ward PJ, et al. Lipid peroxidation in hepatic steatosis in humans is associated with hepatic fibrosis and occurs predominately in acinar zone 3. J Gastroenterol Hepatol. 2001;16:599–606.PubMedCrossRef MacDonald GA, Bridle KR, Ward PJ, et al. Lipid peroxidation in hepatic steatosis in humans is associated with hepatic fibrosis and occurs predominately in acinar zone 3. J Gastroenterol Hepatol. 2001;16:599–606.PubMedCrossRef
42.
go back to reference Fujita N, Miyachi H, Tanaka H, et al. Iron overload is associated with hepatic oxidative damage to DNA in nonalcoholic steatohepatitis. Cancer Epidemiol Biomarkers Prev. 2009;18:424–32.PubMedCrossRef Fujita N, Miyachi H, Tanaka H, et al. Iron overload is associated with hepatic oxidative damage to DNA in nonalcoholic steatohepatitis. Cancer Epidemiol Biomarkers Prev. 2009;18:424–32.PubMedCrossRef
43.
go back to reference Tsukamoto H, Rippe R, Niemela O, Lin M. Roles of oxidative stress in activation of Kupffer and Ito cells in liver fibrogenesis. J Gastroenterol Hepatol. 1995;10 Suppl 1:S50–3.PubMedCrossRef Tsukamoto H, Rippe R, Niemela O, Lin M. Roles of oxidative stress in activation of Kupffer and Ito cells in liver fibrogenesis. J Gastroenterol Hepatol. 1995;10 Suppl 1:S50–3.PubMedCrossRef
44.
go back to reference Tsukamoto H, Lin M, Ohata M, et al. Iron primes hepatic macrophages for NF-kappaB activation in alcoholic liver injury. Am J Physiol. 1999;277:G1240–50.PubMed Tsukamoto H, Lin M, Ohata M, et al. Iron primes hepatic macrophages for NF-kappaB activation in alcoholic liver injury. Am J Physiol. 1999;277:G1240–50.PubMed
45.
go back to reference • Chen L, Xiong S, She H, et al.: Iron causes interactions of TAK1, p21ras, and phosphatidylinositol 3-kinase in caveolae to activate IkappaB kinase in hepatic macrophages. J Biol Chem 2007, 282:5582–5588. An elegant study showing detailed analysis of the effects of iron on several signal transduction pathways within hepatic macrophages. PubMedCrossRef • Chen L, Xiong S, She H, et al.: Iron causes interactions of TAK1, p21ras, and phosphatidylinositol 3-kinase in caveolae to activate IkappaB kinase in hepatic macrophages. J Biol Chem 2007, 282:5582–5588. An elegant study showing detailed analysis of the effects of iron on several signal transduction pathways within hepatic macrophages. PubMedCrossRef
46.
go back to reference Hanada T, Yoshimura A. Regulation of cytokine signaling and inflammation. Cytokine Growth Factor Rev. 2002;13:413–21.PubMedCrossRef Hanada T, Yoshimura A. Regulation of cytokine signaling and inflammation. Cytokine Growth Factor Rev. 2002;13:413–21.PubMedCrossRef
47.
go back to reference Kershenobich SD, Weissbrod AB. Liver fibrosis and inflammation. A review Ann Hepatol. 2003;2:159–63. Kershenobich SD, Weissbrod AB. Liver fibrosis and inflammation. A review Ann Hepatol. 2003;2:159–63.
49.
go back to reference •• Otogawa K, Kinoshita K, Fujii H, et al.: Erythrophagocytosis by liver macrophages (Kupffer cells) promotes oxidative stress, inflammation, and fibrosis in a rabbit model of steatohepatitis: implications for the pathogenesis of human nonalcoholic steatohepatitis. Am J Pathol 2007, 170:967–980. This study provides mechanistic insight into the accumulation or iron in Kupffer cells and strong evidence that iron-laden Kupffer cells increase hepatic oxidative stress, inflammation and fibrosis in an animal model of NASH. PubMedCrossRef •• Otogawa K, Kinoshita K, Fujii H, et al.: Erythrophagocytosis by liver macrophages (Kupffer cells) promotes oxidative stress, inflammation, and fibrosis in a rabbit model of steatohepatitis: implications for the pathogenesis of human nonalcoholic steatohepatitis. Am J Pathol 2007, 170:967–980. This study provides mechanistic insight into the accumulation or iron in Kupffer cells and strong evidence that iron-laden Kupffer cells increase hepatic oxidative stress, inflammation and fibrosis in an animal model of NASH. PubMedCrossRef
50.
go back to reference Fernandez-Real JM, Lopez-Bermejo A, Ricart W. Cross-talk between iron metabolism and diabetes. Diabetes. 2002;51:2348–54.PubMedCrossRef Fernandez-Real JM, Lopez-Bermejo A, Ricart W. Cross-talk between iron metabolism and diabetes. Diabetes. 2002;51:2348–54.PubMedCrossRef
51.
go back to reference Barton JC, Acton RT, Leiendecker-Foster C, et al. Characteristics of participants with self-reported hemochromatosis or iron overload at HEIRS study initial screening. Am J Hematol. 2008;83:126–32.PubMedCrossRef Barton JC, Acton RT, Leiendecker-Foster C, et al. Characteristics of participants with self-reported hemochromatosis or iron overload at HEIRS study initial screening. Am J Hematol. 2008;83:126–32.PubMedCrossRef
52.
go back to reference Kulaksiz H, Fein E, Redecker P, et al. Pancreatic beta-cells express hepcidin, an iron-uptake regulatory peptide. J Endocrinol. 2008;197:241–9.PubMedCrossRef Kulaksiz H, Fein E, Redecker P, et al. Pancreatic beta-cells express hepcidin, an iron-uptake regulatory peptide. J Endocrinol. 2008;197:241–9.PubMedCrossRef
53.
go back to reference Kishimoto M, Endo H, Hagiwara S, et al. Immunohistochemical findings in the pancreatic islets of a patient with transfusional iron overload and diabetes: case report. J Med Invest. 2010;57:345–9.PubMedCrossRef Kishimoto M, Endo H, Hagiwara S, et al. Immunohistochemical findings in the pancreatic islets of a patient with transfusional iron overload and diabetes: case report. J Med Invest. 2010;57:345–9.PubMedCrossRef
54.
go back to reference MacDonald MJ, Cook JD, Epstein ML, Flowers CH. Large amount of (apo)ferritin in the pancreatic insulin cell and its stimulation by glucose. FASEB J. 1994;8:777–81.PubMed MacDonald MJ, Cook JD, Epstein ML, Flowers CH. Large amount of (apo)ferritin in the pancreatic insulin cell and its stimulation by glucose. FASEB J. 1994;8:777–81.PubMed
55.
go back to reference Tiedge M, Lortz S, Drinkgern J, Lenzen S. Relation between antioxidant enzyme gene expression and antioxidative defense status of insulin-producing cells. Diabetes. 1997;46:1733–42.PubMedCrossRef Tiedge M, Lortz S, Drinkgern J, Lenzen S. Relation between antioxidant enzyme gene expression and antioxidative defense status of insulin-producing cells. Diabetes. 1997;46:1733–42.PubMedCrossRef
56.
go back to reference Rumberger JM, Peters Jr T. Burrington C, Green A: Transferrin and iron contribute to the lipolytic effect of serum in isolated adipocytes. Diabetes. 2004;53:2535–41.PubMedCrossRef Rumberger JM, Peters Jr T. Burrington C, Green A: Transferrin and iron contribute to the lipolytic effect of serum in isolated adipocytes. Diabetes. 2004;53:2535–41.PubMedCrossRef
57.
go back to reference Green A, Basile R, Rumberger JM. Transferrin and iron induce insulin resistance of glucose transport in adipocytes. Metabolism. 2006;55:1042–5.PubMedCrossRef Green A, Basile R, Rumberger JM. Transferrin and iron induce insulin resistance of glucose transport in adipocytes. Metabolism. 2006;55:1042–5.PubMedCrossRef
58.
go back to reference Niederau C, Berger M, Stremmel W, et al. Hyperinsulinaemia in non-cirrhotic haemochromatosis: impaired hepatic insulin degradation? Diabetologia. 1984;26:441–4.PubMedCrossRef Niederau C, Berger M, Stremmel W, et al. Hyperinsulinaemia in non-cirrhotic haemochromatosis: impaired hepatic insulin degradation? Diabetologia. 1984;26:441–4.PubMedCrossRef
59.
go back to reference Bertelsen M, Anggard EE, Carrier MJ. Oxidative stress impairs insulin internalization in endothelial cells in vitro. Diabetologia. 2001;44:605–13.PubMedCrossRef Bertelsen M, Anggard EE, Carrier MJ. Oxidative stress impairs insulin internalization in endothelial cells in vitro. Diabetologia. 2001;44:605–13.PubMedCrossRef
60.
go back to reference Krawczyk M, Bonfrate L, Portincasa P. Nonalcoholic fatty liver disease. Best Pract Res Clin Gastroenterol. 2010;24:695–708.PubMedCrossRef Krawczyk M, Bonfrate L, Portincasa P. Nonalcoholic fatty liver disease. Best Pract Res Clin Gastroenterol. 2010;24:695–708.PubMedCrossRef
61.
go back to reference Neuschwander-Tetri BA, Clark JM, Bass NM, et al. Clinical, laboratory and histological associations in adults with nonalcoholic fatty liver disease. Hepatology. 2010;52:913–24.PubMedCrossRef Neuschwander-Tetri BA, Clark JM, Bass NM, et al. Clinical, laboratory and histological associations in adults with nonalcoholic fatty liver disease. Hepatology. 2010;52:913–24.PubMedCrossRef
62.
go back to reference Nelson JE, Kowdley KV. Reply to Manco et al: The wide spectrum of hepatic iron overload. Hepatology. 2011;53:1057–8.CrossRef Nelson JE, Kowdley KV. Reply to Manco et al: The wide spectrum of hepatic iron overload. Hepatology. 2011;53:1057–8.CrossRef
63.
go back to reference Mitsuyoshi H, Yasui K, Harano Y, et al. Analysis of hepatic genes involved in the metabolism of fatty acids and iron in nonalcoholic fatty liver disease. Hepatol Res. 2009;39:366–73.PubMedCrossRef Mitsuyoshi H, Yasui K, Harano Y, et al. Analysis of hepatic genes involved in the metabolism of fatty acids and iron in nonalcoholic fatty liver disease. Hepatol Res. 2009;39:366–73.PubMedCrossRef
64.
go back to reference Graham RM, Chua AC, Carter KW, et al. Hepatic iron loading in mice increases cholesterol biosynthesis. Hepatology. 2010;52:462–71.PubMedCrossRef Graham RM, Chua AC, Carter KW, et al. Hepatic iron loading in mice increases cholesterol biosynthesis. Hepatology. 2010;52:462–71.PubMedCrossRef
65.
go back to reference Petrak J, Myslivcova D, Man P, et al. Proteomic analysis of hepatic iron overload in mice suggests dysregulation of urea cycle, impairment of fatty acid oxidation, and changes in the methylation cycle. Am J Physiol Gastrointest Liver Physiol. 2007;292:G1490–8.PubMedCrossRef Petrak J, Myslivcova D, Man P, et al. Proteomic analysis of hepatic iron overload in mice suggests dysregulation of urea cycle, impairment of fatty acid oxidation, and changes in the methylation cycle. Am J Physiol Gastrointest Liver Physiol. 2007;292:G1490–8.PubMedCrossRef
66.
go back to reference Kirsch R, Sijtsema HP, Tlali M, et al. Effects of iron overload in a rat nutritional model of non-alcoholic fatty liver disease. Liver Int. 2006;26:1258–67.PubMedCrossRef Kirsch R, Sijtsema HP, Tlali M, et al. Effects of iron overload in a rat nutritional model of non-alcoholic fatty liver disease. Liver Int. 2006;26:1258–67.PubMedCrossRef
67.
go back to reference Silva M, Silva ME, de Paula H, et al. Iron overload alters glucose homeostasis, causes liver steatosis, and increases serum triacylglycerols in rats. Nutr Res. 2008;28:391–8.PubMedCrossRef Silva M, Silva ME, de Paula H, et al. Iron overload alters glucose homeostasis, causes liver steatosis, and increases serum triacylglycerols in rats. Nutr Res. 2008;28:391–8.PubMedCrossRef
68.
go back to reference Hevi S, Chuck SL. Ferritins can regulate the secretion of apolipoprotein B. J Biol Chem. 2003;278:31924–9.PubMedCrossRef Hevi S, Chuck SL. Ferritins can regulate the secretion of apolipoprotein B. J Biol Chem. 2003;278:31924–9.PubMedCrossRef
69.
go back to reference Bonkovsky HL, Lambrecht RW, Shan Y. Iron as a co-morbid factor in nonhemochromatotic liver disease. Alcohol. 2003;30:137–44.PubMedCrossRef Bonkovsky HL, Lambrecht RW, Shan Y. Iron as a co-morbid factor in nonhemochromatotic liver disease. Alcohol. 2003;30:137–44.PubMedCrossRef
70.
go back to reference Fargion S, Valenti L, Fracanzani AL. Beyond hereditary hemochromatosis: new insights into the relationship between iron overload and chronic liver diseases. Dig Liver Dis. 2011;43:89–95.PubMedCrossRef Fargion S, Valenti L, Fracanzani AL. Beyond hereditary hemochromatosis: new insights into the relationship between iron overload and chronic liver diseases. Dig Liver Dis. 2011;43:89–95.PubMedCrossRef
71.
go back to reference Bonkovsky HL, Jawaid Q, Tortorelli K, et al. Non-alcoholic steatohepatitis and iron: increased prevalence of mutations of the HFE gene in non-alcoholic steatohepatitis. J Hepatol. 1999;31:421–9.PubMedCrossRef Bonkovsky HL, Jawaid Q, Tortorelli K, et al. Non-alcoholic steatohepatitis and iron: increased prevalence of mutations of the HFE gene in non-alcoholic steatohepatitis. J Hepatol. 1999;31:421–9.PubMedCrossRef
72.
go back to reference Fargion S, Mattioli M, Fracanzani AL, et al. Hyperferritinemia, iron overload, and multiple metabolic alterations identify patients at risk for nonalcoholic steatohepatitis. Am J Gastroenterol. 2001;96:2448–55.PubMedCrossRef Fargion S, Mattioli M, Fracanzani AL, et al. Hyperferritinemia, iron overload, and multiple metabolic alterations identify patients at risk for nonalcoholic steatohepatitis. Am J Gastroenterol. 2001;96:2448–55.PubMedCrossRef
73.
go back to reference Nelson JE, Bhattacharya R, Lindor KD, et al. HFE C282Y mutations are associated with advanced hepatic fibrosis in Caucasians with nonalcoholic steatohepatitis. Hepatology. 2007;46:723–9.PubMedCrossRef Nelson JE, Bhattacharya R, Lindor KD, et al. HFE C282Y mutations are associated with advanced hepatic fibrosis in Caucasians with nonalcoholic steatohepatitis. Hepatology. 2007;46:723–9.PubMedCrossRef
74.
go back to reference Valenti L, Dongiovanni P, Fracanzani AL, et al. Increased susceptibility to nonalcoholic fatty liver disease in heterozygotes for the mutation responsible for hereditary hemochromatosis. Dig Liver Dis. 2003;35:172–8.PubMedCrossRef Valenti L, Dongiovanni P, Fracanzani AL, et al. Increased susceptibility to nonalcoholic fatty liver disease in heterozygotes for the mutation responsible for hereditary hemochromatosis. Dig Liver Dis. 2003;35:172–8.PubMedCrossRef
75.
go back to reference Deguti MM, Sipahi AM, Gayotto LC, et al. Lack of evidence for the pathogenic role of iron and HFE gene mutations in Brazilian patients with nonalcoholic steatohepatitis. Braz J Med Biol Res. 2003;36:739–45.PubMedCrossRef Deguti MM, Sipahi AM, Gayotto LC, et al. Lack of evidence for the pathogenic role of iron and HFE gene mutations in Brazilian patients with nonalcoholic steatohepatitis. Braz J Med Biol Res. 2003;36:739–45.PubMedCrossRef
76.
go back to reference Yamauchi N, Itoh Y, Tanaka Y, et al. Clinical characteristics and prevalence of GB virus C, SEN virus, and HFE gene mutation in Japanese patients with nonalcoholic steatohepatitis. J Gastroenterol. 2004;39:654–60.PubMedCrossRef Yamauchi N, Itoh Y, Tanaka Y, et al. Clinical characteristics and prevalence of GB virus C, SEN virus, and HFE gene mutation in Japanese patients with nonalcoholic steatohepatitis. J Gastroenterol. 2004;39:654–60.PubMedCrossRef
77.
go back to reference Yoneda M, Nozaki Y, Endo H, et al. Serum ferritin is a clinical biomarker in Japanese patients with nonalcoholic steatohepatitis (NASH) independent of HFE gene mutation. Dig Dis Sci. 2010;55:808–14.PubMedCrossRef Yoneda M, Nozaki Y, Endo H, et al. Serum ferritin is a clinical biomarker in Japanese patients with nonalcoholic steatohepatitis (NASH) independent of HFE gene mutation. Dig Dis Sci. 2010;55:808–14.PubMedCrossRef
78.
go back to reference Lin TJ, Lin CL, Wang CS, et al. Prevalence of HFE mutations and relation to serum iron status in patients with chronic hepatitis C and patients with nonalcoholic fatty liver disease in Taiwan. World J Gastroenterol. 2005;11:3905–8.PubMed Lin TJ, Lin CL, Wang CS, et al. Prevalence of HFE mutations and relation to serum iron status in patients with chronic hepatitis C and patients with nonalcoholic fatty liver disease in Taiwan. World J Gastroenterol. 2005;11:3905–8.PubMed
79.
go back to reference Dhillon BK, Das R, Garewal G, et al. Frequency of primary iron overload and HFE gene mutations (C282Y, H63D and S65C) in chronic liver disease patients in north India. World J Gastroenterol. 2007;13:2956–9.PubMed Dhillon BK, Das R, Garewal G, et al. Frequency of primary iron overload and HFE gene mutations (C282Y, H63D and S65C) in chronic liver disease patients in north India. World J Gastroenterol. 2007;13:2956–9.PubMed
80.
go back to reference Duseja A, Das R, Nanda M, et al. Nonalcoholic steatohepatitis in Asian Indians is neither associated with iron overload nor with HFE gene mutations. World J Gastroenterol. 2005;11:393–5.PubMed Duseja A, Das R, Nanda M, et al. Nonalcoholic steatohepatitis in Asian Indians is neither associated with iron overload nor with HFE gene mutations. World J Gastroenterol. 2005;11:393–5.PubMed
81.
go back to reference Lee SH, Jeong SH, Lee D, et al. An epidemiologic study on the incidence and significance of HFE mutations in a Korean cohort with nonalcoholic fatty liver disease. J Clin Gastroenterol. 2010;44:e154–61.PubMedCrossRef Lee SH, Jeong SH, Lee D, et al. An epidemiologic study on the incidence and significance of HFE mutations in a Korean cohort with nonalcoholic fatty liver disease. J Clin Gastroenterol. 2010;44:e154–61.PubMedCrossRef
82.
go back to reference • Hernaez R, Yeung E, Clark JM, et al.: Hemochromatosis gene and nonalcoholic fatty liver disease: A systematic review and meta-analysis. J Hepatol 2011. A large meta-analysis of HFE genotyping studies in NAFLD. The authors conclude that HFE mutations are not more common among Caucasian patients with NAFLD than those without NAFLD. • Hernaez R, Yeung E, Clark JM, et al.: Hemochromatosis gene and nonalcoholic fatty liver disease: A systematic review and meta-analysis. J Hepatol 2011. A large meta-analysis of HFE genotyping studies in NAFLD. The authors conclude that HFE mutations are not more common among Caucasian patients with NAFLD than those without NAFLD.
83.
go back to reference • Petrak J, Myslivcova D, Halada P, et al.: Iron-independent specific protein expression pattern in the liver of HFE-deficient mice. Int J Biochem Cell Biol 2007, 39:1006–1015. A novel study showing unique hepatic protein profiles in HFE-deficient mice compared to iron matched contol mice. The implications of this study are that HFE mutations may effect liver function independent of iron accumulation.PubMedCrossRef • Petrak J, Myslivcova D, Halada P, et al.: Iron-independent specific protein expression pattern in the liver of HFE-deficient mice. Int J Biochem Cell Biol 2007, 39:1006–1015. A novel study showing unique hepatic protein profiles in HFE-deficient mice compared to iron matched contol mice. The implications of this study are that HFE mutations may effect liver function independent of iron accumulation.PubMedCrossRef
84.
go back to reference Wu Y, Fan Y, Xue B, et al. Human glutathione S-transferase P1-1 interacts with TRAF2 and regulates TRAF2-ASK1 signals. Oncogene. 2006;25:5787–800.PubMedCrossRef Wu Y, Fan Y, Xue B, et al. Human glutathione S-transferase P1-1 interacts with TRAF2 and regulates TRAF2-ASK1 signals. Oncogene. 2006;25:5787–800.PubMedCrossRef
85.
go back to reference Redinbo MR, Bencharit S, Potter PM. Human carboxylesterase 1: from drug metabolism to drug discovery. Biochem Soc Trans. 2003;31:620–4.PubMedCrossRef Redinbo MR, Bencharit S, Potter PM. Human carboxylesterase 1: from drug metabolism to drug discovery. Biochem Soc Trans. 2003;31:620–4.PubMedCrossRef
86.
go back to reference Henkel C, Roderfeld M, Weiskirchen R, et al. Identification of fibrosis-relevant proteins using DIGE (difference in gel electrophoresis) in different models of hepatic fibrosis. Z Gastroenterol. 2005;43:23–9.PubMedCrossRef Henkel C, Roderfeld M, Weiskirchen R, et al. Identification of fibrosis-relevant proteins using DIGE (difference in gel electrophoresis) in different models of hepatic fibrosis. Z Gastroenterol. 2005;43:23–9.PubMedCrossRef
87.
go back to reference Chu R, Lim H, Brumfield L, et al. Protein profiling of mouse livers with peroxisome proliferator-activated receptor alpha activation. Mol Cell Biol. 2004;24:6288–97.PubMedCrossRef Chu R, Lim H, Brumfield L, et al. Protein profiling of mouse livers with peroxisome proliferator-activated receptor alpha activation. Mol Cell Biol. 2004;24:6288–97.PubMedCrossRef
88.
go back to reference Kallwitz ER, McLachlan A, Cotler SJ. Role of peroxisome proliferators-activated receptors in the pathogenesis and treatment of nonalcoholic fatty liver disease. World J Gastroenterol. 2008;14:22–8.PubMedCrossRef Kallwitz ER, McLachlan A, Cotler SJ. Role of peroxisome proliferators-activated receptors in the pathogenesis and treatment of nonalcoholic fatty liver disease. World J Gastroenterol. 2008;14:22–8.PubMedCrossRef
89.
go back to reference Milet J, Dehais V, Bourgain C, et al. Common variants in the BMP2, BMP4, and HJV genes of the hepcidin regulation pathway modulate HFE hemochromatosis penetrance. Am J Hum Genet. 2007;81:799–807.PubMedCrossRef Milet J, Dehais V, Bourgain C, et al. Common variants in the BMP2, BMP4, and HJV genes of the hepcidin regulation pathway modulate HFE hemochromatosis penetrance. Am J Hum Genet. 2007;81:799–807.PubMedCrossRef
90.
go back to reference Pietrangelo A. The penetrance of hemochromatosis: mice to the rescue. Gastroenterology. 2007;132:805–8.PubMedCrossRef Pietrangelo A. The penetrance of hemochromatosis: mice to the rescue. Gastroenterology. 2007;132:805–8.PubMedCrossRef
91.
go back to reference Facchini FS, Hua NW, Stoohs RA. Effect of iron depletion in carbohydrate-intolerant patients with clinical evidence of nonalcoholic fatty liver disease. Gastroenterology. 2002;122:931–9.PubMedCrossRef Facchini FS, Hua NW, Stoohs RA. Effect of iron depletion in carbohydrate-intolerant patients with clinical evidence of nonalcoholic fatty liver disease. Gastroenterology. 2002;122:931–9.PubMedCrossRef
92.
go back to reference Valenti L, Fracanzani AL, Fargion S. Effect of iron depletion in patients with nonalcoholic fatty liver disease without carbohydrate intolerance. Gastroenterology. 2003;124:866–7.PubMedCrossRef Valenti L, Fracanzani AL, Fargion S. Effect of iron depletion in patients with nonalcoholic fatty liver disease without carbohydrate intolerance. Gastroenterology. 2003;124:866–7.PubMedCrossRef
93.
go back to reference Valenti L, Fracanzani AL, Dongiovanni P, et al. Iron depletion by phlebotomy improves insulin resistance in patients with nonalcoholic fatty liver disease and hyperferritinemia: evidence from a case–control study. Am J Gastroenterol. 2007;102:1251–8.PubMedCrossRef Valenti L, Fracanzani AL, Dongiovanni P, et al. Iron depletion by phlebotomy improves insulin resistance in patients with nonalcoholic fatty liver disease and hyperferritinemia: evidence from a case–control study. Am J Gastroenterol. 2007;102:1251–8.PubMedCrossRef
94.
go back to reference Sumida Y, Kanemasa K, Fukumoto K, et al. Effect of iron reduction by phlebotomy in Japanese patients with nonalcoholic steatohepatitis: A pilot study. Hepatol Res. 2006;36:315–21.PubMedCrossRef Sumida Y, Kanemasa K, Fukumoto K, et al. Effect of iron reduction by phlebotomy in Japanese patients with nonalcoholic steatohepatitis: A pilot study. Hepatol Res. 2006;36:315–21.PubMedCrossRef
95.
go back to reference Valenti L, Moscatiello S, Vanni E, et al. Venesection for non-alcoholic fatty liver disease unresponsive to lifestyle counselling–a propensity score-adjusted observational study. QJM. 2011;104:141–9.PubMedCrossRef Valenti L, Moscatiello S, Vanni E, et al. Venesection for non-alcoholic fatty liver disease unresponsive to lifestyle counselling–a propensity score-adjusted observational study. QJM. 2011;104:141–9.PubMedCrossRef
96.
go back to reference • Dongiovanni P, Valenti L, Ludovica FA, et al.: Iron depletion by deferoxamine up-regulates glucose uptake and insulin signaling in hepatoma cells and in rat liver. Am J Pathol 2008, 172:738–747. Another novel study providing evidence that iron effects glucose metabolism and insulin signaling and that these effects are mediated by HIF-1α. PubMedCrossRef • Dongiovanni P, Valenti L, Ludovica FA, et al.: Iron depletion by deferoxamine up-regulates glucose uptake and insulin signaling in hepatoma cells and in rat liver. Am J Pathol 2008, 172:738–747. Another novel study providing evidence that iron effects glucose metabolism and insulin signaling and that these effects are mediated by HIF-1α. PubMedCrossRef
97.
go back to reference Kaji K, Yoshiji H, Kitade M, et al. Combination treatment of angiotensin II type I receptor blocker and new oral iron chelator attenuates progression of nonalcoholic steatohepatitis in rats. Am J Physiol Gastrointest Liver Physiol. 2011;300:G1094–104.PubMedCrossRef Kaji K, Yoshiji H, Kitade M, et al. Combination treatment of angiotensin II type I receptor blocker and new oral iron chelator attenuates progression of nonalcoholic steatohepatitis in rats. Am J Physiol Gastrointest Liver Physiol. 2011;300:G1094–104.PubMedCrossRef
Metadata
Title
Iron Metabolism in Nonalcoholic Fatty Liver Disease
Authors
James E. Nelson
Heather Klintworth
Kris V. Kowdley
Publication date
01-02-2012
Publisher
Current Science Inc.
Published in
Current Gastroenterology Reports / Issue 1/2012
Print ISSN: 1522-8037
Electronic ISSN: 1534-312X
DOI
https://doi.org/10.1007/s11894-011-0234-4

Other articles of this Issue 1/2012

Current Gastroenterology Reports 1/2012 Go to the issue

Liver (BR Bacon, Section Editor)

Diagnosis and Treatment of Autoimmune Hepatitis

Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine