Molecular pathogenesis and clinical consequences of iron overload in liver cirrhosis
 
Katarzyna Sikorska, Agnieszka Bernat and Anna Wróblewska
Gdynia, Poland
 
 
Author Affiliations: Department of Tropical Medicine and Epidemiology, Medical University of Gdansk, Powstania Styczniowego 9b, 81-519 Gdynia, Poland (Sikorska K); Pomeranian Center of Infectious Diseases and Tuberculosis, Smoluchowskiego 18, 80-214 Gdansk, Poland (Sikorska K); Intercollegiate Faculty of Biotechnology UG-MUG, Laboratory of Molecular Diagnostics, Kladki 24, 80-822 Gdansk, Poland (Bernat A and Wróblewska A)
Corresponding Author: Katarzyna Sikorska, MD, PhD, Department of Tropical Medicine and Epidemiology, Medical University of Gdansk, Powstania Styczniowego 9b, 81-519, Gdynia, Poland (Tel/Fax: +48-58-3491760; Email: ksikorska@gumed.edu.pl)
 
© 2016, Hepatobiliary Pancreat Dis Int. All rights reserved.
doi: 10.1016/S1499-3872(16)60135-2
Published online September 16, 2016.
 
 
Contributors: SK and WA conceived the article, reviewed the literature and drafted the manuscript. BA contributed to drafting manuscript. All authors contributed to the design and interpretation of the study and to further drafts. SK is the guarantor.
Funding: This study was supported by a grant from Polish National Science Centre (2011/01/B/NZ6/00320).
Ethical approval: Not needed.
Competing interest: No benefits in any form have been received or will be received from a commercial party related directly or indirectly to the subject of this article.
 
 
BACKGROUND: The liver, as the main iron storage compartment and the place of hepcidin synthesis, is the central organ involved in maintaining iron homeostasis in the body. Excessive accumulation of iron is an important risk factor in liver disease progression to cirrhosis and hepatocellular carcinoma. Here, we review the literature on the molecular pathogenesis of iron overload and its clinical consequences in chronic liver diseases.
 
DATA SOURCES: PubMed was searched for English-language articles on molecular genesis of primary and secondary iron overload, as well as on their association with liver disease progression. We have also included literature on adjuvant therapeutic interventions aiming to alleviate detrimental effects of excessive body iron load in liver cirrhosis.
 
RESULTS: Excess of free, unbound iron induces oxidative stress, increases cell sensitivity to other detrimental factors, and can directly affect cellular signaling pathways, resulting in accelerated liver disease progression. Diagnosis of liver cirrhosis is, in turn, often associated with the identification of a pathological accumulation of iron, even in the absence of genetic background of hereditary hemochromatosis. Iron depletion and adjuvant therapy with antioxidants are shown to cause significant improvement of liver functions in patients with iron overload. Phlebotomy can have beneficial effects on liver histology in patients with excessive iron accumulation combined with compensated liver cirrhosis of different etiology.
 
CONCLUSION: Excessive accumulation of body iron in liver cirrhosis is an important predictor of liver failure and available data suggest that it can be considered as target for adjuvant therapy in this condition.
 
(Hepatobiliary Pancreat Dis Int 2016;15:461-479)
 
KEY WORDS: liver cirrhosis; fibrosis; iron overload; hepatocelullar carcinoma; hereditary hemochromatosis
 
 
Introduction
Iron is an essential micronutrient for basic metabolic reactions in living organisms. Iron as a component of many molecules participates in basic metabolic reactions in living organisms. As a non-heme iron it is embedded in the iron-sulfur (Fe-S) centers of enzymes which regulate the expression of many genes encoding enzymes involved in the proliferation and differentiation of cells (protein kinase C-β, isozyme5, acid phosphatase, and p21).[1] High redox potential of iron implicates its toxicity associated with the presence of iron ions in different oxidation states (II to VI), mostly in the form of divalent or trivalent ions (Fe2+ or Fe3+ respectively). Disturbed iron balance leads to an overproduction of toxic reactive oxygen species (ROS), increased oxidative stress, peroxidation of the lipid membrane, protein and DNA resulting in damage to cellular structures which triggers the mechanisms of cell death, i.e. necrosis or apoptosis.[2, 3]
 
Iron accumulates in many tissues (lung, kidney, heart, pancreas, and endocrine glands), but the bone marrow and liver remain the main reservoirs. Since the liver is an iron storage compartment and site of hepcidin synthesis, it is involved in maintaining iron homeostasis in the body and rapidly shows pathology in a case of excessive accumulation of iron. The risk of progression of liver fibrosis and development of cirrhosis finally with symptoms of liver failure is associated with duration of the exposure to excess iron in hepatocytes.[4] Liver iron overload is also significantly correlated with an increased risk of developing hepatocellular carcinoma (HCC).[5] On the other hand, the diagnosis of liver cirrhosis is attributed to signs of pathological accumulation of iron with relatively frequent presence of stainable liver iron. It has been shown in one-third of explanted cirrhotic livers, and in 10% of cases the intensity of iron overload corresponded to the diagnostic criteria for hereditary hemochromatosis (HH).[6] Advancements in knowledge of the molecular mechanisms involved in the control of iron metabolism in the human body have been made in the past 15 years, and have contributed to charting new directions of research into the pathogenesis of iron accumulation in liver cirrhosis, its clinical consequences, and possible therapies.
 
 
The role of the liver in the regulation of iron uptake
Hepatocytes serve as a major storage site of iron and are the predominant cells that synthesize hepcidin. Hepcidin, the “iron hormone”, regulates iron transport out of the cell and plays a role as an important controller of iron content in the extracellular space. Iron in forms of both bound and unbound to transferrin (NTBI: non-transferrin bound iron) is captured by hepatocytes. Transferrin receptors (TfR) 1 and 2, present on the membrane of hepatocytes and showing only a slight degree of similarity, are involved in the uptake of transferrin-bound iron. TfR2 expression in hepatocytes is higher compared to TfR1 and its role becomes clearly predominant with an increasing amount of iron in the blood.[7-9] Only TfR1 synthesis is controlled by iron regulatory proteins (IRPs) and iron responsive elements (IREs), and depends on iron content in the cell. Even when the TfR1 transcript is degraded and its synthesis inhibited in a case of increasing intracellular iron content, the intrahepatocytic system of iron uptake mediated by TfR2 is still possible. According to one of the hypotheses TfR2 acts as an iron sensor through the interaction with hemochromatosis protein (HFE) and signaling pathways for transcription factors involved in synthesis of hepcidin. This is why the liver plays such a significant role in protecting against the toxicity of iron and is significantly involved in the pathogenesis of excessive accumulation of iron, i.e. in HH.[10]
 
NTBI is a highly toxic form of iron that appears in the serum with a drastic increase in transferrin saturation and when iron binding capacity is exceeded. It is particularly hazardous because of the ease of generating ROS. The quick clearance of NTBI from the serum is possible by transporting it into hepatocytes through divalent metal transporter 1 (DMT1).[11] In hepatocytes, iron is incorporated into the ferritin molecule. That compensates for cell damage and preserves iron bioavailability. With increased demand for iron in the organism (e.g. intensified erythropoiesis), it is released from the complex with ferritin and transported out of the cell through the molecular mechanism which has been not fully understood. “Iron-poor” ferritin is also present in the serum and its concentration increases in case of tissue iron overload.[12] Therefore serum ferritin serves as a useful clinical marker of pathological iron accumulation. According to recent discoveries ferritin may be a part of cellular protection under conditions of stress and inflammation. Ferritin synthesis is also regulated by the inflammatory response and activity of cytokines (TNF-α, IL-1α, IL-1β and IL-6), oxidants, growth factors (IGF-1), and hormones [T3, thyrotropin-releasing hormone (TRH) and insulin].[13] Recently, Meyron-Holtz et al[14] gathered data confirming that ferritin may be polarized in epithelial barrier cells and play a role also as an iron transporter, including export out of the cell.
 
 
Molecular pathogenesis of iron overload
The role of hepcidin in the regulation of iron uptake, transport and storage
Although hepcidin was discovered as an antibacterial peptide, soon it was demonstrated that the expression of hepcidin mRNA is strongly associated with liver iron content.[15, 16] Nicolas et al[17] observed massive iron overload after inactivation of the gene encoding for mouse hepcidin (HAMP). In turn, hypochromic life-threatening anemia caused by iron deficiency was observed in transgenic mice with HAMP gene overexpression.[17] In humans, a defect in the synthesis of hepcidin in carriers of HAMP gene mutations causes excessive accumulation of iron, revealed at a young age in the form of multi-organ damage and classified as juvenile HH type 2A.[18] This is an autosomal recessive disease, where the rapid progression of iron overload is accompanied by severe organ dysfunction, especially the heart and endocrine glands. Studies using cellular models have led to the determination of the hepcidin mechanism of action.[19] The basis of its activity is direct binding to ferroportin (Fpn), which acts as a membrane receptor for hepcidin. The strongest expression of Fpn in mammals is observed in cells which are the most important for systemic iron homeostasis: enterocytes, hepatocytes, macrophages of the reticuloendothelial system, and syncytiotrophoblasts of the placenta.[20] The activity of Fpn depends on the content of iron in the cell and is mediated by IRP-IRE binding. As a result of the attachment of hepcidin to the Fpn molecule, Fpn is endocytosed and degraded in lysosomes. Consequently, the release of iron from macrophages is blocked and iron absorption from the gastrointestinal tract into the blood is reduced due to the inhibition of iron excretion from the basolateral enterocyte membrane. These changes in iron turnover lead to a decrease in the concentration of iron in the blood.
 
Signaling pathways which regulate the transcription of hepcidin are strictly controlled. Hepcidin expression is stimulated by inflammation through STAT3 signaling dependent on IL-6 and an increased iron content. The transcriptional regulation of hepcidin depends also on another pathway, which is sensitive to the iron concentration and includes the activation of bone morphogenetic protein (BMP), mainly BMP6 signaling with subsequent phosphorylation of intracellular Smad proteins. BMPs belong to the TGF-β family of peptide growth factors and form a complex with membrane receptors (BMPR), which induces the activation of the signaling pathway involving the transcription of Smad proteins, especially Smad4. Smad proteins translocate into the nucleus of the cell where they affect the transcription of specific genes and activate the expression of HAMP, Smad6, Smad7, and Id1.[21-23] The activity of the BMP/Smad system is controlled by regulatory proteins (repulsive guidance molecules; RGMs) that function as co-receptors modulating the binding of BMP with BMPR. One of such cell-specific co-receptor that modulates hepatic BMP/Smad signaling is hemojuvelin (HJV). In 2004, an HJV mutant was described in patients with severe juvenile hemochromatosis (HH type 2B).[24, 25] BMP-related signaling pathways seem to be involved in the modulation of HFE-dependent hepcidin expression as well.[26]
 
Hepcidin expression is also controlled by hypoxia and mechanisms involved in induction of erythropoiesis. According to data published by Chaston et al[27] hypoxia could repress hepcidin expression through inhibition of BMP/Smad signaling. Recently Sonnweber et al[28] showed that a novel regulatory pathway associated with platelet derived growth factor (PDGF)-BB is responsible for downregulation of hepatic hepcidin expression in hypoxia.
 
The role of HFE protein in the regulation of iron homeostasis
The HFE protein is the product of a gene located on the short arm of chromosome 6 (6p21.3) which was first cloned in 1996.[29] A missense HFE gene mutation involving the exchange of cysteine for tyrosine, i.e. C282Y (Cys282Tyr), has been identified in over 80% of clinically diagnosed HH patients, so called HFE or classical HH, type 1. The HFE protein consists of 343 amino acids forming a heavy chain, three extracellular domains (α1, α2, and α3), transmembrane domain and a short cytoplasmic domain.[30]
 
It is now understood that a mutation in the gene encoding the HFE protein leads to a reduction in hepcidin expression in the liver, inadequate for the iron stores in the body.[31] However, the complete mechanism of action has not been completely described. HFE appears to mediate the cellular uptake of transferrin-bound iron by interacting with TfR, although alone it has no iron-binding capacity. The distinctive structure of the HFE protein allows binding to the α1 and α2 domains of TfR1 protein and the α3 domain of β2-microglobulin (β2M).[32] As binding sites on TfR1 for HFE and holotransferrin overlap, HFE protein plays a role in the competitive inhibition of the attachment of iron-saturated transferrin to TfR1.[33]
 
Gao et al[34] found that the interaction between HFE and TfR2 is necessary for proper transcriptional regulation of hepcidin in response to holotransferrin. It was suggested that an increase in transferrin saturation and binding of holotransferrin to TfR1 displaces the HFE protein and at the same time stabilizes TfR2 to form the complex Tf/TfR2/HFE. Calzolari et al[35] showed that the binding of HFE with TfR2 induced hepcidin expression by activating the kinase cascade through Erk1, Erk2 and p38 mitogen activated protein kinase. However, the role of the HFE/TfR2 interaction in the induction of hepcidin is still debated. Although the interaction of overexpressed HFE and TFR2 has been found in tissue culture, recent in vivo studies on transgenic mice and observations in cell lines have challenged the above thesis. It is now even postulated that HFE and TfR2 may influence hepcidin regulation independently.[36, 37]
 
Regardless of these findings, a key hypothesis regarding the impact of HFE/TfR on the optimal functioning of the BMP/Smad signaling pathway in the activation of hepcidin expression has been endorsed. When studying liver sections from people diagnosed with HH type 1, no correlation was found between the expression of HAMP, Smad7, and Id1 and the severity of iron accumulation in tissue. This observation was not confirmed in patients with iron deposits free of the HFE mutation.[38] D’Alessio et al[39] observed the interaction between HFE, TfR2, and HJV in iron sensing, and found that HFE, TfR2, and HJV form a multi-protein membrane complex on the surface of hepatocytes to regulate hepcidin expression.
 
Recently, Wu et al[40] proposed a novel molecular model of the interaction of HFE with BMP/Smad signaling. They showed that, in cultured hepatocytes and mouse liver, HFE binds to the BMP type I receptor, thereby inhibiting activin receptor like kinase-3 (ALK3) ubiquitination and proteasomal degradation and increasing ALK3 protein expression and accumulation on the cell surface. In the presence of the two HFE mutations C282Y and H63D, regulation of ALK3 protein ubiquitination was different and the final increase in expression of both: ALK3 cell surface and hepcidin.[40]
 
Homozygous C282Y alleles have the strongest impact on the excessive accumulation of iron in patients with HH. In carriers with both mutated alleles, a significant transformation of the spatial structure of the HFE protein occurs. It has been suggested that the disruption of a disulfide bridge within the α3 domain may interfere with β2M protein binding and prevent the normal interaction of HFE with TfR1.[31] In the Caucasian population, the C282Y mutation at a single allele is detected at a frequency ranging from 1:12 to 1:20, and at both alleles at 1:200 to 1:400. Among Afro-Americans, the mutation is recognized much less frequently, at a ratio of 1:4000.[41] In the European population, the frequency of the C282Y mutation is variable and ranges from 0% in southern Europe to 12.5% in Ireland.[42] The presence of the mutation is not definitely and unambiguously correlated with disease, as not all individuals who are homozygous for C282Y develop symptoms. Homozygous C282Y mutations have been detected in 60% to 96% of patients presenting typical symptoms of HH, among inhabitants of Europe, North America, and Australia.[43] The risk determination of phenotypic expression is very controversial, because the research results are significantly variable. Beutler et al, by comparing the frequency of co-occurrence of the symptoms of HH with HFE gene mutations, suggested only a 1% chance for the phenotypic manifestation.[44] Observations conducted among residents of Europe, USA, Canada, Australia, and New Zealand indicated the prevalence of iron accumulation symptoms in 38% to 50%, and the possibility of developing multi-organ pathology caused by iron storage in 10% to 33% of C282Y homozygotes.[45-47] The risk of a disturbance in iron metabolism is much higher for males (28% vs 1%).[48] The reason for a lack of phenotypic expression is not clearly defined, and extensive research is being conducted to identify other genes and factors that increase the risk of disturbances in iron homeostasis like male gender, hepatotropic virus infections, alcohol consumption, or a co-existing metabolic syndrome.[31, 49] Recent discoveries indicate new gene variants which seem to modify clinical course of HFE-HH. Among them a novel variant in the glyceronephosphate O-acyltransferase (GNPAT) is mentioned. Knockdown of its expression in human hepatoma cells led to downregulation of hepcidin expression. Also the coinheritance of GNPAT.p.D519G in some male C282Y homozygotes was associated with severe iron overload.[50]
 
In approximately 4% of HFE-HH patients, a mixed heterozygocity for the C282Y mutation, in combination with the H63D mutation (His63Asp, i.e. the replacement of histidine with asparagine) has been detected.[51] Unlike the C282Y mutant protein, no H63D mutant-specific aberrations in the synthesis, intracellular transport, expression, or binding to β2M have been revealed.[31] Based on the results of these molecular studies, it was initially assumed that the H63D mutation alone does not lead to pathological iron accumulation. However, the higher frequency of H63D allele detection in the population of Caucasian patients with clinical symptoms of HH when compared with the general population points to the potential involvement of this polymorphism in the pathophysiology of iron storage.[31] The expected risk of pathological accumulation of iron with different degrees of severity in mixed heterozygotes is estimated to vary from 1% to 2%.[52] The H63D mutation itself occurs in Caucasian populations with an incidence of 15% to 20%.[31] In homo- and heterozygous carriers, mild symptoms of iron storage can manifest, mainly in the form of disturbances in biochemical parameters of iron metabolism.[53] The significance of these additional mutations as possibly pathogenic has been raised, as they may affect the risk of developing cirrhosis and HCC in the case of co-existing hepatitis virus infections and excessive alcohol consumption.[54-56] On the other hand, according to different observations, simple heterozygotes for either the C282Y or H63D HFE gene did not present evident iron overload. Morbidity for both HFE simple heterozygote groups was similar to that of HFE wild-type participants.[57]
 
Other HFE polymorphisms have been rarely reported, and they are usually detected in patients with the typical clinical symptoms of HH, with no classical HFE HH genotype confirmed.[42] Some of them may lead to significant dysfunction of the HFE protein.[58]
 
The clinical consequences of TfR2 dysfunction, which result from TfR2 gene mutations, are classified as HH type 3. It is clinically similar to HFE-HH and shares the same inheritance pattern but manifests at an earlier age.[9]
 
Excessive accumulation of iron dependent on variations in the gene encoding Fpn
Fpn is the only well-described cellular transporter of iron.[20, 59] It is assumed that the spatial structure of Fpn consists of 9 or 10 transmembrane helices. Pathogenic mutations in the gene encoding Solute Carrier family 11, member A3 (SLC11A3) appear to be associated with autosomal dominant HH. They cause the second most common genetic iron overload disorder after classical HFE-HH, called HH type 4.[60, 61] The molecular consequences of mutations at different locations are divided into two groups. The Fpn mutations A77D, D157G, V162del, N174I, Q182H, Q248H, G323V, and G490D impact on the ability of the protein to reach the cell surface, lead to the loss of Fpn activity and decreased cellular iron export. Retention of iron occurs in macrophages responsible for recovering iron from old erythrocytes and clinically manifests itself as tissue iron accumulation, hyperferritinemia and low serum transferrin saturation.[62] The decreased iron content in the extracellular space is parallel to the reduction of transferrin saturation in the circulatory system, which becomes a signal to intensify iron absorption from the intestinal tract. The described pathology has been defined as ferroportin disease. The second group of mutations (N144H, Y64N, C326Y/S, S338R and Y501C) is associated with increased activity of ferroportin due to the resistance of ferroportin to inhibition by hepcidin or by decreased hepcidin binding ability or impaired internalization. Insensitivity to hepcidin results in an excessive iron absorption from the gastrointestinal tract and a decrease in the amount of this metal in macrophages. All this leads to an increase in the amount of iron in plasma. Clinically, it is called a non-classical Fpn disease characterized by hyperferritinemia, elevated or normal transferrin saturation, and increased iron accumulation, mainly in hepatocytes, which what makes its clinical picture similar to that observed in other types of HH.[63]
 
As a very rare clinical observation, Japanese authors have described a single-point dominant mutation, A49U, in the IRE at the 5’ end of the mRNA of the H-subunit of ferritin members of one family. They all presented symptoms of non-HFE iron overload with excessive accumulation of iron, increased transferrin saturation, and high serum ferritin concentrations.[64, 65]
 
 
Primary and secondary iron overload
Pathological accumulation of iron in the body can generally be divided into primary, i.e. genetically determined (Table 1), or secondary, resulting from other genetic or acquired diseases (Table 2). HH, called primary or inborn, is one of the most common genetic metabolic diseases, with a prevalence in Caucasian population ranging from 3 to 8 cases per 1000 individuals.[66] It is defined as a set of pathological symptoms accompanying iron accumulation in the parenchymal cells of many organs, including liver, pancreas, heart, gonads, and the pituitary. The most common causes of death diagnosed in patients with HH include liver cirrhosis, HCC, diabetes and cardiovascular failure caused by myocardial iron deposition.[67] Hemochromatosis is caused by mutation(s) in genes whose protein products regulate iron homeostasis (Table 1).[68]
 
Secondary iron overload develops in the course of many hematological disorders such as thalassemia syndromes, pyruvate kinase deficiency, hereditary spherocytosis, and myelodysplastic syndrome. Many of these diseases are associated with a need for frequent blood transfusions, which causes an excessive body iron supply. Additionally, impaired erythropoiesis leads to hypoxia which stimulates the synthesis of erythropoietin, type II transmembrane serine proteinase, and growth differentiation factor 15. These molecules, in turn, suppress hepcidin synthesis, leading to even more profound body iron accumulation.[69-71] Aceruloplasminemia, hypotransferrinemia, and DMT1 deficiency are genetic disorders with an autosomal recessive pattern of inheritance which result in secondary hemosiderosis due to ineffective transport of iron. The mutation underlying aceruloplasminemia is localized in the ceruloplasmin gene, encoding for a multicopper feroxidase that is synthesized in hepatocytes and is required for iron mobilization in cells. Deficiency of ceruloplasmin results in iron deposition associated with retinal degeneration, diabetes mellitus, and neurological symptoms accompanied with hyperferritinemia.[72] Lack of transferrin production in patients with hypotransferrinemia or the presence of non-functional DMT1 protein are associated with severe microcytic anemia, and an adaptive increase in iron absorption, which leads to massive liver iron loading.[73, 74] Other factors associated with secondary iron accumulation include chronic liver diseases such as infection with hepatitis C virus (HCV),[75] alcoholic liver disease,[76, 77] non-alcoholic fatty liver disease (NAFLD),[78] and porphyria cutanea tarda (Table 2).[79] Chronic infection with HCV and NAFLD are among the major causes of liver disease and a serious risk factors for cirrhosis and HCC.[80] Secondary iron overload frequently diagnosed in NAFLD or chronic hepatitis C (CHC) is a significant comorbid factor and it associates with disease progression.[81, 82] The molecular mechanisms underlying dysregulation of iron metabolism has not been completely elucidated. The question why, in the same clinical background, some patients develop secondary iron overload and others do not still remains to be answered. It is known that some genetic factors may be involved.[83-86] Also the benefits of application of iron depletion as an adjuvant therapy in these diseases have been extensively discussed.[87, 88]
 
Iron overload in NAFLD
The pathogenesis of NAFLD is considered a multifactorial process. Unhealthy lifestyle leads to changes in gut microflora, abnormal endocrine function of adipose tissue and disturbances in glucose and fat metabolism in the liver. As a result, a chronic, low-grade inflammation develops which causes further damage leading to liver disease progression to steatohepatitis.[89] Excessive of body iron accumulation which is found in about 30% of patients with NAFLD has been termed dysmetabolic iron overload syndrome (DIOS). DIOS is defined as a co-existence of hepatic steatosis with mild to moderate iron deposition in liver biopsies and elevated serum ferritin level with nearly normal transferrin saturation in patients with insulin resistance.[90] However, the reasons why only a subset of patients with NAFLD shows alterations in iron parameters is at present unclear.
 
Elevated serum ferritin concentration has been found as an independent predictor of advanced hepatic fibrosis in patients with NAFLD, and is also associated with hepatic iron deposition, lobular inflammation, steatosis and hepatocyte ballooning.[82, 91] Parenchymal accumulation of iron in NAFLD was associated with higher risk of a fibrosis stage greater than 1 (OR=1.7; 95% CI: 1.2-2.3).[85] Iron depletion therapy of patients with NAFLD and hyperferritinemia resulted in the improvement of liver histology and a decrease of activity of liver enzymes.[92] In NAFLD multiple factors contribute to development of disturbances in body iron metabolism. Unhealthy, imbalanced diet, rich in free fatty acids (FFA) and monosugars, is considered one of the major risk factors for disease development and progression. It was shown that FFA can specifically bind to ferritin and thereby modulate iron uptake and release by this protein. Binding of intracellular FFA to ferritin would enhance iron mineralization as well as prevent iron release, and probably affect downstream iron signaling.[93] Indeed, recently Dongiovanni and colleagues found that a diet rich in FFA directly affects iron metabolism by inducing expression of IRP1 and TfR1 leading to intracellular accumulation of iron.[94] Diet-induced hepatic iron overload in mice appeared prior to the onset on liver steatosis and insulin resistance.[95] Accumulation of iron in the livers of rats fed a high-fat diet was associated with oxidative stress and increased hepatic production of TNF-α, IL-6 and leptin.[96] This diet-induced increase in hepatocyte iron concentration as well as inflammation stimulate hepcidin production in the liver of NAFLD patients. In morbidly obese individuals this hormone is also synthesized by adipose tissue.[97] Additionally glucose was found to stimulate hepcidin release from pancreatic β cells.[98] The resulting increased serum hepcidin causes a reduction of Fpn expression and a decreased iron export from cells which further stimulates hepatocyte iron deposition. Iron perturbations in NAFLD are also linked with inadequate copper bioavailability. Copper is a co-factor for ceruloplasmin ferroxidase, which in turn mobilizes iron from storage sites and is required for Fpn expression and cell surface stability.[99] Low copper concentration and a diminished ceruloplasmin ferroxidase activity in serum of NAFLD patients was associated with low liver Fpn expression and increased hepatic iron stores.[100]
 
Iron overload in CHC
Elevation of total body iron stores is diagnosed in up to 40% cases of CHC and in 50% of patients with both CHC and HCC.[81, 101] Viral infection interferes with cell iron metabolism and iron, in turn, modulates HCV proliferation. Filebeen and colleagues showed a reduction of intracellular Fe levels, reduced iron uptake and increased iron release capacity in Huh7 cell lines stably expressing a subgenomic HCV replicon.[102] They also found a decreased expression of ceruloplasmin and TfR1 as well as an elevated Fpn mRNA levels in these replicon cells.[102] Higher expression of Fpn was also found in the duodenum and the liver of CHC patients.[103-105] Additionally, viral infection upregulates TfR2 expression and mRNA level of TfR2 decreases significantly in parallel with a decline in the liver iron content after a successful eradication of HCV.[106] Recently Nanba and colleagues found a downregulated expression of iron-sensing protein, F-box and leucine-rich repeat protein 5 in CHC patients in comparison to control despite their significantly higher levels of serum iron and ferritin. This effect was associated with an increased iron deposition.[107]
 
HCV infection increases intracellular generation of ROS by directly affecting mitochondrial metabolism and elevating Nox protein expression.[108] ROS can affect iron metabolism, for example by upregulation of Tfr1 expression and iron uptake by the cell[109] or activation of IRP1.[110] HCV-mediated oxidative stress was also found to decrease hepcidin expression in hepatoma cells.[107,111,112] It has been suggested that this effect is responsible for secondary iron overload observed in CHC.[113, 114] Downregulation of hepcidin by HCV core protein involved ROS generation and a displacement of C/EBPα and STAT3 proteins from the hepcidin promoter,[111, 115] while core+/ARFP protein repressed hepcidin transcription via the activator protein 1 binding site.[112] Also patients with CHC display a decreased hepcidin expression in serum and in the liver in comparison to uninfected controls.[113,114,116] After the successful antiviral treatment patients show significantly higher hepcidin values in comparison with nonresponders to the therapy.[113, 117] Kohjima et al found that lower expression of hepcidin and Fpn in untreated CHC patients associates with SVR after PEG-IFN and ribavirin therapy[105] but other studies seem to question the predictive value of hepcidin on therapeutic outcome.[116, 117]
 
Iron inhibits HCV RNA polymerase NS5B and treatment of hepatoma cells with iron donor suppresses subcellular replication of HCV.[118-120] Also viral replication is blocked in Huh7 cells by siRNA-mediated knockdown of hepcidin.[120, 121] Treatment with iron donor and the inhibition of hepcidin both upregulate intracellular ferritin which, and not free intracellular iron, was postulated to influence viral replication.[120] This in vitro effect of iron on HCV is confusing in the light of clinical data suggesting a clear association of excess hepatic or serum iron with disease progression and treatment outcome in CHC.[81, 83, 122, 123] There is still much discussion on the relevance of these findings for HCV infection management.[124-126]
 
 
Iron is an important factor in liver disease progression to cirrhosis
Liver fibrogenesis is a complex and dynamic tissue and cellular process, occurring in the course of chronic liver disease and leading to a progressive accumulation of extracellular matrix (ECM) components in an attempt to limit the consequences of hepatic damage. Progression of hepatic fibrosis leading to the formation of regenerative nodules of liver parenchyma results in disruption of lobular architecture, impairment of the fine vascular structure, development of portal hypertension, and loss of metabolic liver function with the risk of variceal bleeding, encephalopathy, and kidney insufficiency.[127] Liver cirrhosis also poses a significant risk factor for the development of HCC and represents the most common non-neoplastic cause of death in Europe and USA.[128]
 
The molecular mechanisms of the stimulatory effect of iron overload on the development of fibrosis and cirrhosis are not clear. It is known that intracellular iron catalyzes the formation of several ROS, which promote lipid peroxidation and protein damage.[2, 129] This sensitizes hepatocytes to other stresses, finally leading to mitochondrial dysfunction and apoptosis resulting in degenerative changes in liver tissue (ballooning hepatocytes, steatosis).[130] Apoptotic hepatocytes are subsequently phagocytosed by Kupffer cells, which become activated and synthesize a wide range of pro-inflammatory and pro-fibrotic factors including TNF-α, IL-1, IL-10, IFN-γ, TGF-β1, PDGF, bFGF, MCP-1, and ROS.[127, 131] Activation of Kupffer cells can also occur through the direct action of excessive iron accumulation in these cells. Soluble factors produced by activated Kupffer cells as well as by injured hepatocytes lead to the recruitment and infiltration of macrophages and lymphocytes. Inflammatory infiltration localized in portal and periportal areas is correlated with the intensity of fibrosis in HH patients.[130, 132] Stimulated Kupffer cells and damaged hepatocytes also activate hepatic stellate cells (HSCs), which is considered a crucial step in the initiation of fibrogenesis.[133] HSCs, comprising 13%-15% of all liver cells, reside within the space of Disse and are considered a main source of collagen and other components of ECM in normal and fibrotic liver.[134, 135] Activated HSCs trans-differentiate into myofibroblast-like hepatic stellate cells (HSC-MFs). HSC-MFs, together with other types of myofibroblasts (MF), originating from different types of cells, are highly proliferative and synthesize the components of ECM as well as several growth and immunomodulatory factors, and thus are able to sustain and perpetuate fibrosis, chronic inflammation, and angiogenesis in the liver (Fig.).[136] Iron can directly activate the process of fibrogenesis in the liver as HSCs have been found to be activated by ferritin or transferrin, which both bind to their high-affinity receptors on the cell surface.[137, 138] Additionally, HSC activation in HH patients increases with hepatic iron concentration; this process can be reversed by iron removal by phlebotomy.[139-141]
 
 
Clinical consequences of iron overload in liver cirrhosis
Iron overload and risk of infections and liver failure
The relationship between disturbances in body iron homeostasis and the function of the immune system is a notable issue in the context of clinical repercussions. Infections are common reasons for clinical deterioration and decompensation in liver cirrhosis. In post-transplant medical care, infectious complications may influence survival as negative prognostic factors. Thus, excessive accumulation of iron in liver cirrhosis is perceived as a systemic pathology and warrants a thorough diagnosis and treatment, also taking into account the post-transplantation prognosis. Cellular disorders of iron metabolism, theoretically, may affect the normal stimulation of macrophage differentiation and proliferation of lymphocyte subpopulations, and increase their susceptibility to oxidative stress associated with an increased risk of DNA damage.[142, 143] Lowering the content of iron in phagocytic cells, which is observed with the down-regulation of hepcidin, inhibits the translation of pro-inflammatory cytokines such as TNF-α and IL-6.[144] Also, dendritic cell differentiation, their involvement in antigen presentation, and the induction of specific immune cell populations correlate with a decrease in TfR1 expression and the activity of Fpn.[145] Mice with homozygous disruption of the Hfe gene, resulting in low hepcidin and high expression of macrophage Fpn, appear to be a good model for research focused on the impact of decreased intramacrophage iron on the TLR4-activated inflammatory response. Impairment of TLR4 signaling leading to abnormal cytokine production may result in greater susceptibility to some bacterial infections.[146]
 
There are some interesting observations in carriers of C282Y homozygotes which may partially explain increased susceptibility to bacterial and some viral infections in patients with liver cirrhosis and iron overload. Some studies in patients with HFE-HH show a significant reduction in total lymphocytes, especially in the population of CD8 lymphocytes.[147-149] Based on studies in rodents, some authors suggest that excessive accumulation of iron under the conditions of the experiment did not alter the number of cells, but affected the mechanisms of cytokine regulation.[150, 151] However, in cases of excessive iron accumulation, the number of NK cells and CD8 cells in liver tissue is decreased.[152] Porto et al showed a significant correlation between an increased CD4/CD8 ratio in the blood with the severity of iron storage and liver fibrosis.[153] It has even been suggested that this indicator could be a predictive factor in determining the severity of the illness. Undoubtedly, the impact of iron overload on innate or acquired immunity requires further research, based both on experimental and clinical observations, particularly in view of the rapid progress in studies on cellular iron metabolism.[154, 155]
 
Clinical observations confirm that the serum concentration of ferritin, considered not only as an acute phase protein, but also an indicator of iron overload in patients with cirrhosis, appears to be associated with markers of liver insufficiency.[156] In other research, serum ferritin was described as an independent predictor of mortality and liver-related clinical events in patients awaiting liver transplantation.[157] Moreover, in this study, the authors showed that patients with elevated serum ferritin had more intense iron accumulation in explanted livers. Maiwall et al found the same correlation between higher serum ferritin and early mortality in patients with decompensated cirrhosis, excluding from the study subjects with secondary iron overload.[158] In turn, Abu Rajab et al demonstrated that marked hepatic hemosiderosis secondary to cirrhosis may be associated with the risk of extrahepatic complications, especially pancreatic and cardiac, due to an increased body iron content.[159] Dysregulation of iron homeostasis with elevated serum parameters of iron metabolism and low hepcidin is associated with acute-on-chronic liver failure and multi-organ insufficiency. Maras et al even suggested that the measurement of plasma transferrin saturation could serve as a predictor of poor outcome and early mortality in this group of patients.[160] The results from the National Transplant Registry indicate that hepatic iron overload may be associated with decreased survival after liver transplantation.[161-163]
 
Thus, it seems important for the clinical prognosis to establish both the diagnostic rules for accurate assessment of the iron content in liver cirrhosis and possible methods of safe iron removal therapy. It has been reported in patients who were qualified for liver transplantation that iron reduction with preoperative venesection reduced the risk of cardiac and infection complications postoperatively.[164]
 
Iron and HCC
HCC is the sixth most common cancer in the world and the leading cause of death from malignancy.[165] Liver cirrhosis is the primary risk factor for developing HCC as it is diagnosed in 70%-90% of all cases of HCC worldwide.[166] The microenvironment of a cirrhotic liver, with cytokines, growth factors, and products of oxidative stress released as a result of activation of HSC and Kupffer cells, can stimulate neoplastic transformation.[127] Also, the inhibition of the hepatocyte proliferation rate, observed in cirrhosis, can facilitate clonal expansion of preneoplastic cells in the liver and thus lead to HCC development.[167]
 
Hepatic iron overload is linked with an increased risk of HCC in patients with cirrhosis of different etiology. HCC occurs in approximately 20% of cirrhotic HH patients and is one of the most common causes of death in non-treated HH.[168, 169] In patients with cirrhosis without a determined background, the presence of iron deposition in liver regenerative nodules has been associated with the presence of HCC.[170] Elevated serum transferrin saturation correlates with an increased incidence of cirrhosis and HCC, especially in patients with high alcohol consumption.[171] The prevalence of both iron overload and HCC was found to be high in cirrhotic patients with viral hepatitis C or hepatitis B, but relatively low in patients with biliary cirrhosis.[5, 82] The critical role of the iron-overload state in the development of HCC was shown in patients with CHC. A combination of phlebotomies and low-iron diet significantly reduced the level of hepatic oxidative markers and was suggested to lower the risk of HCC.[172, 173]
 
Excessive hepatic iron can cause neoplastic transformation in the presence of cirrhosis or it can be directly hepatocarcinogenic. Exogenous iron administration is associated with enhanced tumor development and growth in HCC cell lines and murine models.[174-176] Additionally, HCC has been documented to occur in iron-overloaded patients in the absence of cirrhosis, albeit rarely.[177] The majority of diagnosed HCC cases, however, develop as a consequence of cirrhosis, and iron overload accelerates the progression of the disease.
 
Free, unbound iron catalyzes the formation of highly toxic ROS. The accumulation of ROS-mediated DNA damage can facilitate the progression of chronic liver injury to HCC. This process of cell immortalization is accompanied by the induction of telomerase activity and overexpression of pro-oncogenic microRNA miR-92.[178] Oxidative stress in HCC diagnosed in HH patients has also been found to be associated with an increased mutation rate in the p53 tumor suppressor. In fact both, genetic instability and p53 inactivation play critical and cooperative roles in the progression of HCC.[179] The background of cirrhotic liver with chronic cycles of hepatocyte death and regeneration further stimulates disease progression. Iron overload can also act synergistically with other HCC risk factors such as aflatoxin B1 or alcohol, and increase the mutagenesis rate.[180, 181] Chronic severe iron overload resulting in damage to hepatocytes may lead to the epigenetic abnormalities found in HCC. In patients with HH, aberrant hypermethylation has been detected in pre-neoplastic liver tissue. Among the tested genes, the tumor suppressors RASSF1A and p16-CDKN2A, and the cell-cycle regulator cyclin D2 exhibited differential methylation, albeit to a different extent (55%, 45%, and 32%, respectively). Moreover, changes in methylation patterns were observed in the liver tissue adjacent to HCC.[182] In a rodent model of chronic iron overload, both tumor suppressive as well as tumor-promoting mechanisms were found to be activated in liver tissue. Among the pro-carcinogenic mechanisms, a decrease in p53 levels and increases in p52, p46, and β-catenin levels were found, while protective responses included an increase in C/EBPα and a decrease in Yes-associated protein. In this study, no tumors were observed in iron-loaded rats; the authors suspected that this was due to the high resistance of rat models to the development of cirrhosis in response to iron overload, which contrasts with observations made in humans and eliminates this important risk factor for HCC.[183]
 
Nitric oxide (NO) produced by cytokine-stimulated macrophages and hepatocytes can inhibit the proliferation of tumor cells. The non-heme iron content is a key factor modulating the effect of NO on cell viability.[184] Finally, excess iron may prevent efficient removal of arising tumor cells by influencing immune system functions, since exogenous iron reduces the tumoricidal activity of macrophages,[185] inhibits lymphocyte proliferation, and modulates the balance between different populations of T cells.[154, 186]
 
The development of HCC is associated with changes in the expression of iron regulatory genes, as the mRNA levels of HAMP, ceruloplasmin, transferrin, and TfR2 are reduced while TfR1 expression is elevated in HCC.[187] Interestingly, HAMP expression is downregulated in cancerous tissue regardless of the degree of tumor differentiation and the serum concentration of hepcidin-25.[188] Boyault et al identified 16 genes, among them the HAMP gene, whose expression could correctly distinguish between different subgroups of HCC.[189] Hepcidin suppression may be linked with decreased p53 activity, frequently observed in HCC.[179] The promoter region of HAMP gene contains a putative p53 response element and its expression is induced by the activation of p53.[190]
 
 
Targeting iron in treatment of liver cirrhosis
Iron depletion therapy
Reduction of body iron stores in the early stages of hepatic fibrosis can slow the disease progression and thereby improve survival. Phlebotomy is a safe, cheap and easy to perform iron depletion therapy in patients with iron overload. It was proven to be especially effective in HH patients with fibrosis where it leads to normalization of ALT levels, alleviation of some of the associated symptoms of HH such as abdominal pain, malaise, fatigue, skin pigmentation and insulin resistance. However, other clinical features of HH such as arthropathy and hypogonadism respond poorly to iron depletion or are not responsive at all.[141, 191, 192] Among 185 HH patients treated with phlebotomy 42 (23%) had a documented decrease in liver fibrosis stage during a 14-year observation period.[193]
 
Phlebotomy can also be useful in reversing liver fibrosis in a certain types of secondary iron overload. Venesection resulted in a significant reduction in serum procollagen III, a marker of fibrogenesis, in 22% of CHC patients who previously did not respond to IFN monotherapy.[194] Long-term phlebotomy (56±28 months) improved liver function tests as well as histopathology of the liver in 15 out of 30 patients with CHC.[195] Likewise the severity of liver fibrosis decreased significantly during a 5-year phlebotomy maintenance program of CHC patients in comparison to IFN treated, non-responsive control CHC group. At the same time the intensity of liver inflammation remained unchanged as compared to significant progression observed in the controls.[196] The results of phase II clinical trial on effect of phlebotomy in 31 NAFLD patients suggest that this treatment may improve the overall liver histology (measured as NAFLD activity score).[197]
 
Once cirrhosis has developed, iron depletion therapy is generally considered ineffective in the reversal of the fibrosis process. Cirrhotic HH patients have an increased risk of HCC and death regardless of treatment.[191] It is generally accepted, that for patients with liver failure due to cirrhosis or HCC in cirrhotic liver, an orthotopic liver transplantation is the only solution.[191] However, there are some reports on successful therapy by venesection in cirrhotic patients with a diagnosed primary[141, 198] or secondary[199] iron overload. Falize and colleagues reported a regression of advanced fibrosis, defined as a decrease of at least 2 units on a METAVIR scale, during a 2-year period in 17 among 36 HH patients treated with phlebotomy. In this study a subgroup of 23 patients with histological evidence of cirrhosis, 8 subjects presented regression of fibrosis. In this study, authors also defined serum gamma-globulins, prothrombin activity, and platelet count as biochemical parameters which can predict regression of the fibrosis.[141] What was important, they recruited patients with stable liver function, without severe abnormalities in mean values of serum albumin concentration, prothrombin activity which could indicate liver failure. Phlebotomy was shown to improve liver function and thrombocytopenia, a frequent hematological complication of advanced fibrosis, in cirrhotic patients with iron overload.[200] Given this data and the safety of phlebotomy, this treatment should be considered as an important adjuvant therapy in patients with advanced fibrosis, even liver cirrhosis. But in cirrhotic patients an especial attention should be paid to exponents of good liver function. Advanced portal hypertension, unstable erythropoiesis are obvious contraindications to this form of therapy.
 
Indications for therapeutic phlebotomy should take into account not only serum markers of iron overload: elevated iron and ferritin concentrations, transferrin saturation but if possible also the intensity of tissue iron accumulation. The use of histopathological examination to assess liver iron deposits semiquantitatively or quantitative spectrophotometric analysis of iron content in liver biopsy specimens is limited. In case of cirrhosis biopsy samples are not sufficiently representative due to advanced fibrosis with nodular transformation of liver parenchyma. MR imaging appeared to be useful diagnostic procedure to detect liver iron overload in liver cirrhosis.[201] Another method of iron removal, alternative to phlebotomy, hypothetically safer in cirrhotic patients is erythrocytapheresis. The costs of the procedure are the same as with phlebotomy, and because a larger amount of iron can be removed fewer sessions of erythrocytapheresis are required.[88] Generally patients are treated until near-iron depletion is reached that is ferritin concentration 50 ng/mL or lower and serum transferrin saturation below 45%. The subsequent phlebotomy sessions (usually once per 2-3 months) aim to maintain these levels.[88, 197, 200, 202]
 
For patients who cannot tolerate phlebotomy because of anemia or other comorbidities such as cardiac dysfunction, iron chelation therapy could be considered. Among licensed iron chelators deferoxamine must be administered by intravenous or subcutaneous injections, while deferiprone and deferasirox can be given orally.[203] Numerous clinical trials showed that these drugs decrease organ iron toxicity in patients with severe iron overload and dramatically improve their survival. Therefore, despite their unexplained low iron chelation activity in some patients and various side effect (such as agranulocytosis, neutropenia, arthropathy) chelators are the drugs of choice when there is no other iron removal therapy applicable.[204, 205]
 
Just as phlebotomy, iron chelation therapy can improve liver functions. For example early trials showed that deferoxamine prevented progression of hepatic fibrosis in children.[206] Treatment with deferiprone resulted in a reversal of liver fibrosis in 15 out of 17 patients with β-thalassemia with iron overload during a 3-year period.[207] Deugnier et al observed an improvement in liver fibrosis in iron-overloaded β-thalassaemia patients treated with deferasirox for at least 3 years. Liver fibrosis was reversed or stabilized in 83% of 219 patients. Interestingly, this effect was independent of a reduction in the concentration of hepatic iron concentration.[208] Treatment with deferasirox inhibited hepatic fibrogenesis in a rat model of non-alcoholic steatohepatitis.[209] However, in a mutant mouse model of liver fibrosis, apart from reduction of expression of fibrogenic genes, no effect was observed.[210] The reason for this inconsistency might be due to the differences in doses and treatment periods applied in these studies and further research is needed to elucidate the potential antifibrotic activity of deferasirox. Moreover, expanded clinical observational studies are needed to study the risk of fungal infections associated with interaction between iron chelators and iron-uptaking siderophores of zygomycetes (invasive fungi). Unlike deferoxamine, oral iron chelators deferiprone and deferasirox probably may present antifungal activity.[211]
 
Reduction of iron-related oxidative damage
Liver diseases is closely linked with oxidative stress and many antioxidants were shown to exhibit hepatoprotective activity in animal models of liver injury.[212] Considering important role of iron in mediating oxidative damage it is interesting that a number of natural antioxidants also possess iron-chelating properties. For example curcuminoids from Curcuma longa can bind ferric and ferrous ions and were found to decrease oxidative stress and serum NTBI levels in β-thalassemia patients.[213] Polyphenols from green tea exhibited hepatoprotective effect on iron-loaded hepatocytes,[214] and decreased serum iron indices and lipid peroxidation in iron-loaded mice.[215] Drinking tea with meals reduced dietary iron absorption in patients with metabolic syndrome,[216] as well as the frequency of phlebotomies needed for the management of patients with HH.[217] Importantly, green tea extract inhibited or delayed hepatic iron deposition in regularly iron-loaded thalassemic mice.[218] Another well known natural polyphenol, resveratrol, was found to protect the liver from iron-mediated injury. Although resveratrol did not affect the degree of hepatic iron-overload in livers of mouse models of primary and secondary iron overload, it showed significant antiinflammatory, antioxidant, antiapoptotic and antifibrotic activity.[219]
 
Silymarin from Silybum marianum is a complex mixture of phenolic compounds with a recognized hepatoprotective, immunomodulatory and antioxidant properties. Silymarin, and one of its flavonolignan constituents, silybin, bind Fe(III) with high affinity, and have shown to reduce body iron indices in patients with CHC.[220] A number of clinical trials reported significant decrease of serum iron indices in β-thalassemia patients treated with a combination of silymarin with conventional iron-chelators, in comparison with chelator alone.[221] In HFE-HH patients silymarin reduced dietary iron absorption of non-heme iron by over 40%.[222]
 
Although many antioxidants prevent hepatic damage, reduce inflammation and oxidative stress markers, they are very often much less effective for treatment of already established liver disease. So far beneficial effects of antioxidants application for patients with liver cirrhosis have not been unequivocally confirmed, although some studies reported a significant decrease of liver-related mortality rates in treatment groups.[223, 224] According to ClinicalTrials.gov several antioxidants were recently tested in clinical trials as complementary therapeutics in patients with hepatic fibrosis and cirrhosis. However, no study addressed so far any potential beneficial effects of antioxidants with iron chelating properties on patients with liver cirrhosis with elevated body iron stores. It remains to be studied if these compounds could be specifically useful in this group of patients. Nevertheless application of adjuvant therapy with antioxidants in cirrhotic patients should be taken with caution as degree of liver damage associates with changes in bioavailability of some compounds,[225] and adverse effects of particular antioxidants on liver function have also been reported.[226]
 
 
Conclusion
Disturbances in body iron balance occur commonly in liver cirrhosis. They may be of genetic origin like in HH, which is one of the most common genetic metabolic diseases of humans. Iron overload is an important factor in liver disease progression to cirrhosis. In turn, the diagnosis of liver cirrhosis is often attributed to signs of pathological accumulation of iron, even in the absence of genetic abnormalities. Very few research studies have been devoted to the analysis of changes in the expression of iron regulatory proteins in liver cirrhosis. Possible complex disturbances in iron homeostasis may be a consequence of both genetic factors and morphological changes in the liver, including the disruption of lobular architecture, progressive fibrosis, and loss of parenchyma. As iron overload in liver cirrhosis is suggested to associate with the risk of liver failure, severe infections, pancreatic and cardiac complications, and the development of HCC, further studies on the role of iron in advanced liver disease are needed. Better knowledge of the mechanisms associated with disturbed iron homeostasis and its clinical consequences is desirable to implement treatment for iron overload in liver cirrhosis and improve the prognosis in this condition. 
 
 
References
1 Ganz T. Systemic iron homeostasis. Physiol Rev 2013;93:1721-1741. PMID: 24137020
2 Niemelä O, Parkkila S, Britton RS, Brunt E, Janney C, Bacon B. Hepatic lipid peroxidation in hereditary hemochromatosis and alcoholic liver injury. J Lab Clin Med 1999;133:451-460. PMID: 10235128
3 Pietrangelo A. Metals, oxidative stress, and hepatic fibrogenesis. Semin Liver Dis 1996;16:13-30. PMID: 8723320
4 Hübscher SG. Iron overload, inflammation and fibrosis in genetic haemochromatosis. J Hepatol 2003;38:521-525. PMID: 12663247
5 Ko C, Siddaiah N, Berger J, Gish R, Brandhagen D, Sterling RK, et al. Prevalence of hepatic iron overload and association with hepatocellular cancer in end-stage liver disease: results from the National Hemochromatosis Transplant Registry. Liver Int 2007;27:1394-1401. PMID: 17927713
6 Ludwig J, Hashimoto E, Porayko MK, Moyer TP, Baldus WP. Hemosiderosis in cirrhosis: a study of 447 native livers. Gastroenterology 1997;112:882-888. PMID: 9041250
7 Kawabata H, Yang R, Hirama T, Vuong PT, Kawano S, Gombart AF, et al. Molecular cloning of transferrin receptor 2. A new member of the transferrin receptor-like family. J Biol Chem 1999;274:20826-20832. PMID: 10409623
8 Crichton R. Cellular iron uptake in mammals. In: inorganic biochemistry of iron metabolism: from molecular mechanisms to clinical consequences, 3rd ed. John Wiley & Sons; 2009:141-182.
9 Camaschella C, Roetto A. TFR2-related hereditary hemochromatosis. In: Pagon RA, Adam MP, Ardinger HH, Wallace SE, Amemiya A, Bean LJH, Bird TD, Dolan CR, Fong CT, Smith RJH, Stephens K, editors. GeneReviews. Seattle (WA): University of Washington, Seattle; 2011:1993-2015.
10 Fleming RE, Migas MC, Holden CC, Waheed A, Britton RS, Tomatsu S, et al. Transferrin receptor 2: continued expression in mouse liver in the face of iron overload and in hereditary hemochromatosis. Proc Natl Acad Sci U S A 2000;97:2214-2219. PMID: 10681454
11 Brissot P, Ropert M, Le Lan C, Loréal O. Non-transferrin bound iron: a key role in iron overload and iron toxicity. Biochim Biophys Acta 2012;1820:403-410. PMID: 21855608
12 Finazzi D, Arosio P. Biology of ferritin in mammals: an update on iron storage, oxidative damage and neurodegeneration. Arch Toxicol 2014;88:1787-1802. PMID: 25119494
13 Torti FM, Torti SV. Regulation of ferritin genes and protein. Blood 2002;99:3505-3516. PMID: 11986201
14 Meyron-Holtz EG, Cohen LA, Fahoum L, Haimovich Y, Lifshitz L, Magid-Gold I, et al. Ferritin polarization and iron transport across monolayer epithelial barriers in mammals. Front Pharmacol 2014;5:194. PMID: 25202274
15 Park CH, Valore EV, Waring AJ, Ganz T. Hepcidin, a urinary antimicrobial peptide synthesized in the liver. J Biol Chem 2001;276:7806-7810. PMID: 11113131
16 Nicolas G, Bennoun M, Devaux I, Beaumont C, Grandchamp B, Kahn A, et al. Lack of hepcidin gene expression and severe tissue iron overload in upstream stimulatory factor 2 (USF2) knockout mice. Proc Natl Acad Sci U S A 2001;98:8780-8785. PMID: 11447267
17 Nicolas G, Bennoun M, Porteu A, Mativet S, Beaumont C, Grandchamp B, et al. Severe iron deficiency anemia in transgenic mice expressing liver hepcidin. Proc Natl Acad Sci U S A 2002;99:4596-4601. PMID: 11930010
18 Roetto A, Papanikolaou G, Politou M, Alberti F, Girelli D, Christakis J, et al. Mutant antimicrobial peptide hepcidin is associated with severe juvenile hemochromatosis. Nat Genet 2003;33:21-22. PMID: 12469120
19 Nemeth E, Tuttle MS, Powelson J, Vaughn MB, Donovan A, Ward DM, et al. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 2004;306:2090-2093. PMID: 15514116
20 Abboud S, Haile DJ. A novel mammalian iron-regulated protein involved in intracellular iron metabolism. J Biol Chem 2000;275:19906-19912. PMID: 10747949
21 Babitt JL, Huang FW, Wrighting DM, Xia Y, Sidis Y, Samad TA, et al. Bone morphogenetic protein signaling by hemojuvelin regulates hepcidin expression. Nat Genet 2006;38:531-539. PMID: 16604073
22 Andriopoulos B Jr, Corradini E, Xia Y, Faasse SA, Chen S, Grgurevic L, et al. BMP6 is a key endogenous regulator of hepcidin expression and iron metabolism. Nat Genet 2009;41:482-487. PMID: 19252486
23 Meynard D, Kautz L, Darnaud V, Canonne-Hergaux F, Coppin H, Roth MP. Lack of the bone morphogenetic protein BMP6 induces massive iron overload. Nat Genet 2009;41:478-481. PMID: 19252488
24 Papanikolaou G, Samuels ME, Ludwig EH, MacDonald ML, Franchini PL, Dubé MP, et al. Mutations in HFE2 cause iron overload in chromosome 1q-linked juvenile hemochromatosis. Nat Genet 2004;36:77-82. PMID: 14647275
25 Lee PL, Beutler E, Rao SV, Barton JC. Genetic abnormalities and juvenile hemochromatosis: mutations of the HJV gene encoding hemojuvelin. Blood 2004;103:4669-4671. PMID: 14982867
26 Worthen CA, Enns CA. The role of hepatic transferrin receptor 2 in the regulation of iron homeostasis in the body. Front Pharmacol 2014;5:34. PMID: 24639653
27 Chaston TB, Matak P, Pourvali K, Srai SK, McKie AT, Sharp PA. Hypoxia inhibits hepcidin expression in HuH7 hepatoma cells via decreased SMAD4 signaling. Am J Physiol Cell Physiol 2011;300:C888-895. PMID: 21289291
28 Sonnweber T, Nachbaur D, Schroll A, Nairz M, Seifert M, Demetz E, et al. Hypoxia induced downregulation of hepcidin is mediated by platelet derived growth factor BB. Gut 2014;63:1951-1959. PMID: 24598129
29 Feder JN, Gnirke A, Thomas W, Tsuchihashi Z, Ruddy DA, Basava A, et al. A novel MHC class I-like gene is mutated in patients with hereditary haemochromatosis. Nat Genet 1996;13: 399-408. PMID: 8696333
30 Parkkila S, Waheed A, Britton RS, Bacon BR, Zhou XY, Tomatsu S, et al. Association of the transferrin receptor in human placenta with HFE, the protein defective in hereditary hemochromatosis. Proc Natl Acad Sci U S A 1997;94:13198-13202. PMID: 9371823
31 Barton JC, Edwards CQ, Acton RT. HFE gene: structure, function, mutations, and associated iron abnormalities. Gene 2015;574:179-192. PMID: 26456104
32 Bennett MJ, Lebrón JA, Bjorkman PJ. Crystal structure of the hereditary haemochromatosis protein HFE complexed with transferrin receptor. Nature 2000;403:46-53. PMID: 10638746
33 West AP Jr, Giannetti AM, Herr AB, Bennett MJ, Nangiana JS, Pierce JR, et al. Mutational analysis of the transferrin receptor reveals overlapping HFE and transferrin binding sites. J Mol Biol 2001;313:385-397. PMID: 11800564
34 Gao J, Chen J, Kramer M, Tsukamoto H, Zhang AS, Enns CA. Interaction of the hereditary hemochromatosis protein HFE with transferrin receptor 2 is required for transferrin-induced hepcidin expression. Cell Metab 2009;9:217-227. PMID: 19254567
35 Calzolari A, Raggi C, Deaglio S, Sposi NM, Stafsnes M, Fecchi K, et al. TfR2 localizes in lipid raft domains and is released in exosomes to activate signal transduction along the MAPK pathway. J Cell Sci 2006;119:4486-4498. PMID: 17046995
36 Schmidt PJ, Fleming MD. Transgenic HFE-dependent induction of hepcidin in mice does not require transferrin receptor-2. Am J Hematol 2012;87:588-595. PMID: 22460705
37 Rishi G, Crampton EM, Wallace DF, Subramaniam VN. In situ proximity ligation assays indicate that hemochromatosis proteins Hfe and transferrin receptor 2 (Tfr2) do not interact. PLoS One 2013;8:e77267. PMID: 24155934
38 Bolondi G, Garuti C, Corradini E, Zoller H, Vogel W, Finkenstedt A, et al. Altered hepatic BMP signaling pathway in human HFE hemochromatosis. Blood Cells Mol Dis 2010;45:308-312. PMID: 20863724
39 D’Alessio F, Hentze MW, Muckenthaler MU. The hemochromatosis proteins HFE, TfR2, and HJV form a membrane-associated protein complex for hepcidin regulation. J Hepatol 2012;57:1052-1060. PMID: 22728873
40 Wu XG, Wang Y, Wu Q, Cheng WH, Liu W, Zhao Y, et al. HFE interacts with the BMP type I receptor ALK3 to regulate hepcidin expression. Blood 2014;124:1335-1343. PMID: 24904118
41 Harrison SA, Bacon BR. Hereditary hemochromatosis: update for 2003. J Hepatol 2003;38:S14-23. PMID: 12591183
42 European Association For The Study Of The Liver. EASL clinical practice guidelines for HFE hemochromatosis. J Hepatol 2010;53:3-22. PMID: 20471131
43 Adams P, Brissot P, Powell LW. EASL International Consensus Conference on Haemochromatosis. J Hepatol 2000;33:485-504. PMID: 11020008
44 Beutler E, Felitti VJ, Koziol JA, Ho NJ, Gelbart T. Penetrance of 845G--> A (C282Y) HFE hereditary haemochromatosis mutation in the USA. Lancet 2002;359:211-218. PMID: 11812557
45 Olynyk JK, Hagan SE, Cullen DJ, Beilby J, Whittall DE. Evolution of untreated hereditary hemochromatosis in the Busselton population: a 17-year study. Mayo Clin Proc 2004;79:309-313. PMID: 15008603
46 Andersen RV, Tybjaerg-Hansen A, Appleyard M, Birgens H, Nordestgaard BG. Hemochromatosis mutations in the general population: iron overload progression rate. Blood 2004;103:2914-2919. PMID: 15070663
47 Whitlock EP, Garlitz BA, Harris EL, Beil TL, Smith PR. Screening for hereditary hemochromatosis: a systematic review for the U.S. Preventive Services Task Force. Ann Intern Med 2006;145:209-223. PMID: 16880463
48 Allen KJ, Gurrin LC, Constantine CC, Osborne NJ, Delatycki MB, Nicoll AJ, et al. Iron-overload-related disease in HFE hereditary hemochromatosis. N Engl J Med 2008;358:221-230. PMID: 18199861
49 Ekanayake D, Roddick C, Powell LW. Recent advances in hemochromatosis: a 2015 update : a summary of proceedings of the 2014 conference held under the auspices of Hemochromatosis Australia. Hepatol Int 2015;9:174-182. PMID: 25788196
50 McLaren CE, Emond MJ, Subramaniam VN, Phatak PD, Barton JC, Adams PC, et al. Exome sequencing in HFE C282Y homozygous men with extreme phenotypes identifies a GNPAT variant associated with severe iron overload. Hepatology 2015;62:429-439. PMID: 25605615
51 Hanson EH, Imperatore G, Burke W. HFE gene and hereditary hemochromatosis: a HuGE review. Human Genome Epidemiology. Am J Epidemiol 2001;154:193-206. PMID: 11479183
52 Pietrangelo A. Hereditary hemochromatosis--a new look at an old disease. N Engl J Med 2004;350:2383-2397. PMID: 15175440
53 Gochee PA, Powell LW, Cullen DJ, Du Sart D, Rossi E, Olynyk JK. A population-based study of the biochemical and clinical expression of the H63D hemochromatosis mutation. Gastroenterology 2002;122:646-651. PMID: 11874997
54 Fargion S, Valenti L, Dongiovanni P, Scaccabarozzi A, Fracanzani AL, Taioli E, et al. Tumor necrosis factor alpha promoter polymorphisms influence the phenotypic expression of hereditary hemochromatosis. Blood 2001;97:3707-3712. PMID: 11389006
55 Erhardt A, Maschner-Olberg A, Mellenthin C, Kappert G, Adams O, Donner A, et al. HFE mutations and chronic hepatitis C: H63D and C282Y heterozygosity are independent risk factors for liver fibrosis and cirrhosis. J Hepatol 2003;38:335-342. PMID: 12586300
56 Machado MV, Ravasco P, Martins A, Almeida MR, Camilo ME, Cortez-Pinto H. Iron homeostasis and H63D mutations in alcoholics with and without liver disease. World J Gastroenterol 2009;15:106-111. PMID: 19115475
57 Zaloumis SG, Allen KJ, Bertalli NA, Turkovic L, Delatycki MB, Nicoll AJ, et al. Natural history of HFE simple heterozygosity for C282Y and H63D: a prospective 12-year study. J Gastroenterol Hepatol 2015;30:719-725. PMID: 25311314
58 Piperno A, Arosio C, Fossati L, Viganò M, Trombini P, Vergani A, et al. Two novel nonsense mutations of HFE gene in five unrelated italian patients with hemochromatosis. Gastroenterology 2000;119:441-445. PMID: 10930379
59 Donovan A, Brownlie A, Zhou Y, Shepard J, Pratt SJ, Moynihan J, et al. Positional cloning of zebrafish ferroportin1 identifies a conserved vertebrate iron exporter. Nature 2000;403:776-781. PMID: 10693807
60 Njajou OT, Vaessen N, Joosse M, Berghuis B, van Dongen JW, Breuning MH, et al. A mutation in SLC11A3 is associated with autosomal dominant hemochromatosis. Nat Genet 2001;28:213-214. PMID: 11431687
61 Pietrangelo A. The ferroportin disease. Blood Cells Mol Dis 2004;32:131-138. PMID: 14757427
62 Schimanski LM, Drakesmith H, Merryweather-Clarke AT, Viprakasit V, Edwards JP, Sweetland E, et al. In vitro functional analysis of human ferroportin (FPN) and hemochromatosis-associated FPN mutations. Blood 2005;105:4096-4102. PMID: 15692071
63 Létocart E, Le Gac G, Majore S, Ka C, Radio FC, Gourlaouen I, et al. A novel missense mutation in SLC40A1 results in resistance to hepcidin and confirms the existence of two ferroportin-associated iron overload diseases. Br J Haematol 2009;147:379-385. PMID: 19709084
64 Kato J, Fujikawa K, Kanda M, Fukuda N, Sasaki K, Takayama T, et al. A mutation, in the iron-responsive element of H ferritin mRNA, causing autosomal dominant iron overload. Am J Hum Genet 2001;69:191-197. PMID: 11389486
65 Kato J, Niitsu Y. Recent advance in molecular iron metabolism: translational disorders of ferritin. Int J Hematol 2002;76:208-212. PMID: 12416730
66 Phatak PD, Sham RL, Raubertas RF, Dunnigan K, O’Leary MT, Braggins C, et al. Prevalence of hereditary hemochromatosis in 16031 primary care patients. Ann Intern Med 1998;129:954-961. PMID: 9867748
67 Strohmeyer G, Niederau C, Stremmel W. Survival and causes of death in hemochromatosis. Observations in 163 patients. Ann N Y Acad Sci 1988;526:245-257. PMID: 3389643
68 Pietrangelo A. Hereditary hemochromatosis: pathogenesis, diagnosis, and treatment. Gastroenterology 2010;139:393-408. PMID: 20542038
69 Tanno T, Bhanu NV, Oneal PA, Goh SH, Staker P, Lee YT, et al. High levels of GDF15 in thalassemia suppress expression of the iron regulatory protein hepcidin. Nat Med 2007;13:1096-1101. PMID: 17721544
70 Lakhal S, Schödel J, Townsend AR, Pugh CW, Ratcliffe PJ, Mole DR. Regulation of type II transmembrane serine proteinase TMPRSS6 by hypoxia-inducible factors: new link between hypoxia signaling and iron homeostasis. J Biol Chem 2011;286:4090-4097. PMID: 20966077
71 Mastrogiannaki M, Matak P, Mathieu JR, Delga S, Mayeux P, Vaulont S, et al. Hepatic hypoxia-inducible factor-2 down-regulates hepcidin expression in mice through an erythropoietin-mediated increase in erythropoiesis. Haematologica 2012;97:827-834. PMID: 22207682
72 Yoshida K, Furihata K, Takeda S, Nakamura A, Yamamoto K, Morita H, et al. A mutation in the ceruloplasmin gene is associated with systemic hemosiderosis in humans. Nat Genet 1995;9:267-272. PMID: 7539672
73 Beaumont-Epinette MP, Delobel JB, Ropert M, Deugnier Y, Loréal O, Jouanolle AM, et al. Hereditary hypotransferrinemia can lead to elevated transferrin saturation and, when associated to HFE or HAMP mutations, to iron overload. Blood Cells Mol Dis 2015;54:151-154. PMID: 25486930
74 Iolascon A, Camaschella C, Pospisilova D, Piscopo C, Tchernia G, Beaumont C. Natural history of recessive inheritance of DMT1 mutations. J Pediatr 2008;152:136-139. PMID: 18154916
75 Lange CM, Kutalik Z, Morikawa K, Bibert S, Cerny A, Dollenmaier G, et al. Serum ferritin levels are associated with a distinct phenotype of chronic hepatitis C poorly responding to pegylated interferon-alpha and ribavirin therapy. Hepatology 2012;55:1038-1047. PMID: 22095909
76 Dostalikova-Cimburova M, Balusikova K, Kratka K, Chmelikova J, Hejda V, Hnanicek J, et al. Role of duodenal iron transporters and hepcidin in patients with alcoholic liver disease. J Cell Mol Med 2014;18:1840-1850. PMID: 24894955
77 Ioannou GN, Dominitz JA, Weiss NS, Heagerty PJ, Kowdley KV. The effect of alcohol consumption on the prevalence of iron overload, iron deficiency, and iron deficiency anemia. Gastroenterology 2004;126:1293-1301. PMID: 15131790
78 Dongiovanni P, Fracanzani AL, Fargion S, Valenti L. Iron in fatty liver and in the metabolic syndrome: a promising therapeutic target. J Hepatol 2011;55:920-932. PMID: 21718726
79 Singal AK, Kormos-Hallberg C, Lee C, Sadagoparamanujam VM, Grady JJ, Freeman DH Jr, et al. Low-dose hydroxychloroquine is as effective as phlebotomy in treatment of patients with porphyria cutanea tarda. Clin Gastroenterol Hepatol 2012;10:1402-1409. PMID: 22985607
80 Blachier M, Leleu H, Peck-Radosavljevic M, Valla DC, Roudot-Thoraval F. The burden of liver disease in Europe: a review of available epidemiological data. J Hepatol 2013;58:593-608. PMID: 23419824
81 Chapoutot C, Esslimani M, Joomaye Z, Ramos J, Perney P, Laurent C, et al. Liver iron excess in patients with hepatocellular carcinoma developed on viral C cirrhosis. Gut 2000;46:711-714. PMID: 10764717
82 Kowdley KV, Belt P, Wilson LA, Yeh MM, Neuschwander-Tetri BA, Chalasani N, et al. Serum ferritin is an independent predictor of histologic severity and advanced fibrosis in patients with nonalcoholic fatty liver disease. Hepatology 2012;55:77-85. PMID: 21953442
83 Sikorska K, Stalke P, Izycka-Swieszewska E, Romanowski T, Bielawski KP. The role of iron overload and HFE gene mutations in the era of pegylated interferon and ribavirin treatment of chronic hepatitis C. Med Sci Monit 2010;16:CR137-143. PMID: 20190684
84 Valenti L, Pulixi EA, Arosio P, Cremonesi L, Biasiotto G, Dongiovanni P, et al. Relative contribution of iron genes, dysmetabolism and hepatitis C virus (HCV) in the pathogenesis of altered iron regulation in HCV chronic hepatitis. Haematologica 2007;92:1037-1042. PMID: 17640859
85 Valenti L, Fracanzani AL, Bugianesi E, Dongiovanni P, Galmozzi E, Vanni E, et al. HFE genotype, parenchymal iron accumulation, and liver fibrosis in patients with nonalcoholic fatty liver disease. Gastroenterology 2010;138:905-912. PMID: 19931264
86 Sikorska K, Romanowski T, Stalke P, I?ycka-?wieszewska E, Bielawski KP. Iron overload and HFE gene mutations in Polish patients with liver cirrhosis. Hepatobiliary Pancreat Dis Int 2011;10:270-275. PMID: 21669570
87 Adams LA, Crawford DH, Stuart K, House MJ, St Pierre TG, Webb M, et al. The impact of phlebotomy in nonalcoholic fatty liver disease: A prospective, randomized, controlled trial. Hepatology 2015;61:1555-1564. PMID: 25524401
88 Assi TB, Baz E. Current applications of therapeutic phlebotomy. Blood Transfus 2014;12:s75-83. PMID: 24120605
89 Takaki A, Kawai D, Yamamoto K. Multiple hits, including oxidative stress, as pathogenesis and treatment target in non-alcoholic steatohepatitis (NASH). Int J Mol Sci 2013;14:20704- 20728. PMID: 24132155
90 Datz C, Felder TK, Niederseer D, Aigner E. Iron homeostasis in the metabolic syndrome. Eur J Clin Invest 2013;43:215-224. PMID: 23289518
91 Bugianesi E, Manzini P, D’Antico S, Vanni E, Longo F, Leone N, et al. Relative contribution of iron burden, HFE mutations, and insulin resistance to fibrosis in nonalcoholic fatty liver. Hepatology 2004;39:179-187. PMID: 14752836
92 Valenti L, Fracanzani AL, Dongiovanni P, Rovida S, Rametta R, Fatta E, et al. A randomized trial of iron depletion in patients with nonalcoholic fatty liver disease and hyperferritinemia. World J Gastroenterol 2014;20:3002-3010. PMID: 24659891
93 Bu W, Liu R, Cheung-Lau JC, Dmochowski IJ, Loll PJ, Eckenhoff RG. Ferritin couples iron and fatty acid metabolism. FASEB J 2012;26:2394-2400. PMID: 22362897
94 Dongiovanni P, Lanti C, Gatti S, Rametta R, Recalcati S, Maggioni M, et al. High fat diet subverts hepatocellular iron uptake determining dysmetabolic iron overload. PLoS One 2015;10:e0116855. PMID: 25647178
95 Tsuchiya H, Ebata Y, Sakabe T, Hama S, Kogure K, Shiota G. High-fat, high-fructose diet induces hepatic iron overload via a hepcidin-independent mechanism prior to the onset of liver steatosis and insulin resistance in mice. Metabolism 2013;62:62-69. PMID: 22854109
96 Meli R, Mattace Raso G, Irace C, Simeoli R, Di Pascale A, Paciello O, et al. High fat diet induces liver steatosis and early dysregulation of iron metabolism in rats. PLoS One 2013;8:e66570. PMID: 23805238
97 Bekri S, Gual P, Anty R, Luciani N, Dahman M, Ramesh B, et al. Increased adipose tissue expression of hepcidin in severe obesity is independent from diabetes and NASH. Gastroenterology 2006;131:788-796. PMID: 16952548
98 Aigner E, Felder TK, Oberkofler H, Hahne P, Auer S, Soyal S, et al. Glucose acts as a regulator of serum iron by increasing serum hepcidin concentrations. J Nutr Biochem 2013;24:112-117. PMID: 22819549
99 Musci G, Polticelli F, Bonaccorsi di Patti MC. Ceruloplasmin-ferroportin system of iron traffic in vertebrates. World J Biol Chem 2014;5:204-215. PMID: 24921009
100 Aigner E, Weiss G, Datz C. Dysregulation of iron and copper homeostasis in nonalcoholic fatty liver. World J Hepatol 2015;7:177-188. PMID: 25729473
101 Di Bisceglie AM, Axiotis CA, Hoofnagle JH, Bacon BR. Measurements of iron status in patients with chronic hepatitis. Gastroenterology 1992;102:2108-2113. PMID: 1587431
102 Fillebeen C, Muckenthaler M, Andriopoulos B, Bisaillon M, Mounir Z, Hentze MW, et al. Expression of the subgenomic hepatitis C virus replicon alters iron homeostasis in Huh7 cells. J Hepatol 2007;47:12-22. PMID: 17399844
103 Takeo M, Kobayashi Y, Fujita N, Urawa N, Iwasa M, Horiike S, et al. Upregulation of transferrin receptor 2 and ferroportin 1 mRNA in the liver of patients with chronic hepatitis C. J Gastroenterol Hepatol 2005;20:562-569. PMID: 15836704
104 Ma L, Zou T, Yuan Y, Lv J, Dong X, Yang G, et al. Duodenal ferroportin is up-regulated in patients with chronic hepatitis C. PLoS One 2014;9:e110658. PMID: 25330009
105 Kohjima M, Yoshimoto T, Enjoji M, Fukushima N, Fukuizumi K, Nakamura T, et al. Hepcidin/ferroportin expression levels involve efficacy of pegylated-interferon plus ribavirin in hepatitis C virus-infected liver. World J Gastroenterol 2015;21:3291-3299. PMID: 25805936
106 Mifuji R, Kobayashi Y, Ma N, Qiang QL, Urawa N, Horiike S, et al. Role of transferrin receptor 2 in hepatic accumulation of iron in patients with chronic hepatitis C. J Gastroenterol Hepatol 2006;21:144-151. PMID: 16706826
107 Nanba S, Ikeda F, Baba N, Takaguchi K, Senoh T, Nagano T, et al. Association of hepatic oxidative stress and iron dysregulation with HCC development after interferon therapy in chronic hepatitis C. J Clin Pathol 2016;69:226-233. PMID: 26290259
108 Choi J, Corder NL, Koduru B, Wang Y. Oxidative stress and hepatic Nox proteins in chronic hepatitis C and hepatocellular carcinoma. Free Radic Biol Med 2014;72:267-284. PMID: 24816297
109 Andriopoulos B, Hegedüsch S, Mangin J, Riedel HD, Hebling U, Wang J, et al. Sustained hydrogen peroxide induces iron uptake by transferrin receptor-1 independent of the iron regulatory protein/iron-responsive element network. J Biol Chem 2007;282:20301-20308. PMID: 17517884
110 Mueller S, Pantopoulos K, Hübner CA, Stremmel W, Hentze MW. IRP1 activation by extracellular oxidative stress in the perfused rat liver. J Biol Chem 2001;276:23192-23196. PMID: 11297549
111 Miura K, Taura K, Kodama Y, Schnabl B, Brenner DA. Hepatitis C virus-induced oxidative stress suppresses hepcidin expression through increased histone deacetylase activity. Hepatology 2008;48:1420-1429. PMID: 18671304
112 Kotta-Loizou I, Vassilaki N, Pissas G, Kakkanas A, Bakiri L, Bartenschlager R, et al. Hepatitis C virus core+1/ARF protein decreases hepcidin transcription through an AP1 binding site. J Gen Virol 2013;94:1528-1534. PMID: 23580428
113 Fujita N, Sugimoto R, Motonishi S, Tomosugi N, Tanaka H, Takeo M, et al. Patients with chronic hepatitis C achieving a sustained virological response to peginterferon and ribavirin therapy recover from impaired hepcidin secretion. J Hepatol 2008;49:702-710. PMID: 18620776
114 Girelli D, Pasino M, Goodnough JB, Nemeth E, Guido M, Castagna A, et al. Reduced serum hepcidin levels in patients with chronic hepatitis C. J Hepatol 2009;51:845-852. PMID: 19729219
115 Nishina S, Hino K, Korenaga M, Vecchi C, Pietrangelo A, Mizukami Y, et al. Hepatitis C virus-induced reactive oxygen species raise hepatic iron level in mice by reducing hepcidin transcription. Gastroenterology 2008;134:226-238. PMID: 18166355
116 Sikorska K, Romanowski T, Stalke P, Izycka Swieszewska E, Bielawski KP. Association of hepcidin mRNA expression with hepatocyte iron accumulation and effects of antiviral therapy in chronic hepatitis C infection. Hepat Mon 2014;14:e21184. PMID: 25598789
117 Strnad P, Nuraldeen R, Fischer J, Berg T, Trautwein C. Hepcidin as a predictor of treatment response in chronic hepatitis C infection. Hepatology 2014;59:1648. PMID: 23907834
118 Fillebeen C, Pantopoulos K. Iron inhibits replication of infectious hepatitis C virus in permissive Huh7.5.1 cells. J Hepatol 2010;53:995-999. PMID: 20813419
119 Fillebeen C, Rivas-Estilla AM, Bisaillon M, Ponka P, Muckenthaler M, Hentze MW, et al. Iron inactivates the RNA polymerase NS5B and suppresses subgenomic replication of hepatitis C virus. J Biol Chem 2005;280:9049-9057. PMID: 15637067
120 Bartolomei G, Cevik RE, Marcello A. Modulation of hepatitis C virus replication by iron and hepcidin in Huh7 hepatocytes. J Gen Virol 2011;92:2072-2081. PMID: 21593278
121 Tai AW, Benita Y, Peng LF, Kim SS, Sakamoto N, Xavier RJ, et al. A functional genomic screen identifies cellular cofactors of hepatitis C virus replication. Cell Host Microbe 2009;5:298-307. PMID: 19286138
122 Sikorska K, Stalke P, Romanowski T, Rzepko R, Bielawski KP. Liver steatosis correlates with iron overload but not with HFE gene mutations in chronic hepatitis C. Hepatobiliary Pancreat Dis Int 2013;12:377-384. PMID: 23924495
123 Fujita N, Sugimoto R, Urawa N, Araki J, Mifuji R, Yamamoto M, et al. Hepatic iron accumulation is associated with disease progression and resistance to interferon/ribavirin combination therapy in chronic hepatitis C. J Gastroenterol Hepatol 2007;22:1886-1893. PMID: 17914965
124 Sikorska K, Stalke P, Bielawski KP. Is there any association between HCV multiplication and iron induced liver injury in chronic hepatitis C? J Hepatol 2011;55:235-237. PMID: 21236307
125 Price L, Kowdley KV. The role of iron in the pathophysiology and treatment of chronic hepatitis C. Can J Gastroenterol 2009;23:822-828. PMID: 20011735
126 Ryan JD, Crowe J. Hepatocyte iron accumulation: a new string to ribavirin’s antiviral bow? J Hepatol 2011;55:237-238. PMID: 21349299
127 Novo E, Cannito S, Paternostro C, Bocca C, Miglietta A, Parola M. Cellular and molecular mechanisms in liver fibrogenesis. Arch Biochem Biophys 2014;548:20-37. PMID: 24631571
128 Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012;380:2095-2128. PMID: 23245604
129 Cabantchik ZI. Labile iron in cells and body fluids: physiology, pathology, and pharmacology. Front Pharmacol 2014;5:45. PMID: 24659969
130 Bridle KR, Crawford DH, Fletcher LM, Smith JL, Powell LW, Ramm GA. Evidence for a sub-morphological inflammatory process in the liver in haemochromatosis. J Hepatol 2003;38:426-433. PMID: 12663233
131 Fargion S, Mattioli M, Fracanzani AL, Sampietro M, Tavazzi D, Fociani P, et al. Hyperferritinemia, iron overload, and multiple metabolic alterations identify patients at risk for nonalcoholic steatohepatitis. Am J Gastroenterol 2001;96:2448-2455. PMID: 11513189
132 Deugnier YM, Loréal O, Turlin B, Guyader D, Jouanolle H, Moirand R, et al. Liver pathology in genetic hemochromatosis: a review of 135 homozygous cases and their bioclinical correlations. Gastroenterology 1992;102:2050-2059. PMID: 1587423
133 Heymann F, Trautwein C, Tacke F. Monocytes and macrophages as cellular targets in liver fibrosis. Inflamm Allergy Drug Targets 2009;8:307-318. PMID: 19534673
134 Friedman SL. Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol Rev 2008;88:125-172. PMID: 18195085
135 Duval F, Moreno-Cuevas JE, González-Garza MT, Rodríguez-Montalvo C, Cruz-Vega DE. Liver fibrosis and protection mechanisms action of medicinal plants targeting apoptosis of hepatocytes and hepatic stellate cells. Adv Pharmacol Sci 2014;2014:373295. PMID: 25505905
136 Zhang M, Wang FL, Zhu JY, Zheng YB, Zhao QY, Gu YR, et al. Liver myofibroblasts regulate the phenotype and function of monocytes through soluble factors in cirrhosis. Exp Ther Med 2013;5:143-149. PMID: 23251256
137 Ramm GA, Ruddell RG, Subramaniam VN. Identification of ferritin receptors: their role in iron homeostasis, hepatic injury, and inflammation. Gastroenterology 2009;137:1849-1851. PMID: 19789086
138 Bridle KR, Crawford DH, Ramm GA. Identification and characterization of the hepatic stellate cell transferrin receptor. Am J Pathol 2003;162:1661-1667. PMID: 12707050
139 Ramm GA, Li SC, Li L, Britton RS, O’Neill R, Kobayashi Y, et al. Chronic iron overload causes activation of rat lipocytes in vivo. Am J Physiol 1995;268:G451-458. PMID: 7900806
140 Ramm GA, Crawford DH, Powell LW, Walker NI, Fletcher LM, Halliday JW. Hepatic stellate cell activation in genetic haemochromatosis. Lobular distribution, effect of increasing hepatic iron and response to phlebotomy. J Hepatol 1997;26:584-592. PMID: 9075666
141 Falize L, Guillygomarc’h A, Perrin M, Lainé F, Guyader D, Brissot P, et al. Reversibility of hepatic fibrosis in treated genetic hemochromatosis: a study of 36 cases. Hepatology 2006;44:472-477. PMID: 16871557
142 Pietrangelo A. Iron-induced oxidant stress in alcoholic liver fibrogenesis. Alcohol 2003;30:121-129. PMID: 12957296
143 Kruszewski M, Iwane��ko T, Bart?omiejczyk T, Woli��ski J, Starzy��ski RR, Gralak MA, et al. Hepatic iron content corresponds with the susceptibility of lymphocytes to oxidative stress in neonatal pigs. Mutat Res 2008;657:146-149. PMID: 18824129
144 Wang L, Johnson EE, Shi HN, Walker WA, Wessling-Resnick M, Cherayil BJ. Attenuated inflammatory responses in hemochromatosis reveal a role for iron in the regulation of macrophage cytokine translation. J Immunol 2008;181:2723-2731. PMID: 18684963
145 Porto G, De Sousa M. Iron overload and immunity. World J Gastroenterol 2007;13:4707-4715. PMID: 17729392
146 Wang L, Harrington L, Trebicka E, Shi HN, Kagan JC, Hong CC, et al. Selective modulation of TLR4-activated inflammatory responses by altered iron homeostasis in mice. J Clin Invest 2009;119:3322-3328. PMID: 19809161
147 Barton JC, Wiener HW, Acton RT, Go RC. Total blood lymphocyte counts in hemochromatosis probands with HFE C282Y homozygosity: relationship to severity of iron overload and HLA-A and -B alleles and haplotypes. BMC Blood Disord 2005;5:5. PMID: 16042809
148 Macedo MF, Cruz E, Lacerda R, Porto G, de Sousa M. Low serum transferrin levels in HFE C282Y homozygous subjects are associated with low CD8(+) T lymphocyte numbers. Blood Cells Mol Dis 2005;35:319-325. PMID: 16140024
149 Costa M, Cruz E, Oliveira S, Benes V, Ivacevic T, Silva MJ, et al. Lymphocyte gene expression signatures from patients and mouse models of hereditary hemochromatosis reveal a function of HFE as a negative regulator of CD8+ T-lymphocyte activation and differentiation in vivo. PLoS One 2015;10:e0124246. PMID: 25880808
150 Mencacci A, Cenci E, Boelaert JR, Bucci P, Mosci P, Fè d’Ostiani C, et al. Iron overload alters innate and T helper cell responses to Candida albicans in mice. J Infect Dis 1997;175:1467-1476. PMID: 9180188
151 Melo RA, Garcia AB, Viana SR, Falcão RP. Lymphocyte subsets in experimental hemochromatosis. Acta Haematol 1997;98:72-75. PMID: 9286302
152 Cardoso EM, Hagen K, de Sousa M, Hultcrantz R. Hepatic damage in C282Y homozygotes relates to low numbers of CD8+ cells in the liver lobuli. Eur J Clin Invest 2001;31:45-53. PMID: 11168438
153 Porto G, Vicente C, Teixeira MA, Martins O, Cabeda JM, Lacerda R, et al. Relative impact of HLA phenotype and CD4-CD8 ratios on the clinical expression of hemochromatosis. Hepatology 1997;25:397-402. PMID: 9021953
154 Cherayil BJ. Iron and immunity: immunological consequences of iron deficiency and overload. Arch Immunol Ther Exp (Warsz) 2010;58:407-415. PMID: 20878249
155 Ward RJ, Crichton RR, Taylor DL, Della Corte L, Srai SK, Dexter DT. Iron and the immune system. J Neural Transm (Vienna) 2011;118:315-328. PMID: 20878427
156 Ripoll C, Keitel F, Hollenbach M, Greinert R, Zipprich A. Serum ferritin in patients with cirrhosis is associated with markers of liver insufficiency and circulatory dysfunction, but not of portal hypertension. J Clin Gastroenterol 2015;49:784-789. PMID: 25599219
157 Walker NM, Stuart KA, Ryan RJ, Desai S, Saab S, Nicol JA, et al. Serum ferritin concentration predicts mortality in patients awaiting liver transplantation. Hepatology 2010;51:1683-1691. PMID: 20225256
158 Maiwall R, Kumar S, Chaudhary AK, Maras J, Wani Z, Kumar C, et al. Serum ferritin predicts early mortality in patients with decompensated cirrhosis. J Hepatol 2014;61:43-50. PMID: 24681346
159 Abu Rajab M, Guerin L, Lee P, Brown KE. Iron overload secondary to cirrhosis: a mimic of hereditary haemochromatosis? Histopathology 2014;65:561-569. PMID: 24635122
160 Maras JS, Maiwall R, Harsha HC, Das S, Hussain MS, Kumar C, et al. Dysregulated iron homeostasis is strongly associated with multiorgan failure and early mortality in acute-on-chronic liver failure. Hepatology 2015;61:1306-1320. PMID: 25475192
161 Kowdley KV, Brandhagen DJ, Gish RG, Bass NM, Weinstein J, Schilsky ML, et al. Survival after liver transplantation in patients with hepatic iron overload: the national hemochromatosis transplant registry. Gastroenterology 2005;129:494-503. PMID: 16083706
162 Brandhagen DJ, Alvarez W, Therneau TM, Kruckeberg KE, Thibodeau SN, Ludwig J, et al. Iron overload in cirrhosis-HFE genotypes and outcome after liver transplantation. Hepatology 2000;31:456-460. PMID: 10655270
163 Fenton H, Torbenson M, Vivekanandan P, Yeh MM, Hart J, Ferrell L. Marked iron in liver explants in the absence of major hereditary hemochromatosis gene defects: a risk factor for cardiac failure. Transplantation 2009;87:1256-1260. PMID: 19384175
164 Dar FS, Faraj W, Zaman MB, Bartlett A, Bomford A, O’Sullivan A, et al. Outcome of liver transplantation in hereditary hemochromatosis. Transpl Int 2009;22:717-724. PMID: 19490544
165 Venook AP, Papandreou C, Furuse J, de Guevara LL. The incidence and epidemiology of hepatocellular carcinoma: a global and regional perspective. Oncologist 2010;15:5-13. PMID: 21115576
166 Sanyal AJ, Yoon SK, Lencioni R. The etiology of hepatocellular carcinoma and consequences for treatment. Oncologist 2010;15:14-22. PMID: 21115577
167 van Gijssel HE, Maassen CB, Mulder GJ, Meerman JH. p53 protein expression by hepatocarcinogens in the rat liver and its potential role in mitoinhibition of normal hepatocytes as a mechanism of hepatic tumour promotion. Carcinogenesis 1997;18:1027-1033. PMID: 9163691
168 Alexander J, Kowdley KV. HFE-associated hereditary hemochromatosis. Genet Med 2009;11:307-313. PMID: 19444013
169 Beaton MD, Adams PC. Prognostic factors and survival in patients with hereditary hemochromatosis and cirrhosis. Can J Gastroenterol 2006;20:257-260. PMID: 16609753
170 Ito K, Mitchell DG, Gabata T, Hann HW, Kim PN, Fujita T, et al. Hepatocellular carcinoma: association with increased iron deposition in the cirrhotic liver at MR imaging. Radiology 1999;212:235-240. PMID: 10405747
171 Ioannou GN, Weiss NS, Kowdley KV. Relationship between transferrin-iron saturation, alcohol consumption, and the incidence of cirrhosis and liver cancer. Clin Gastroenterol Hepatol 2007;5:624-629. PMID: 17478349
172 Kato J, Kobune M, Nakamura T, Kuroiwa G, Takada K, Takimoto R, et al. Normalization of elevated hepatic 8-hydroxy-2’-deoxyguanosine levels in chronic hepatitis C patients by phlebotomy and low iron diet. Cancer Res 2001;61:8697-8702. PMID: 11751387
173 Kato J, Miyanishi K, Kobune M, Nakamura T, Takada K, Takimoto R, et al. Long-term phlebotomy with low-iron diet therapy lowers risk of development of hepatocellular carcinoma from chronic hepatitis C. J Gastroenterol 2007;42:830-836. PMID: 17940836
174 Hann HW, Stahlhut MW, Hann CL. Effect of iron and desferoxamine on cell growth and in vitro ferritin synthesis in human hepatoma cell lines. Hepatology 1990;11:566-569. PMID: 2158479
175 Marrogi AJ, Khan MA, van Gijssel HE, Welsh JA, Rahim H, Demetris AJ, et al. Oxidative stress and p53 mutations in the carcinogenesis of iron overload-associated hepatocellular carcinoma. J Natl Cancer Inst 2001;93:1652-1655. PMID: 11698570
176 Asare GA, Paterson AC, Kew MC, Khan S, Mossanda KS. Iron-free neoplastic nodules and hepatocellular carcinoma without cirrhosis in Wistar rats fed a diet high in iron. J Pathol 2006;208:82-90. PMID: 16278820
177 Bralet MP, Régimbeau JM, Pineau P, Dubois S, Loas G, Degos F, et al. Hepatocellular carcinoma occurring in nonfibrotic liver: epidemiologic and histopathologic analysis of 80 French cases. Hepatology 2000;32:200-204. PMID: 10915724
178 Cardin R, Piciocchi M, Sinigaglia A, Lavezzo E, Bortolami M, Kotsafti A, et al. Oxidative DNA damage correlates with cell immortalization and mir-92 expression in hepatocellular carcinoma. BMC Cancer 2012;12:177. PMID: 22587342
179 Farazi PA, Glickman J, Horner J, Depinho RA. Cooperative interactions of p53 mutation, telomere dysfunction, and chronic liver damage in hepatocellular carcinoma progression. Cancer Res 2006;66:4766-4773. PMID: 16651430
180 Asare GA, Bronz M, Naidoo V, Kew MC. Interactions between aflatoxin B1 and dietary iron overload in hepatic mutagenesis. Toxicology. 2007;234:157-166. PMID: 17399882
181 Asare GA, Bronz M, Naidoo V, Kew MC. Synergistic interaction between excess hepatic iron and alcohol ingestion in hepatic mutagenesis. Toxicology 2008;254:11-18. PMID: 18852013
182 Lehmann U, Wingen LU, Brakensiek K, Wedemeyer H, Becker T, Heim A, et al. Epigenetic defects of hepatocellular carcinoma are already found in non-neoplastic liver cells from patients with hereditary haemochromatosis. Hum Mol Genet 2007;16:1335-1342. PMID: 17412760
183 Bloomer SA, Brown KE. Tumour promotion versus tumour suppression in chronic hepatic iron overload. Cell Biochem Funct 2015;33:241-248. PMID: 26059599
184 Kim S, Ponka P. Role of nitric oxide in cellular iron metabolism. Biometals 2003;16:125-135. PMID: 12572672
185 Green R, Esparza I, Schreiber R. Iron inhibits the nonspecific tumoricidal activity of macrophages. A possible contributory mechanism for neoplasia in hemochromatosis. Ann N Y Acad Sci 1988;526:301-309. PMID: 2455463
186 Matsui K, Ezoe S, Yokota T, Ishibashi T, Oritani K, Kanakura Y. Iron overload effects on immune system through the cytokine secretion by macrophage. Blood 2013;122:1047.
187 Tseng HH, Chang JG, Hwang YH, Yeh KT, Chen YL, Yu HS. Expression of hepcidin and other iron-regulatory genes in human hepatocellular carcinoma and its clinical implications. J Cancer Res Clin Oncol 2009;135:1413-1420. PMID: 19387685
188 Kijima H, Sawada T, Tomosugi N, Kubota K. Expression of hepcidin mRNA is uniformly suppressed in hepatocellular carcinoma. BMC Cancer 2008;8:167. PMID: 18541040
189 Boyault S, Rickman DS, de Reyniès A, Balabaud C, Rebouissou S, Jeannot E, et al. Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology 2007;45:42-52. PMID: 17187432
190 Weizer-Stern O, Adamsky K, Margalit O, Ashur-Fabian O, Givol D, Amariglio N, et al. Hepcidin, a key regulator of iron metabolism, is transcriptionally activated by p53. Br J Haematol 2007;138:253-262. PMID: 17593032
191 Bacon BR, Adams PC, Kowdley KV, Powell LW, Tavill AS; American Association for the Study of Liver Diseases. Diagnosis and management of hemochromatosis: 2011 practice guideline by the American Association for the Study of Liver Diseases. Hepatology 2011;54:328-343. PMID: 21452290
192 McDonnell SM, Preston BL, Jewell SA, Barton JC, Edwards CQ, Adams PC, et al. A survey of 2,851 patients with hemochromatosis: symptoms and response to treatment. Am J Med 1999;106:619-624. PMID: 10378618
193 Niederau C, Fischer R, Pürschel A, Stremmel W, Häussinger D, Strohmeyer G. Long-term survival in patients with hereditary hemochromatosis. Gastroenterology 1996;110:1107-1119. PMID: 8613000
194 Alexander J, Tung BY, Croghan A, Kowdley KV. Effect of iron depletion on serum markers of fibrogenesis, oxidative stress and serum liver enzymes in chronic hepatitis C: results of a pilot study. Liver Int 2007;27:268-273. PMID: 17311623
195 Sartori M, Andorno S, Rossini A, Boldorini R, Bozzola C, Carmagnola S, et al. A case-control histological study on the effects of phlebotomy in patients with chronic hepatitis C. Eur J Gastroenterol Hepatol 2011;23:1178-1184. PMID: 22002003
196 Yano M, Hayashi H, Wakusawa S, Sanae F, Takikawa T, Shiono Y, et al. Long term effects of phlebotomy on biochemical and histological parameters of chronic hepatitis C. Am J Gastroenterol 2002;97:133-137. PMID: 11808937
197 Beaton MD, Chakrabarti S, Levstik M, Speechley M, Marotta P, Adams P. Phase II clinical trial of phlebotomy for non-alcoholic fatty liver disease. Aliment Pharmacol Ther 2013;37:720-729. PMID: 23441892
198 Knauer CM, Gamble CN, Monroe LS. The reversal of hemochromatotic cirrhosis by multiple phlebotomies. Report of a case. Gastroenterology 1965;49:667-671. PMID: 5846420
199 Hasan MK, Siddiqui AA, Mesiya S, Fazili J, Pitha J, Talbert M. Reversal of advance liver fibrosis with phlebotomy in a patient with porphyria cutanea tarda and hepatitis C. The Am J Gastroenterol 2003;98:S201.
200 Franchini M. Hereditary iron overload: update on pathophysiology, diagnosis, and treatment. Am J Hematol 2006;81:202-209. PMID: 16493621
201 Szurowska E, Sikorska K, Izycka-Swieszewska E, Nowicki T, Romanowski T, Bielawski KP, et al. The role of MR imaging in detection of hepatic iron overload in patients with cirrhosis of different origins. BMC Gastroenterol 2010;10:13. PMID: 20105330
202 Adams PC, Barton JC. How I treat hemochromatosis. Blood 2010;116:317-325. PMID: 20308595
203 Poggiali E, Cassinerio E, Zanaboni L, Cappellini MD. An update on iron chelation therapy. Blood Transfus 2012;10:411-422. PMID: 22790257
204 Maggio A, D’Amico G, Morabito A, Capra M, Ciaccio C, Cianciulli P, et al. Deferiprone versus deferoxamine in patients with thalassemia major: a randomized clinical trial. Blood Cells Mol Dis 2002;28:196-208. PMID: 12064916
205 Cohen AR, Galanello R, Piga A, De Sanctis V, Tricta F. Safety and effectiveness of long-term therapy with the oral iron chelator deferiprone. Blood 2003;102:1583-1587. PMID: 12763939
206 Barry M, Flynn DM, Letsky EA, Risdon RA. Long-term chelation therapy in thalassaemia major: effect on liver iron concentration, liver histology, and clinical progress. Br Med J 1974;2:16-20. PMID: 4821036
207 Wu SF, Peng CT, Wu KH, Tsai CH. Liver fibrosis and iron levels during long-term deferiprone treatment of thalassemia major patients. Hemoglobin 2006;30:215-218. PMID: 16798646
208 Deugnier Y, Turlin B, Ropert M, Cappellini MD, Porter JB, Giannone V, et al. Improvement in liver pathology of patients with β-thalassemia treated with deferasirox for at least 3 years. Gastroenterology 2011;141:1202-1211. PMID: 21741344
209 Kaji K, Yoshiji H, Kitade M, Ikenaka Y, Noguchi R, Shirai Y, 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:G1094-1104. PMID: 21372165
210 Sobbe A, Bridle KR, Jaskowski L, de Guzman CE, Santrampurwala N, Clouston AD, et al. Inconsistent hepatic antifibrotic effects with the iron chelator deferasirox. J Gastroenterol Hepatol 2015;30:638-645. PMID: 25168203
211 Symeonidis AS. The role of iron and iron chelators in zygomycosis. Clin Microbiol Infect 2009;15:26-32. PMID: 19754753
212 Li S, Tan HY, Wang N, Zhang ZJ, Lao L, Wong CW, et al. The role of oxidative stress and antioxidants in liver diseases. Int J Mol Sci 2015;16:26087-26124. PMID: 26540040
213 Weeraphan C, Srisomsap C, Chokchaichamnankit D, Subhasitanont P, Hatairaktham S, Charoensakdi R, et al. Role of curcuminoids in ameliorating oxidative modification in β-thalassemia/Hb E plasma proteome. J Nutr Biochem 2013;24:578-585. PMID: 22818714
214 Srichairatanakool S, Kulprachakarn K, Pangjit K, Pattanapanyasat K, Fuchaeron S. Green tea extract and epigallocatechin 3-gallate reduced labile iron pool and protected oxidative stress in iron-loaded cultured hepatocytes. Adv Biosci Biotechnol 2012;3:1140-1150. PMID: 38140
215 Ounjaijean S, Thephinlap C, Khansuwan U, Phisalapong C, Fucharoen S, Porter JB, et al. Effect of green tea on iron status and oxidative stress in iron-loaded rats. Med Chem 2008;4:365-370. PMID: 18673149
216 Basu A, Betts NM, Mulugeta A, Tong C, Newman E, Lyons TJ. Green tea supplementation increases glutathione and plasma antioxidant capacity in adults with the metabolic syndrome. Nutr Res 2013;33:180-187. PMID: 23507223
217 Kaltwasser JP, Werner E, Schalk K, Hansen C, Gottschalk R, Seidl C. Clinical trial on the effect of regular tea drinking on iron accumulation in genetic haemochromatosis. Gut 1998;43:699-704. PMID: 9824354
218 Saewong T, Ounjaijean S, Mundee Y, Pattanapanyasat K, Fucharoen S, Porter JB, et al. Effects of green tea on iron accumulation and oxidative stress in livers of iron-challenged thalassemic mice. Med Chem 2010;6:57-64. PMID: 20218965
219 Das SK, DesAulniers J, Dyck JR, Kassiri Z, Oudit GY. Resveratrol mediates therapeutic hepatic effects in acquired and genetic murine models of iron-overload. Liver Int 2016;36:246-257. PMID: 26077449
220 Bares JM, Berger J, Nelson JE, Messner DJ, Schildt S, Standish LJ, et al. Silybin treatment is associated with reduction in serum ferritin in patients with chronic hepatitis C. J Clin Gastroenterol 2008;42:937-944. PMID: 18458640
221 Darvishi Khezri H, Salehifar E, Kosaryan M, Aliasgharian A, Jalali H, Hadian Amree A. Potential effects of silymarin and its flavonolignan components in patients with β-Thalassemia major: a comprehensive review in 2015. Adv Pharmacol Sci 2016;2016:3046373. PMID: 26997953
222 Hutchinson C, Bomford A, Geissler CA. The iron-chelating potential of silybin in patients with hereditary haemochromatosis. Eur J Clin Nutr 2010;64:1239-1241. PMID: 20628405
223 Hellerbrand C, Schattenberg JM, Peterburs P, Lechner A, Brignoli R. The potential of silymarin for the treatment of hepatic disorders Clin Phytosci 2016;2:7.
224 Rambaldi A, Jacobs BP, Gluud C. Milk thistle for alcoholic and/or hepatitis B or C virus liver diseases. Cochrane Database Syst Rev 2007;(4):CD003620. PMID: 17943794
225 Canini L, DebRoy S, Mariño Z, Conway JM, Crespo G, Navasa M, et al. Severity of liver disease affects HCV kinetics in patients treated with intravenous silibinin monotherapy. Antivir Ther 2015;20:149-155. PMID: 24912382
226 Bjelakovic G, Gluud LL, Nikolova D, Bjelakovic M, Nagorni A, Gluud C. Antioxidant supplements for liver diseases. Cochrane Database Syst Rev 2011;(3):CD007749. PMID: 21412909
 
Received March 10, 2016
Accepted after revision July 22, 2016