Next Article in Journal
Deficiency of NEIL3 Enhances the Chemotherapy Resistance of Prostate Cancer
Previous Article in Journal
Adiponectin Deregulation in Systemic Autoimmune Rheumatic Diseases
Previous Article in Special Issue
Possible Roles of Interleukin-4 and -13 and Their Receptors in Gastric and Colon Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Iron at the Interface of Hepatocellular Carcinoma

1
Institute of Comparative Molecular Endocrinology, Ulm University, 89081 Ulm, Germany
2
Department of Internal Medicine I, University Hospital Ulm, 89081 Ulm, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(8), 4097; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22084097
Submission received: 19 February 2021 / Revised: 6 April 2021 / Accepted: 13 April 2021 / Published: 15 April 2021
(This article belongs to the Special Issue Biological Interfaces in Gastrointestinal Cancer)

Abstract

:
Cancer incidence and mortality are rapidly growing, with liver cancer being the sixth most diagnosed cancer worldwide and the third leading cause of cancer death in 2020. A number of risk factors have been identified that trigger the progression to hepatocellular carcinoma. In this review, we focus on iron as a potential risk factor for liver carcinogenesis. Molecules involved in the regulation of iron metabolism are often upregulated in cancer cells, in order to provide a supply of this essential trace element for all stages of tumor development, survival, proliferation, and metastasis. Thus, cellular and systemic iron levels must be tightly regulated to prevent or delay liver cancer progression. Disorders associated with dysregulated iron metabolism are characterized with increased susceptibility to hepatocellular carcinoma. This review discusses the association of iron with metabolic disorders such as hereditary hemochromatosis, non-alcoholic fatty liver disease, obesity, and type 2 diabetes, in the background of hepatocellular carcinoma.

1. Global Cancer Incidence

Cancer incidence rates and cancer mortality are rapidly growing worldwide. The number of new cases in the last 20 years has almost doubled, from 10.9 million cases in 2002 to 19.3 million in 2020, and cases are expected to rise in the future [1,2]. This trend is also observed within cancer mortality, with the death toll rising from 6.7 million in 2002, to 10 million in 2020 [1,2]. Global analysis of the incidence and mortality of various cancer types in both genders placed lung cancer as one of the most lethal, followed by female breast cancer, colorectal cancer, prostate cancer, stomach cancer, and liver cancer [1]. The prevalence and distribution of the main risk factors for cancer, several of which are associated with ageing, socioeconomic development and lifestyle, reproductive system, dietary choices, and metabolic and hormonal status, contribute to differing cancer profiles between world regions [3]. For instance, the incidence rate of lung cancers in men living in western countries is 39% in comparison to 10.3% in developing countries [1]. On the other hand, infection-associated cancers, such as cervical cancer, show the opposite ratio, with an incidence rate of 11.3% in the western world and 18.8% in developing countries [1].

2. Liver Cancer and Its Origin

Liver cancer is the sixth most diagnosed cancer type worldwide, and the third leading cause of cancer mortality in 2020 [1]. By incidence, China is ranked first, accounting for 46.7% of new cases of liver cancer last year, followed by the USA with 4.5% of new cases, with both being linked to an increased presence of metabolic disorders [1,4]. In twenty countries located predominantly in Sub-Saharan Africa and South-Eastern Asia, liver cancer is the leading cause of death due to its association with viral infections prevalent in these countries [5]. Liver cancer is relatively rare in Germany; however, it is one of the most common cancer-related deaths due to its high mortality rate and poor prognosis. In 2016, the incidence rate of liver cancer in Germany was approximately 9000 new cases per year and nearly 8000 deaths. The frequency of liver cancer in women is less than half of that in men, while the relative 5-year survival rate is 15% for both genders [6].
Liver cancer is a general denomination, and each form of cancer is named after the cell type that becomes cancerous. For example, hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC) are the most frequently occurring types of primary liver cancer, and together are among the most common cancers worldwide, with more than 900,000 new cases globally in 2020 [1]. In particular, HCC is the most common type of liver cancer, accounting for 75–85% of all liver cancers and resulting in more than 700,000 deaths/year worldwide [1]. It is the fifth most often diagnosed cancer in males, and the ninth most diagnosed in females [1]. HCC originates from the predominant type of liver parenchymal cells, hepatocytes, and usually develops within the presence of advanced chronic liver disease [5,7]. In contrast to HCC, ICC arises primarily from the epithelial lining of the bile duct (intra- and extra-hepatic bile duct) that carries bile to the gall bladder. The relative incidence of ICC is around 10–15% of all liver cancers [1]. The third most common primary malignancy of the liver is hepatic angiosarcoma (HA), that begins in endothelial cells of blood or lymphatic vessels [8]. HA is particularly rare, accounting for just 2% of liver cancers, and is characterized by a very rapid progression of the illness, with an average life expectancy of around 10 months after the first diagnosis [8].

3. Risk Factors for HCC

The type of liver damage development is dependent upon the nature and severity of the lesion. In general, liver carcinogenesis is a progressive multi-stage process that proceeds with chronic inflammation, followed by fibrosis, and a culmination into cirrhosis. Cirrhosis is the most advanced stage of liver fibrosis and is associated with a higher risk of malignant liver transformation into HCC. Major risk factors for HCC include viral infections such as hepatitis B and C, chronic alcohol consumption, and non-alcoholic fatty liver disease (NAFLD) [9,10]. Moreover, aflatoxin exposure, Wilson’s disease, glycogen storage disease, α1-trypsin deficiency, autoimmune hepatitis, obesity, diabetes, and primary biliary cirrhosis are further examples of conditions that induce chronic liver damage and thereby increase the susceptibility to HCC [11,12,13,14,15,16,17,18,19].
In this review, we particularly focus on iron as a risk factor for HCC.

4. Iron as a Risk Factor for Liver Carcinogenesis

4.1. Mechanisms of Iron-Mediated Carcinogenesis

Iron is one of the most common elements on our planet and an essential micronutrient for numerous biological processes. It is involved in a variety of fundamental cellular and biochemical processes, including oxygen transport, DNA synthesis, cell survival, energy metabolism, and cellular respiration due to its integration within heme- and iron-sulphur cluster-containing proteins [20,21,22]. Typically, cancer cells require iron during all stages of tumor development, survival, proliferation, and metastasis [23,24,25,26,27,28,29,30]. Molecules involved in the regulation of iron metabolism, such as transferrin receptor, ferritin H, and divalent metal transporter 1 (DMT1), are often upregulated in cancer cells in order to induce iron accumulation [31,32]. Observations of enhanced iron uptake and iron retention in cancer cells suggest that cancers are dependent on iron.
Various iron-driven mechanisms have been described to induce HCC, or cancer in general. The most important of these is the generation of reactive oxygen species (ROS) and the resulting oxidative stress. The hypothesis that oxidative stress is the underlying cause of liver damage in iron-excess conditions has emerged from the role of ‘free’ iron which is present when the iron-binding capacities of plasma transferrin or intracellular iron-storage molecule ferritin are surpassed. The redox-active properties of iron account for its toxicity exerted in “Fenton-type” reactions, where iron mediates conversion of generally inactive H2O2 into extremely reactive ·HO (Fe2+ + H2O2 → reactive intermediates → Fe3+ + HO + OH−) [33]. Superoxide anions can reduce Fe3+ back to Fe2+ (Fe3+ + O·−2 → Fe2+ + O2), thus allowing iron to act as a catalyst of the reaction. A transient pool of redox-active iron is present in the cytosol and mirrors the overall iron status of the cell. Pathological accumulation of iron within cells and tissues enhances generation of ROS and elicits toxic effects, reflected in the production of hydroxyl and lipid radicals, which attack proteins, nucleic acids, and carbohydrates [34]. In addition, ROS promotes carcinogenesis via mutagenesis, genomic instability, and DNA repair defects, thereby compromising cell viability and promoting cell death [35,36]. Besides the direct role of ROS in promoting carcinogenesis, ROS can serve as secondary messengers in intracellular signaling cascades, promoting and maintaining the oncogenic phenotype of cancer cells [37,38].

4.2. Role of Cellular Iron Metabolism in the Pathogenesis of HCC

Iron metabolism is a tightly controlled process at the cellular level. Cellular iron levels are regulated by F-box and leucine-rich repeat protein 5 (FBXL5), and iron regulatory protein 2 (IRP2) [39]. A recent publication from Nakayama’s lab described the importance of FBXL5-mediated cellular iron homeostasis in liver carcinogenesis [40]. Mice lacking Fbxl5 are characterized as having excessive iron accumulation, resulting in subsequent oxidative stress, inflammation, and IRP2 accumulation. Exposure of Fbxl5 knockout mice to the chemical carcinogen, diethylnitrosamine (DEN), or to virus-induced HCC, resulted in an increased mutational load and liver tumor formation [40]. Deletion of Irp2 restored the lack of FBXL5 in the DEN model [40]. Moreover, low levels of Fbxl5 expression in HCC patients were associated with a poor disease prognosis. This data highlights the importance of strict regulation of cellular iron levels, a major elicitor of oxidative stress, in protection against hepatocarcinogenesis.

4.3. Role of Systemic Iron Overload in the Pathogenesis of HCC

Likewise, iron metabolism is a precisely controlled process at the systemic level. The small hepatocyte-derived peptide hormone, hepcidin, synchronizes systemic iron fluxes to regulate adequate supplies of iron to tissues and cells, while preventing iron overload and iron-related toxicity [41,42,43]. Hepcidin acts by binding to the cellular iron exporter ferroportin (FPN) to trigger its internalization and degradation [44]. Hepcidin expression is regulated by several signals, including plasma iron levels, hepatic iron stores, inflammatory states, erythropoietic activity, and hypoxia [43].
Systemic iron overload, due to dietary, environmental, or genetic factors, can significantly promote tumorigenesis. In particular, excessive iron in the liver may act both directly and indirectly to induce carcinogenesis. One such direct role of excessive iron in the pathogenesis of HCC was confirmed in an animal model that was fed an iron-rich diet, which resulted in HCC developing in the absence of fibrosis and cirrhosis [45]. The findings by this study confirm a direct mutagenic and hepatocarcinogenic effect of free iron, elicited through the generation of ROS and oxidative damage [45]. The contribution of iron overload in cancer development is well-documented in patients suffering from a genetic iron overload disorder (hereditary hemochromatosis, HH), caused by mutations within the HFE gene. HFE-HH is characterized by excessive iron absorption from the diet and increased iron deposition in tissues [46,47,48]. Genetic data in mice and patients demonstrated a relative deficiency of iron-hormone hepcidin to be the hallmark of HFE-HH disorders [49,50,51,52,53,54]. Since there is no regulated mechanism to remove the excess of iron, progressive iron accumulation in various tissues occurs, which may consequently cause serious organ dysfunctions [46,55,56,57,58]. Excessive iron overload exerts a toxic effect on the liver and correlates with an increased risk of cirrhosis [59,60]. It has been widely reported that mutations in the HFE gene correlate with increased cancer risk [61], and that HH patients have a 20- to 200-fold higher probability to develop liver cancer compared to control patients, culminating in deaths in 45% of all HCC patients [59,62,63,64]. Moreover, a meta-analysis study identified a statistically significant association between the HFE H63D polymorphism and hepatocellular carcinoma, especially in Asian and African populations [65]. Interestingly, HCC has been documented to develop in iron-overload patients even in the context of non-fibrotic and non-cirrhotic livers, suggesting a more direct role of hepatic iron overload in triggering the malignant transformation of hepatocytes [66,67,68,69]. Of note, HFE-HH patients show higher rates of p53 mutations (64–71%), compared to sporadic HCC patients, supporting a role of p53 in the hepatocarcinogenesis induced by iron overload [70,71]. p53 is one of the most common onco-suppressor genes mutated in cancer [72]. The importance of p53 in tumor suppression is due to its ability to act as a transcription factor, and to regulate a series of target genes necessary for cell survival and death [73]. An earlier study by Hussain et al. demonstrated that increased oxidative stress (due to high levels of etheno-DNA adducts formed from oxyradical-induced lipid peroxidation) in non-tumorous human liver of HFE-HH patients, caused mutations in the p53 tumor suppressor gene [74]. More recent studies showed that iron overload directly decreased p53 protein level and its activity in the liver, thereby facilitating hepatocarcinogenesis [75]. Consistently, p53 expression is decreased in the liver of Hfe−/− mice and in wild-type mice fed with an iron-rich diet [75]. The molecular mechanism behind p53 vanishing has been ascertained in HepG2 cell lines: iron-containing porphyrin (heme) binds to p53 at the C-terminal end of the protein, thus interfering with p53-DNA interactions and triggering both the nuclear export and cytosolic degradation of p53 via proteasomes [75]. Additionally, p53 is involved in the regulation of key iron sensors such as iron-sulphur cluster assembly enzyme (ISCU) and hepcidin. More precisely, p53 controls the intracellular iron pool by specifically modulating IRP1 RNA binding activity via transcriptional regulation of the ISCU [76,77]. Furthermore, the promoter of hepcidin contains putative p53-responsive elements and thus can be activated by p53 and decreased when p53 is silenced [78]. Therefore, following chronic iron overload, increased intracellular iron levels, the presence of oxidative stress, and reduced p53 activity may co-promote hepatocarcinogenesis.
Based on the above findings, it would be paramount to determine if p53 expression is decreased in other forms of HH, such as those caused by the mutations in hepcidin, hemojuvelin, and ferroportin genes. These investigations may reveal whether low p53 levels may be considered a common denominator of HCC in HH conditions. Moreover, given that iron chelators such as desferrioxamine show an antitumor effect in patients with advanced HCC, it is crucial to investigate if iron chelation would result in the normalization of p53 expression in the liver of HH-mice and patients [79].

5. Iron as a Co-Risk Factor in the Pathogenesis of HCC

Iron may also act synergistically with other risk factors in the pathogenesis of HCC, since chronic liver diseases such as chronic hepatitis, alcoholic liver disease, and NAFLD, associated with insulin resistance and the metabolic syndrome, are associated with iron overload.

5.1. Iron Overload and Viral Hepatitis

Hepatitis B virus (HBV) and hepatitis C virus (HCV) are major causes of viral hepatitis that lead to the development of cirrhosis and HCC [80,81]. The prevalence of HCC development in HBV carriers is up to 20-fold higher than the rate in non-carriers, as the DNA of HBV can integrate and replicate within cells, giving rise to chronic inflammation which is a predominant cause for fibrosis and cirrhosis [82,83]. It is estimated that 70–90% of HBV-associated HCC cases were identified in patients with cirrhotic liver [84]. Apart from random integration, HBV is able to disrupt cell cycle checkpoints and to induce chromosomal instability by protein HBx, thereby reinforcing the malignant transformation of hepatocytes. In addition to the prolonged HBV contagious period, HCV infection contributes as a major risk factor for liver cirrhosis [85]. Markedly, HCV-infected patients have an approximately 17-fold increased risk of developing HCC [86]. However, due to the RNA genome, the means by which HCV facilitates hepatocarcinogenesis differ greatly from mechanisms induced by HBV. So far, it is evident that structural and non-structural proteins of HCV are able to potentiate oncogenic transformation of hepatocytes by encouraging cell proliferation, DNA synthesis, steatosis, inflammatory processes, mitochondrial dysfunction, and insulin resistance, all of which lead to oxidative stress, genetic instability, and DNA damage; with cirrhosis and HCC as a potential outcome [87]. HCC risk drastically increases at the cirrhotic liver stage, suggesting a close association. The corresponding interplay of inflammatory responses, gene activation, and viral clearance suppression results in a conditioned environment that promotes cellular mutations leading to HCC.
HCV infection has been demonstrated to alter systemic and cellular iron homeostasis and its regulator hepcidin [88]. In the first phase of infection, hepcidin is more highly expressed in order to sustain the virus internalization though the iron importer, transferrin receptor 1 (TfR1) [89]. TfR1 was identified as a fundamental factor for HCV import in hepatocytes. Upon the binding of viral particles, TfR1 undergoes endocytosis, thus translocating viruses into intracellular compartments [89,90]. Indeed, TfR1 knockdown and antibody blocking inhibit HCV infection in Huh7 hepatoma cells [89]. In the following chronic phase, HCV infection leads to the downregulation of hepcidin in the plasma, with a resulting increase in iron absorption, as patient data clearly show [91,92]. Accordingly, these patients displayed increased transferrin saturation, raising the concentration of plasma ferritin and iron deposition in the liver [93]. Of interest is that the higher the iron levels are, the lower the response to the canonical therapy used for HCV infection is (interferon and ribavirin treatment) [94]. Moreover, the reduction in excess liver and body iron stores by phlebotomy was shown to ameliorate the course of hepatitis C and significantly improved the interferon effect [95,96].
Up to now, it is clear that HCV infection can induce iron overload in the liver and then behave as cofactors to worsen hepatic disease, characterized by advanced fibrosis, cirrhosis, and eventually even HCC. Yet, no data have demonstrated that iron increases susceptibility to HCV; quite the opposite, many studies now agree that the presence of HFE mutations alone does not significantly increase the risk of HCV disease [97].

5.2. Iron Overload and Chronic Alcoholic Liver Disease

Chronic alcohol consumption is a risk factor for the development of liver cancer, as demonstrated by its contribution to 30% of global HCC deaths [98,99,100,101]. Alcohol consumption increases the incidence of HCC in conjunction with other risk factors for chronic liver disease, especially viral hepatitis, diabetes, and obesity [102,103]. Chronic alcohol intake alters the liver architecture and compromises the functional capacity of the liver by triggering steatosis, steatohepatitis, and cirrhosis [104,105]. These pathological events are subsequently sustained and participate in the induction of carcinogenic processes. The mechanisms by which alcohol can mediate carcinogenesis are well known and were recently reviewed in [105]. Ethanol is first metabolized into acetaldehyde, which causes major cell damage by forming DNA and protein adducts [105]. In addition, alcohol increases gut permeability and the translocation of bacteria-derived lipopolysaccharide (LPS) from the gut to the liver, where the Kupffer cells start producing pro-inflammatory cytokines, triggering major biological pathways involved in hepatocarcinogenesis [106,107]. Moreover, the elevated production of cytochrome P450 2E1 (CYP2E1) and iron-induced ROS further aggravates the liver function by the impairment of antioxidant defenses and DNA repair mechanisms [108]. Hepatic iron accumulation and chronic alcohol consumption have long been associated together [109]. High liver iron was found to be predictive of HCC development or death in patients with alcoholic cirrhosis [110,111]. Both in vitro and in vivo models show that alcohol exposure directly downregulates hepcidin and simultaneously increases the expression of iron transporter proteins (DMT1 and FPN) in the duodenum, exacerbating the iron overload condition [112]. This effect of alcohol on hepcidin can be abolished with antioxidant treatment, pointing out the essential role of oxidative stress in the regulation of hepcidin expression [112]. In conclusion, alcohol abrogates the protective effect of hepcidin in iron overload by rendering the synthesis of hepcidin in the liver insensitive to body iron levels. Deregulation of hepcidin synthesis in the liver may be one of the underlying mechanisms by which alcohol consumption leads to iron overload.

5.3. Iron Overload as a Risk Factor in Progression of NAFLD to Non-Alcoholic Steatohepatitis (NASH), Cirrhosis and HCC

In several studies conducted in western countries, 30 to 40% of patients with HCC did not have chronic hepatitis infection, implying the presence of other causes of disease such as fatty liver disease, due to either alcohol abuse or NAFLD. A meta-analysis of 86 studies on 8.5 million subjects from 22 countries revealed that the global prevalence of NAFLD was 25.24% [113]. In particular, in the USA, 83.1 million people (~25% of the population) were diagnosed with NAFLD in 2015 and that number is expected to increase to 100.9 million by 2030 [114]. The pandemic of NAFLD represents the heaviest burden among modern liver diseases, not only due to its epidemiology, but also because of the risk of progression to cirrhosis and its associated complications, such as hepatic decompensation, HCC, and death. One study demonstrated that the standardized incidence ratio of HCC in patients with NAFLD was 4.4 after 16 years of follow-up [115]. The prevalence of NAFLD-related HCC is also rising worldwide and 4–22% of HCC cases in western countries are now attributed to NAFLD [115,116,117,118]. Meanwhile, in Asia, where viral hepatitis remains endemic, 1–2% of HCC cases are attributable to NAFLD [119,120]. Moreover, non-alcoholic fatty liver disease is present in up to 90% of all obese people and in up to 70% of people with type 2 diabetes (T2D), being a further possible risk factor for HCC [121,122,123,124].
NAFLD is a broad spectrum of liver diseases ranging from simple steatosis, such as the presence of fatty liver alone, to non-alcoholic steatohepatitis (NASH) and cirrhosis, that may progress to HCC [125,126]. A major hallmark of NAFLD is the accumulation of triglycerides in the cytoplasm of hepatocytes, that arises from an imbalance between lipid acquisition (i.e., fatty acid uptake and de novo lipogenesis) and lipid removal (i.e., mitochondrial fatty acid oxidation and export as a component of VLDL (very-low-density lipoprotein) particles [127,128].
Accumulating evidence suggests a link between altered iron metabolism and NAFLD. Iron has been implicated in the pathogenesis of NAFLD via oxidative stress. An excess of iron is the major cause of ROS production and oxidative stress has been demonstrated to be the starting point of the hepatic and extrahepatic damage in NAFLD [129,130]. Hepatic iron loading was shown to up-regulate cholesterol biosynthesis pathways and has been proposed as an additional mechanism contributing to iron-induced liver injury in fatty liver disease [131]. Concurrently, the presence of oxidative stress in the hepatocytes stimulates liver stellate cells to increase the formation of extracellular matrix components, which often causes fibrosis and cirrhosis and might lead to HCC development [19,47]. Last but not least, iron may also contribute to liver injury in NAFLD by generating endoplasmic reticulum (ER) stress. Tan et al. demonstrated that iron overload in conditions of alcohol- and obesity-induced liver injury significantly increased the extent of liver injury, steatohepatitis, and liver fibrosis through the induction of unfolded protein response and ER stress [132,133].
Progression from simple steatosis to NASH, which is a serious risk factor for cirrhosis and HCC, correlates with an increased concentration of iron in hepatocytes, increased production of lipid peroxides, organelle dysfunction, and eventually cell death [134,135]. In NASH, oxidative stress leads to cell death via depletion of ATP, NAD, and glutathione, and by direct damage to DNA, lipids, and proteins within hepatocytes [130]. Furthermore, oxidative stress leads to an increase in the production of pro-inflammatory cytokines and a fibrogenic response [130].
Iron accumulation in the liver is believed to be an aggravating factor. Elevated levels of serum ferritin, iron, and transferrin saturation, and an increase in hepatic iron staining were detected in more than 30% of patients with NAFLD and correlated with insulin resistance, advanced fibrosis, NASH, and an increased long-term risk of death [136,137,138,139]. Nelson et al. performed a study in which they divided NAFLD patients into three groups depending on iron localization in the liver cells (hepatocytes, endothelial cells, and Kupffer cells) [140]. The authors demonstrated that iron retention in endothelial cells and Kupffer cells aggravated NAFLD and increased fibrosis, inflammation, hepatocyte ballooning, and definite NASH compared to patients with iron deposition in hepatocytes and to those having mixed iron patterns [140]. Sorrentino et al. demonstrated that the hepatic iron was significantly higher in HCC-NASH patients than in HCC-free NASH controls, and, more importantly, in the former group the liver iron overload was mainly associated with Kupffer and sinusoidal cells [141]. Similarly, HFE-HH patients with NAFLD have more severe liver damage due to the iron deposition in hepatocytes and in non-parenchymal cells, and furthermore, these patients develop NASH even in the presence of less severe metabolic abnormalities, suggesting that iron overload may contribute to NAFLD onset and exacerbate the progression to NASH [142,143,144]. By contrast, hepatocellular iron excess in mouse models of hemochromatosis (such as Hfe−/−, Hjv−/−, and Hfe−/−/Hjv−/− double knock-out) did not aggravate diet-induced steatosis to steatohepatitis or early liver fibrosis [145]. Collectively, these studies reveal a still unresolved controversy in the field on whether iron excess in hepatocytes or Kupffer cells, or both, acts as a driver for progression of NAFLD to NASH and advanced liver disease.
Recently, Tsurusaki et al., showed that ferroptosis, a type of “programmed necrosis” described as an iron- and lipid hydroperoxide-dependent nonapoptotic cell death, may be the trigger for initiating inflammation in NAFLD and could be observed as the first event towards steatohepatitis [146,147]. Nonetheless, more investigations are needed to demonstrate the implication of ferroptosis in NASH and the possible effectiveness of ferroptosis inhibition, both in murine models and patients with NASH.

5.4. Iron as a Risk Factor in NAFLD Associated with Insulin Resistance and the Metabolic Syndrome

T2D has been associated with an augmented risk of developing different types of cancers, including liver cancers [148,149,150,151,152,153,154]. In a large cohort study, Inoue et al. showed that the risk of malignancy was 2.8-fold higher in patients with T2D than in those without [149]. Similar conclusions were drawn from case-control studies conducted in the population of the USA, where the proportion of HCC patients with diabetes was twice the controls, demonstrating that T2D increased the risk for HCC [150]. Although further evaluation is required to draw definitive conclusions, it is estimated that T2D can increase the risk of HCC by roughly two- to three-fold, and the development of HCC is the most worrisome liver-related complication in diabetic patients. Clinical observation studies demonstrated an increased incidence of T2D in patients with pathologic iron overload, including HH (13–22% prevalence), beta thalassemia (6–14% incidence), and Friedreich ataxia [155,156,157,158,159,160,161]. Importantly, even high levels of dietary iron or transfusional iron overload impart an increase in diabetes-related risks [162,163,164,165]. Iron overload in hepatocytes may induce insulin resistance via oxidative stress, increased accumulation of ROS, and subsequent impairment of signaling cascades such as phosphatidylinositol 3-kinase (PI-3K), adiponectin, interleukin, and TNF-α [166,167,168,169,170]. Placing obese mice or mice fed a high-fat diet on iron chelation therapy, or on an iron-deficient diet, resulted in significant protection from diabetes that was relayed by both increased insulin secretion and sensitivity [171]. Although the exact molecular link between iron and insulin resistance remains unknown, it is postulated that dysregulated expression of hepcidin and of molecules involved in cellular iron export may play a role. Interestingly, metformin, the first-line drug for T2D therapy, was recently shown to modulate iron metabolism by inhibiting bone morphogenetic protein 6 (BMP6)-induced hepcidin expression independently from protein kinase AMP-activated catalytic (AMPK) activation (canonical metformin activity) [172,173]. Metformin increases the orphan nuclear receptor SHP which interacts with SMAD1 (small mothers against decapentaplegic) proteins and suppresses BMP6-mediated recruitment of the SMAD1–SMAD4 complex to the hepcidin gene promoter [172,174]. Another proposed mechanism by which metformin regulates hepcidin levels is through inhibition of STAT3 activation by promoting the proteasome-mediated degradation of JAK2 [175]. Indeed, metformin pre-treatment significantly reduced the IL-6 response with a consequent reduction in pSTAT3 and hepcidin levels [175]. Moreover, metformin was associated with decreased serum hepcidin levels in Chinese T2D patients [175]. Thus, beneficial effects of metformin in inhibiting tumorigenesis could be, at least in part, ascribed to its ability to modulate hepcidin expression and, thus, the iron metabolism, since a reduction of 31% in cancer incidence was noted in diabetic patients undergoing metformin treatment [176]. Given the very high prevalence of the metabolic syndrome worldwide, and that even a small increase in risk related to obesity or diabetes could potentially translate into a large number of HCC cases, future investigations into the role of iron and its metabolism in the progression of HCC in the background of metabolic syndrome are urgently needed.

6. Conclusions

Iron overload, as it is observed in patients with hereditary hemochromatosis, is an important risk factor in liver carcinogenesis. The accumulation of iron in hepatocytes promotes oxidative stress, cell death, and compulsory liver regeneration. These factors lead to an accumulation of DNA damage, and subsequently to the malignant transformation of hepatocytes. There is experimental evidence that iron supports a carcinogenic or co-carcinogenic role in carcinogenesis. So far it is not clear if the accumulation of iron is a main driver for liver carcinogenesis, but it is definitely known that iron overload is an additional factor which will lead (in combination with disease models of liver cancer) to an increased mutational load and to tumor formation. Therefore, the modification of the cellular and systemic iron content might demonstrate an important factor for liver cancer prevention or a delay in liver cancer progression. Due to the limited understanding of the role of iron in liver cancer, the biology of iron metabolism, and the dysfunctional regulation of iron, it offers new challenges to unravel liver tumor pathogenesis and to develop new therapeutic strategies based on iron regulation.

Author Contributions

Conceptualization, M.V.S.; writing—original draft preparation, R.P. and M.V.S.; writing—review and editing, R.P., A.L., and M.V.S.; funding acquisition, M.V.S. All authors have read and agreed to the published version of the manuscript. We thank Peter Steele-Perkins for the critical reading of the manuscript.

Funding

Funded by the DFG, grant number VU 75/4-1 (to M.V.S.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021. [Google Scholar] [CrossRef] [PubMed]
  2. Parkin, D.M.; Bray, F.; Ferlay, J.; Pisani, P. Global cancer statistics, 2002. CA Cancer J. Clin. 2005, 55, 74–108. [Google Scholar] [CrossRef] [PubMed]
  3. Fidler, M.M.; Bray, F.; Soerjomataram, I. The global cancer burden and human development: A review. Scand. J. Public Health 2018, 46, 27–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. GLOBOCAN 2020 Graph Production: Global Cancer Observatory © International Agency for Research on Cancer 2021. Available online: https://gco.iarc.fr/today/data/factsheets/populations/900-world-fact-sheets.pdf (accessed on 15 April 2021).
  5. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Cancer in Germany in 2015/2016, 12th ed.; Robert Koch Institute, the Association of Population-Based Cancer Registries in Germany, Eds.; Berlin, Germany, 2020. Available online: https://www.krebsdaten.de/Krebs/EN/Content/Publications/Cancer_in_Germany/cancer_chapters_2015_2016/cancer_germany_2015_2016.pdf?__blob=publicationFile (accessed on 15 April 2021). [CrossRef]
  7. Torre, L.A.; Bray, F.; Siegel, R.L.; Ferlay, J.; Lortet-Tieulent, J.; Jemal, A. Global cancer statistics, 2012. CA Cancer J. Clin. 2015, 65, 87–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Mani, H.; Van Thiel, D.H. Mesenchymal tumors of the liver. Clin. Liver Dis. 2001, 5, 219–257. [Google Scholar] [CrossRef]
  9. El-Serag, H.B. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology 2012, 142, 1264–1273.e1261. [Google Scholar] [CrossRef] [Green Version]
  10. Zoller, H.; Tilg, H. Nonalcoholic fatty liver disease and hepatocellular carcinoma. Metabolism 2016, 65, 1151–1160. [Google Scholar] [CrossRef]
  11. Hamid, A.S.; Tesfamariam, I.G.; Zhang, Y.; Zhang, Z.G. Aflatoxin B1-induced hepatocellular carcinoma in developing countries: Geographical distribution, mechanism of action and prevention. Oncol. Lett. 2013, 5, 1087–1092. [Google Scholar] [CrossRef] [Green Version]
  12. Xu, R.; Hajdu, C.H. Wilson disease and hepatocellular carcinoma. Gastroenterol. Hepatol. 2008, 4, 438–439. [Google Scholar]
  13. Manzia, T.M.; Angelico, R.; Toti, L.; Cillis, A.; Ciano, P.; Orlando, G.; Anselmo, A.; Angelico, M.; Tisone, G. Glycogen storage disease type Ia and VI associated with hepatocellular carcinoma: Two case reports. Transplant. Proc. 2011, 43, 1181–1183. [Google Scholar] [CrossRef] [PubMed]
  14. van Ginkel, W.G.; Gouw, A.S.; van der Jagt, E.J.; de Jong, K.P.; Verkade, H.J.; van Spronsen, F.J. Hepatocellular carcinoma in tyrosinemia type 1 without clear increase of AFP. Pediatrics 2015, 135, e749–e752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. El-Serag, H.B.; Kanwal, F. Obesity and hepatocellular carcinoma: Hype and reality. Hepatology 2014, 60, 779–781. [Google Scholar] [CrossRef] [PubMed]
  16. Heidelbaugh, J.J.; Bruderly, M. Cirrhosis and chronic liver failure: Part I. Diagnosis and evaluation. Am. Fam. Physician 2006, 74, 756–762. [Google Scholar] [PubMed]
  17. Sanyal, A.J.; Yoon, S.K.; Lencioni, R. The etiology of hepatocellular carcinoma and consequences for treatment. Oncologist 2010, 15 (Suppl. 4), 14–22. [Google Scholar] [CrossRef] [Green Version]
  18. Findor, J.; He, X.S.; Sord, J.; Terg, R.; Gershwin, M.E. Primary biliary cirrhosis and hepatocellular carcinoma. Autoimmun. Rev. 2002, 1, 220–225. [Google Scholar] [CrossRef]
  19. Kew, M.C. Hepatic iron overload and hepatocellular carcinoma. Cancer Lett. 2009, 286, 38–43. [Google Scholar] [CrossRef] [Green Version]
  20. Veatch, J.R.; McMurray, M.A.; Nelson, Z.W.; Gottschling, D.E. Mitochondrial dysfunction leads to nuclear genome instability via an iron-sulfur cluster defect. Cell 2009, 137, 1247–1258. [Google Scholar] [CrossRef] [Green Version]
  21. Puig, S.; Ramos-Alonso, L.; Romero, A.M.; Martinez-Pastor, M.T. The elemental role of iron in DNA synthesis and repair. Metallomics 2017, 9, 1483–1500. [Google Scholar] [CrossRef] [Green Version]
  22. Oexle, H.; Gnaiger, E.; Weiss, G. Iron-dependent changes in cellular energy metabolism: Influence on citric acid cycle and oxidative phosphorylation. Biochim. Biophys. Acta 1999, 1413, 99–107. [Google Scholar] [CrossRef] [Green Version]
  23. Ludwig, H.; Evstatiev, R.; Kornek, G.; Aapro, M.; Bauernhofer, T.; Buxhofer-Ausch, V.; Fridrik, M.; Geissler, D.; Geissler, K.; Gisslinger, H.; et al. Iron metabolism and iron supplementation in cancer patients. Wien. Klin. Wochenschr. 2015, 127, 907–919. [Google Scholar] [CrossRef] [Green Version]
  24. Torti, S.V.; Torti, F.M. Iron and cancer: More ore to be mined. Nat. Rev. Cancer 2013, 13, 342–355. [Google Scholar] [CrossRef] [Green Version]
  25. Daniels, T.R.; Bernabeu, E.; Rodriguez, J.A.; Patel, S.; Kozman, M.; Chiappetta, D.A.; Holler, E.; Ljubimova, J.Y.; Helguera, G.; Penichet, M.L. The transferrin receptor and the targeted delivery of therapeutic agents against cancer. Biochim. Biophys. Acta 2012, 1820, 291–317. [Google Scholar] [CrossRef] [Green Version]
  26. Forciniti, S.; Greco, L.; Grizzi, F.; Malesci, A.; Laghi, L. Iron Metabolism in Cancer Progression. Int. J. Mol. Sci. 2020, 21, 2257. [Google Scholar] [CrossRef] [Green Version]
  27. Torti, S.V.; Manz, D.H.; Paul, B.T.; Blanchette-Farra, N.; Torti, F.M. Iron and Cancer. Annu. Rev. Nutr. 2018, 38, 97–125. [Google Scholar] [CrossRef]
  28. Zhang, C.; Zhang, F. Iron homeostasis and tumorigenesis: Molecular mechanisms and therapeutic opportunities. Protein Cell 2015, 6, 88–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Brown, R.A.M.; Richardson, K.L.; Kabir, T.D.; Trinder, D.; Ganss, R.; Leedman, P.J. Altered Iron Metabolism and Impact in Cancer Biology, Metastasis, and Immunology. Front. Oncol. 2020, 10, 476. [Google Scholar] [CrossRef] [PubMed]
  30. Jung, M.; Mertens, C.; Tomat, E.; Brune, B. Iron as a Central Player and Promising Target in Cancer Progression. Int. J. Mol. Sci. 2019, 20, 273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Adachi, M.; Kai, K.; Yamaji, K.; Ide, T.; Noshiro, H.; Kawaguchi, A.; Aishima, S. Transferrin receptor 1 overexpression is associated with tumour de-differentiation and acts as a potential prognostic indicator of hepatocellular carcinoma. Histopathology 2019, 75, 63–73. [Google Scholar] [CrossRef]
  32. Hsu, M.Y.; Mina, E.; Roetto, A.; Porporato, P.E. Iron: An Essential Element of Cancer Metabolism. Cells 2020, 9, 2591. [Google Scholar] [CrossRef] [PubMed]
  33. Fenton, H.J.H. LXXIII—Oxidation of tartaric acid in presence of iron. J. Chem. Soc. Trans. 1894, 65, 899–910. [Google Scholar] [CrossRef] [Green Version]
  34. D’Autreaux, B.; Toledano, M.B. ROS as signalling molecules: Mechanisms that generate specificity in ROS homeostasis. Nat. Rev. Mol. Cell Biol. 2007, 8, 813–824. [Google Scholar] [CrossRef] [PubMed]
  35. Paul, V.D.; Lill, R. Biogenesis of cytosolic and nuclear iron-sulfur proteins and their role in genome stability. Biochim. Biophys. Acta 2015, 1853, 1528–1539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Zhang, C. Essential functions of iron-requiring proteins in DNA replication, repair and cell cycle control. Protein Cell 2014, 5, 750–760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Huang, X. Does iron have a role in breast cancer? Lancet Oncol. 2008, 9, 803–807. [Google Scholar] [CrossRef] [Green Version]
  38. Storz, P. Reactive oxygen species in tumor progression. Front. Biosci. 2005, 10, 1881–1896. [Google Scholar] [CrossRef] [Green Version]
  39. Moroishi, T.; Nishiyama, M.; Takeda, Y.; Iwai, K.; Nakayama, K.I. The FBXL5-IRP2 axis is integral to control of iron metabolism in vivo. Cell Metab. 2011, 14, 339–351. [Google Scholar] [CrossRef] [Green Version]
  40. Muto, Y.; Moroishi, T.; Ichihara, K.; Nishiyama, M.; Shimizu, H.; Eguchi, H.; Moriya, K.; Koike, K.; Mimori, K.; Mori, M.; et al. Disruption of FBXL5-mediated cellular iron homeostasis promotes liver carcinogenesis. J. Exp. Med. 2019, 216, 950–965. [Google Scholar] [CrossRef] [Green Version]
  41. Camaschella, C.; Nai, A.; Silvestri, L. Iron metabolism and iron disorders revisited in the hepcidin era. Haematologica 2020, 105, 260–272. [Google Scholar] [CrossRef] [Green Version]
  42. Muckenthaler, M.U.; Rivella, S.; Hentze, M.W.; Galy, B. A Red Carpet for Iron Metabolism. Cell 2017, 168, 344–361. [Google Scholar] [CrossRef] [Green Version]
  43. Hentze, M.W.; Muckenthaler, M.U.; Galy, B.; Camaschella, C. Two to tango: Regulation of Mammalian iron metabolism. Cell 2010, 142, 24–38. [Google Scholar] [CrossRef] [Green Version]
  44. Ganz, T. Systemic iron homeostasis. Physiol. Rev. 2013, 93, 1721–1741. [Google Scholar] [CrossRef] [Green Version]
  45. Asare, G.A.; Mossanda, K.S.; Kew, M.C.; Paterson, A.C.; Kahler-Venter, C.P.; Siziba, K. Hepatocellular carcinoma caused by iron overload: A possible mechanism of direct hepatocarcinogenicity. Toxicology 2006, 219, 41–52. [Google Scholar] [CrossRef] [PubMed]
  46. Pietrangelo, A. Hereditary hemochromatosis: Pathogenesis, diagnosis, and treatment. Gastroenterology 2010, 139, 393–408, 408 e391–392. [Google Scholar] [CrossRef] [PubMed]
  47. Sebastiani, G.; Walker, A.P. HFE gene in primary and secondary hepatic iron overload. World J. Gastroenterol. 2007, 13, 4673–4689. [Google Scholar] [CrossRef]
  48. Vujic, M. Molecular basis of HFE-hemochromatosis. Front. Pharmacol. 2014, 5, 42. [Google Scholar] [CrossRef] [PubMed]
  49. Ahmad, K.A.; Ahmann, J.R.; Migas, M.C.; Waheed, A.; Britton, R.S.; Bacon, B.R.; Sly, W.S.; Fleming, R.E. Decreased liver hepcidin expression in the Hfe knockout mouse. Blood Cells Mol. Dis. 2002, 29, 361–366. [Google Scholar] [CrossRef]
  50. Bridle, K.R.; Frazer, D.M.; Wilkins, S.J.; Dixon, J.L.; Purdie, D.M.; Crawford, D.H.; Subramaniam, V.N.; Powell, L.W.; Anderson, G.J.; Ramm, G.A. Disrupted hepcidin regulation in HFE-associated haemochromatosis and the liver as a regulator of body iron homoeostasis. Lancet 2003, 361, 669–673. [Google Scholar] [CrossRef]
  51. Muckenthaler, M.; Roy, C.N.; Custodio, A.O.; Minana, B.; deGraaf, J.; Montross, L.K.; Andrews, N.C.; Hentze, M.W. Regulatory defects in liver and intestine implicate abnormal hepcidin and Cybrd1 expression in mouse hemochromatosis. Nat. Genet. 2003, 34, 102–107. [Google Scholar] [CrossRef] [PubMed]
  52. Lesbordes-Brion, J.C.; Viatte, L.; Bennoun, M.; Lou, D.Q.; Ramey, G.; Houbron, C.; Hamard, G.; Kahn, A.; Vaulont, S. Targeted disruption of the hepcidin 1 gene results in severe hemochromatosis. Blood 2006, 108, 1402–1405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Wallace, D.F.; Summerville, L.; Crampton, E.M.; Frazer, D.M.; Anderson, G.J.; Subramaniam, V.N. Combined deletion of Hfe and transferrin receptor 2 in mice leads to marked dysregulation of hepcidin and iron overload. Hepatology 2009, 50, 1992–2000. [Google Scholar] [CrossRef]
  54. Vujic Spasic, M.; Kiss, J.; Herrmann, T.; Galy, B.; Martinache, S.; Stolte, J.; Grone, H.J.; Stremmel, W.; Hentze, M.W.; Muckenthaler, M.U. Hfe acts in hepatocytes to prevent hemochromatosis. Cell Metab. 2008, 7, 173–178. [Google Scholar] [CrossRef] [Green Version]
  55. Davies, P.S.; Enns, C.A. Expression of the hereditary hemochromatosis protein HFE increases ferritin levels by inhibiting iron export in HT29 cells. J. Biol. Chem. 2004, 279, 25085–25092. [Google Scholar] [CrossRef] [Green Version]
  56. Camaschella, C. Understanding iron homeostasis through genetic analysis of hemochromatosis and related disorders. Blood 2005, 106, 3710–3717. [Google Scholar] [CrossRef] [Green Version]
  57. Guggenbuhl, P.; Deugnier, Y.; Boisdet, J.F.; Rolland, Y.; Perdriger, A.; Pawlotsky, Y.; Chales, G. Bone mineral density in men with genetic hemochromatosis and HFE gene mutation. Osteoporos. Int. 2005, 16, 1809–1814. [Google Scholar] [CrossRef] [PubMed]
  58. Wagner, A.; Alan, B.; Yilmaz, D.; Ahmad, M.; Liu, P.; Tangudu, N.K.; Tuckermann, J.P.; Vujic Spasic, M. Despite Genetic Iron Overload, Hfe-Hemochromatosis Mice Do Not Show Bone Loss. JBMR Plus 2019, 3, e10206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Kew, M.C. Hepatic iron overload and hepatocellular carcinoma. Liver Cancer 2014, 3, 31–40. [Google Scholar] [CrossRef] [PubMed]
  60. Macias-Rodriguez, R.U.; Inzaugarat, M.E.; Ruiz-Margain, A.; Nelson, L.J.; Trautwein, C.; Cubero, F.J. Reclassifying Hepatic Cell Death during Liver Damage: Ferroptosis-A Novel Form of Non-Apoptotic Cell Death? Int. J. Mol. Sci. 2020, 21, 1651. [Google Scholar] [CrossRef] [Green Version]
  61. Fargion, S.; Valenti, L.; Fracanzani, A.L. Hemochromatosis gene (HFE) mutations and cancer risk: Expanding the clinical manifestations of hereditary iron overload. Hepatology 2010, 51, 1119–1121. [Google Scholar] [CrossRef]
  62. 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. [Google Scholar] [CrossRef]
  63. Bradbear, R.A.; Bain, C.; Siskind, V.; Schofield, F.D.; Webb, S.; Axelsen, E.M.; Halliday, J.W.; Bassett, M.L.; Powell, L.W. Cohort study of internal malignancy in genetic hemochromatosis and other chronic nonalcoholic liver diseases. J. Natl. Cancer Inst. 1985, 75, 81–84. [Google Scholar]
  64. Fracanzani, A.L.; Conte, D.; Fraquelli, M.; Taioli, E.; Mattioli, M.; Losco, A.; Fargion, S. Increased cancer risk in a cohort of 230 patients with hereditary hemochromatosis in comparison to matched control patients with non-iron-related chronic liver disease. Hepatology 2001, 33, 647–651. [Google Scholar] [CrossRef]
  65. Shen, L.L.; Gu, D.Y.; Zhao, T.T.; Tang, C.J.; Xu, Y.; Chen, J.F. Implicating the H63D polymorphism in the HFE gene in increased incidence of solid cancers: A meta-analysis. Genet. Mol. Res. 2015, 14, 13735–13745. [Google Scholar] [CrossRef]
  66. Turlin, B.; Juguet, F.; Moirand, R.; Le Quilleuc, D.; Loreal, O.; Campion, J.P.; Launois, B.; Ramee, M.P.; Brissot, P.; Deugnier, Y. Increased liver iron stores in patients with hepatocellular carcinoma developed on a noncirrhotic liver. Hepatology 1995, 22, 446–450. [Google Scholar]
  67. Blanc, J.F.; De Ledinghen, V.; Bernard, P.H.; de Verneuil, H.; Winnock, M.; Le Bail, B.; Carles, J.; Saric, J.; Balabaud, C.; Bioulac-Sage, P. Increased incidence of HFE C282Y mutations in patients with iron overload and hepatocellular carcinoma developed in non-cirrhotic liver. J. Hepatol. 2000, 32, 805–811. [Google Scholar] [CrossRef]
  68. Bralet, M.P.; Regimbeau, J.M.; Pineau, P.; Dubois, S.; Loas, G.; Degos, F.; Valla, D.; Belghiti, J.; Degott, C.; Terris, B. Hepatocellular carcinoma occurring in nonfibrotic liver: Epidemiologic and histopathologic analysis of 80 French cases. Hepatology 2000, 32, 200–204. [Google Scholar] [CrossRef]
  69. Chung, H.; Kudo, M.; Kawasaki, T.; Kitano, M.; Minami, Y.; Suetomi, Y.; Onda, H. Hepatocellular carcinoma associated with secondary haemochromatosis in non-cirrhotic liver: A case report. Hepatol. Res. 2003, 26, 254–258. [Google Scholar] [CrossRef]
  70. Vautier, G.; Bomford, A.B.; Portmann, B.C.; Metivier, E.; Williams, R.; Ryder, S.D. p53 mutations in british patients with hepatocellular carcinoma: Clustering in genetic hemochromatosis. Gastroenterology 1999, 117, 154–160. [Google Scholar] [CrossRef]
  71. Marrogi, A.J.; Khan, M.A.; van Gijssel, H.E.; Welsh, J.A.; Rahim, H.; Demetris, A.J.; Kowdley, K.V.; Hussain, S.P.; Nair, J.; Bartsch, H.; et al. Oxidative stress and p53 mutations in the carcinogenesis of iron overload-associated hepatocellular carcinoma. J. Natl. Cancer Inst. 2001, 93, 1652–1655. [Google Scholar] [CrossRef] [Green Version]
  72. Kandoth, C.; McLellan, M.D.; Vandin, F.; Ye, K.; Niu, B.; Lu, C.; Xie, M.; Zhang, Q.; McMichael, J.F.; Wyczalkowski, M.A.; et al. Mutational landscape and significance across 12 major cancer types. Nature 2013, 502, 333–339. [Google Scholar] [CrossRef] [Green Version]
  73. Harms, K.; Nozell, S.; Chen, X. The common and distinct target genes of the p53 family transcription factors. Cell Mol. Life Sci. 2004, 61, 822–842. [Google Scholar] [CrossRef]
  74. Hussain, S.P.; Raja, K.; Amstad, P.A.; Sawyer, M.; Trudel, L.J.; Wogan, G.N.; Hofseth, L.J.; Shields, P.G.; Billiar, T.R.; Trautwein, C.; et al. Increased p53 mutation load in nontumorous human liver of wilson disease and hemochromatosis: Oxyradical overload diseases. Proc. Natl. Acad. Sci. USA 2000, 97, 12770–12775. [Google Scholar] [CrossRef] [Green Version]
  75. Shen, J.; Sheng, X.; Chang, Z.; Wu, Q.; Wang, S.; Xuan, Z.; Li, D.; Wu, Y.; Shang, Y.; Kong, X.; et al. Iron metabolism regulates p53 signaling through direct heme-p53 interaction and modulation of p53 localization, stability, and function. Cell Rep. 2014, 7, 180–193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Zhang, F.; Wang, W.; Tsuji, Y.; Torti, S.V.; Torti, F.M. Post-transcriptional modulation of iron homeostasis during p53-dependent growth arrest. J. Biol. Chem. 2008, 283, 33911–33918. [Google Scholar] [CrossRef] [Green Version]
  77. Funauchi, Y.; Tanikawa, C.; Yi Lo, P.H.; Mori, J.; Daigo, Y.; Takano, A.; Miyagi, Y.; Okawa, A.; Nakamura, Y.; Matsuda, K. Regulation of iron homeostasis by the p53-ISCU pathway. Sci. Rep. 2015, 5, 16497. [Google Scholar] [CrossRef] [Green Version]
  78. Weizer-Stern, O.; Adamsky, K.; Margalit, O.; Ashur-Fabian, O.; Givol, D.; Amariglio, N.; Rechavi, G. Hepcidin, a key regulator of iron metabolism, is transcriptionally activated by p53. Br. J. Haematol. 2007, 138, 253–262. [Google Scholar] [CrossRef]
  79. Yamasaki, T.; Terai, S.; Sakaida, I. Deferoxamine for advanced hepatocellular carcinoma. N. Engl. J. Med. 2011, 365, 576–578. [Google Scholar] [CrossRef]
  80. WHO. Hepatitis B. Available online: https://www.who.int/news-room/fact-sheets/detail/hepatitis-b (accessed on 27 July 2020).
  81. WHO. Hepatitis C. Available online: https://www.who.int/news-room/fact-sheets/detail/hepatitis-c (accessed on 27 July 2020).
  82. Shi, J.; Zhu, L.; Liu, S.; Xie, W.F. A meta-analysis of case-control studies on the combined effect of hepatitis B and C virus infections in causing hepatocellular carcinoma in China. Br. J. Cancer 2005, 92, 607–612. [Google Scholar] [CrossRef] [Green Version]
  83. Donato, F.; Boffetta, P.; Puoti, M. A meta-analysis of epidemiological studies on the combined effect of hepatitis B and C virus infections in causing hepatocellular carcinoma. Int. J. Cancer 1998, 75, 347–354. [Google Scholar] [CrossRef]
  84. Yang, J.D.; Kim, W.R.; Coelho, R.; Mettler, T.A.; Benson, J.T.; Sanderson, S.O.; Therneau, T.M.; Kim, B.; Roberts, L.R. Cirrhosis is present in most patients with hepatitis B and hepatocellular carcinoma. Clin. Gastroenterol. Hepatol. 2011, 9, 64–70. [Google Scholar] [CrossRef] [Green Version]
  85. Fattovich, G.; Giustina, G.; Degos, F.; Tremolada, F.; Diodati, G.; Almasio, P.; Nevens, F.; Solinas, A.; Mura, D.; Brouwer, J.T.; et al. Morbidity and mortality in compensated cirrhosis type C: A retrospective follow-up study of 384 patients. Gastroenterology 1997, 112, 463–472. [Google Scholar] [CrossRef] [PubMed]
  86. Donato, F.; Tagger, A.; Gelatti, U.; Parrinello, G.; Boffetta, P.; Albertini, A.; Decarli, A.; Trevisi, P.; Ribero, M.L.; Martelli, C.; et al. Alcohol and hepatocellular carcinoma: The effect of lifetime intake and hepatitis virus infections in men and women. Am. J. Epidemiol. 2002, 155, 323–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Irshad, M.; Gupta, P.; Irshad, K. Molecular basis of hepatocellular carcinoma induced by hepatitis C virus infection. World J. Hepatol. 2017, 9, 1305–1314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Mancinelli, R.; Rosa, L.; Cutone, A.; Lepanto, M.S.; Franchitto, A.; Onori, P.; Gaudio, E.; Valenti, P. Viral Hepatitis and Iron Dysregulation: Molecular Pathways and the Role of Lactoferrin. Molecules 2020, 25, 1997. [Google Scholar] [CrossRef] [PubMed]
  89. Martin, D.N.; Uprichard, S.L. Identification of transferrin receptor 1 as a hepatitis C virus entry factor. Proc. Natl. Acad. Sci. USA 2013, 110, 10777–10782. [Google Scholar] [CrossRef] [Green Version]
  90. Fillebeen, C.; Pantopoulos, K. Hepatitis C virus infection causes iron deficiency in Huh7.5.1 cells. PLoS ONE 2013, 8, e83307. [Google Scholar] [CrossRef]
  91. Girelli, D.; Pasino, M.; Goodnough, J.B.; Nemeth, E.; Guido, M.; Castagna, A.; Busti, F.; Campostrini, N.; Martinelli, N.; Vantini, I.; et al. Reduced serum hepcidin levels in patients with chronic hepatitis C. J. Hepatol. 2009, 51, 845–852. [Google Scholar] [CrossRef] [Green Version]
  92. Fujita, N.; Sugimoto, R.; Takeo, M.; Urawa, N.; Mifuji, R.; Tanaka, H.; Kobayashi, Y.; Iwasa, M.; Watanabe, S.; Adachi, Y.; et al. Hepcidin expression in the liver: Relatively low level in patients with chronic hepatitis C. Mol. Med. 2007, 13, 97–104. [Google Scholar] [CrossRef] [PubMed]
  93. Fujita, N.; Takei, Y. Iron, hepatitis C virus, and hepatocellular carcinoma: Iron reduction preaches the gospel for chronic hepatitis C. J. Gastroenterol. 2007, 42, 923–926. [Google Scholar] [CrossRef]
  94. Lange, C.M.; Kutalik, Z.; Morikawa, K.; Bibert, S.; Cerny, A.; Dollenmaier, G.; Dufour, J.F.; Gerlach, T.J.; Heim, M.H.; Malinverni, R.; 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. [Google Scholar] [CrossRef]
  95. Franchini, M.; Targher, G.; Capra, F.; Montagnana, M.; Lippi, G. The effect of iron depletion on chronic hepatitis C virus infection. Hepatol. Int. 2008, 2, 335–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Desai, T.K.; Jamil, L.H.; Balasubramaniam, M.; Koff, R.; Bonkovsky, H.L. Phlebotomy improves therapeutic response to interferon in patients with chronic hepatitis C: A meta-analysis of six prospective randomized controlled trials. Dig. Dis. Sci. 2008, 53, 815–822. [Google Scholar] [CrossRef]
  97. Gattoni, A.; Parlato, A.; Vangieri, B.; Bresciani, M.; Derna, R.; Baldassarre, R. Role of hemochromatosis genes in chronic hepatitis C. Clin. Ter. 2006, 157, 61–68. [Google Scholar]
  98. Collaborators, G.B.D.A. Alcohol use and burden for 195 countries and territories, 1990-2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet 2018, 392, 1015–1035. [Google Scholar] [CrossRef] [Green Version]
  99. Global Burden of Disease Liver Cancer Collaboration. The Burden of Primary Liver Cancer and Underlying Etiologies From 1990 to 2015 at the Global, Regional, and National Level: Results From the Global Burden of Disease Study 2015. JAMA Oncol. 2017, 3, 1683–1691. [Google Scholar] [CrossRef] [PubMed]
  100. Makarova-Rusher, O.V.; Altekruse, S.F.; McNeel, T.S.; Ulahannan, S.; Duffy, A.G.; Graubard, B.I.; Greten, T.F.; McGlynn, K.A. Population attributable fractions of risk factors for hepatocellular carcinoma in the United States. Cancer 2016, 122, 1757–1765. [Google Scholar] [CrossRef] [PubMed]
  101. Welzel, T.M.; Graubard, B.I.; Quraishi, S.; Zeuzem, S.; Davila, J.A.; El-Serag, H.B.; McGlynn, K.A. Population-attributable fractions of risk factors for hepatocellular carcinoma in the United States. Am. J. Gastroenterol. 2013, 108, 1314–1321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Hassan, M.M.; Hwang, L.Y.; Hatten, C.J.; Swaim, M.; Li, D.; Abbruzzese, J.L.; Beasley, P.; Patt, Y.Z. Risk factors for hepatocellular carcinoma: Synergism of alcohol with viral hepatitis and diabetes mellitus. Hepatology 2002, 36, 1206–1213. [Google Scholar] [CrossRef]
  103. Grewal, P.; Viswanathen, V.A. Liver cancer and alcohol. Clin. Liver Dis. 2012, 16, 839–850. [Google Scholar] [CrossRef]
  104. Seitz, H.K.; Bataller, R.; Cortez-Pinto, H.; Gao, B.; Gual, A.; Lackner, C.; Mathurin, P.; Mueller, S.; Szabo, G.; Tsukamoto, H. Alcoholic liver disease. Nat. Rev. Dis. Primers 2018, 4, 16. [Google Scholar] [CrossRef]
  105. Ganne-Carrie, N.; Nahon, P. Hepatocellular carcinoma in the setting of alcohol-related liver disease. J. Hepatol. 2019, 70, 284–293. [Google Scholar] [CrossRef] [Green Version]
  106. Parlesak, A.; Schafer, C.; Schutz, T.; Bode, J.C.; Bode, C. Increased intestinal permeability to macromolecules and endotoxemia in patients with chronic alcohol abuse in different stages of alcohol-induced liver disease. J. Hepatol. 2000, 32, 742–747. [Google Scholar] [CrossRef]
  107. Park, E.J.; Lee, J.H.; Yu, G.Y.; He, G.; Ali, S.R.; Holzer, R.G.; Osterreicher, C.H.; Takahashi, H.; Karin, M. Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell 2010, 140, 197–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Niemela, O.; Parkkila, S.; Pasanen, M.; Iimuro, Y.; Bradford, B.; Thurman, R.G. Early alcoholic liver injury: Formation of protein adducts with acetaldehyde and lipid peroxidation products, and expression of CYP2E1 and CYP3A. Alcohol. Clin. Exp. Res. 1998, 22, 2118–2124. [Google Scholar] [CrossRef] [PubMed]
  109. Powell, L.W. The role of alcoholism in hepatic iron storage disease. Ann. N. Y. Acad. Sci. 1975, 252, 124–134. [Google Scholar] [CrossRef]
  110. Nahon, P.; Sutton, A.; Rufat, P.; Ziol, M.; Thabut, G.; Schischmanoff, P.O.; Vidaud, D.; Charnaux, N.; Couvert, P.; Ganne-Carrie, N.; et al. Liver iron, HFE gene mutations, and hepatocellular carcinoma occurrence in patients with cirrhosis. Gastroenterology 2008, 134, 102–110. [Google Scholar] [CrossRef] [Green Version]
  111. Ganne-Carrie, N.; Christidis, C.; Chastang, C.; Ziol, M.; Chapel, F.; Imbert-Bismut, F.; Trinchet, J.C.; Guettier, C.; Beaugrand, M. Liver iron is predictive of death in alcoholic cirrhosis: A multivariate study of 229 consecutive patients with alcoholic and/or hepatitis C virus cirrhosis: A prospective follow up study. Gut 2000, 46, 277–282. [Google Scholar] [CrossRef] [PubMed]
  112. Harrison-Findik, D.D.; Schafer, D.; Klein, E.; Timchenko, N.A.; Kulaksiz, H.; Clemens, D.; Fein, E.; Andriopoulos, B.; Pantopoulos, K.; Gollan, J. Alcohol metabolism-mediated oxidative stress down-regulates hepcidin transcription and leads to increased duodenal iron transporter expression. J. Biol. Chem. 2006, 281, 22974–22982. [Google Scholar] [CrossRef] [Green Version]
  113. Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016, 64, 73–84. [Google Scholar] [CrossRef] [Green Version]
  114. Estes, C.; Razavi, H.; Loomba, R.; Younossi, Z.; Sanyal, A.J. Modeling the epidemic of nonalcoholic fatty liver disease demonstrates an exponential increase in burden of disease. Hepatology 2018, 67, 123–133. [Google Scholar] [CrossRef] [PubMed]
  115. Sorensen, H.T.; Mellemkjaer, L.; Jepsen, P.; Thulstrup, A.M.; Baron, J.; Olsen, J.H.; Vilstrup, H. Risk of cancer in patients hospitalized with fatty liver: A Danish cohort study. J. Clin. Gastroenterol. 2003, 36, 356–359. [Google Scholar] [CrossRef]
  116. Hucke, F.; Sieghart, W.; Schoniger-Hekele, M.; Peck-Radosavljevic, M.; Muller, C. Clinical characteristics of patients with hepatocellular carcinoma in Austria—Is there a need for a structured screening program? Wien. Klin. Wochenschr. 2011, 123, 542–551. [Google Scholar] [CrossRef]
  117. Ertle, J.; Dechene, A.; Sowa, J.P.; Penndorf, V.; Herzer, K.; Kaiser, G.; Schlaak, J.F.; Gerken, G.; Syn, W.K.; Canbay, A. Non-alcoholic fatty liver disease progresses to hepatocellular carcinoma in the absence of apparent cirrhosis. Int. J. Cancer 2011, 128, 2436–2443. [Google Scholar] [CrossRef] [PubMed]
  118. Malik, S.M.; Gupte, P.A.; de Vera, M.E.; Ahmad, J. Liver transplantation in patients with nonalcoholic steatohepatitis-related hepatocellular carcinoma. Clin. Gastroenterol. Hepatol. 2009, 7, 800–806. [Google Scholar] [CrossRef] [PubMed]
  119. Kawada, N.; Imanaka, K.; Kawaguchi, T.; Tamai, C.; Ishihara, R.; Matsunaga, T.; Gotoh, K.; Yamada, T.; Tomita, Y. Hepatocellular carcinoma arising from non-cirrhotic nonalcoholic steatohepatitis. J. Gastroenterol. 2009, 44, 1190–1194. [Google Scholar] [CrossRef]
  120. Tokushige, K.; Hashimoto, E.; Horie, Y.; Taniai, M.; Higuchi, S. Hepatocellular carcinoma in Japanese patients with nonalcoholic fatty liver disease, alcoholic liver disease, and chronic liver disease of unknown etiology: Report of the nationwide survey. J. Gastroenterol. 2011, 46, 1230–1237. [Google Scholar] [CrossRef] [PubMed]
  121. Caldwell, S.H.; Crespo, D.M.; Kang, H.S.; Al-Osaimi, A.M. Obesity and hepatocellular carcinoma. Gastroenterology 2004, 127, S97–S103. [Google Scholar] [CrossRef] [PubMed]
  122. Larsson, S.C.; Wolk, A. Overweight, obesity and risk of liver cancer: A meta-analysis of cohort studies. Br. J. Cancer 2007, 97, 1005–1008. [Google Scholar] [CrossRef]
  123. Neuschwander-Tetri, B.A.; Caldwell, S.H. Nonalcoholic steatohepatitis: Summary of an AASLD Single Topic Conference. Hepatology 2003, 37, 1202–1219. [Google Scholar] [CrossRef]
  124. El-Serag, H.B. Hepatocellular carcinoma. N. Engl. J. Med. 2011, 365, 1118–1127. [Google Scholar] [CrossRef]
  125. Friedman, S.L.; Neuschwander-Tetri, B.A.; Rinella, M.; Sanyal, A.J. Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 2018, 24, 908–922. [Google Scholar] [CrossRef] [PubMed]
  126. Loomba, R.; Sanyal, A.J. The global NAFLD epidemic. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 686–690. [Google Scholar] [CrossRef] [PubMed]
  127. Caligiuri, A.; Gentilini, A.; Marra, F. Molecular Pathogenesis of NASH. Int. J. Mol. Sci. 2016, 17, 1575. [Google Scholar] [CrossRef] [Green Version]
  128. Straub, B.K.; Schirmacher, P. Pathology and biopsy assessment of non-alcoholic fatty liver disease. Dig. Dis. 2010, 28, 197–202. [Google Scholar] [CrossRef] [PubMed]
  129. Masarone, M.; Rosato, V.; Dallio, M.; Gravina, A.G.; Aglitti, A.; Loguercio, C.; Federico, A.; Persico, M. Role of Oxidative Stress in Pathophysiology of Nonalcoholic Fatty Liver Disease. Oxid. Med. Cell Longev. 2018, 2018, 9547613. [Google Scholar] [CrossRef]
  130. Rolo, A.P.; Teodoro, J.S.; Palmeira, C.M. Role of oxidative stress in the pathogenesis of nonalcoholic steatohepatitis. Free Radic. Biol. Med. 2012, 52, 59–69. [Google Scholar] [CrossRef]
  131. Graham, R.M.; Chua, A.C.; Carter, K.W.; Delima, R.D.; Johnstone, D.; Herbison, C.E.; Firth, M.J.; O’Leary, R.; Milward, E.A.; Olynyk, J.K.; et al. Hepatic iron loading in mice increases cholesterol biosynthesis. Hepatology 2010, 52, 462–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Tan, T.C.; Crawford, D.H.; Jaskowski, L.A.; Subramaniam, V.N.; Clouston, A.D.; Crane, D.I.; Bridle, K.R.; Anderson, G.J.; Fletcher, L.M. Excess iron modulates endoplasmic reticulum stress-associated pathways in a mouse model of alcohol and high-fat diet-induced liver injury. Lab. Investig. 2013, 93, 1295–1312. [Google Scholar] [CrossRef] [Green Version]
  133. Vecchi, C.; Montosi, G.; Zhang, K.; Lamberti, I.; Duncan, S.A.; Kaufman, R.J.; Pietrangelo, A. ER stress controls iron metabolism through induction of hepcidin. Science 2009, 325, 877–880. [Google Scholar] [CrossRef] [Green Version]
  134. Matteoni, C.A.; Younossi, Z.M.; Gramlich, T.; Boparai, N.; Liu, Y.C.; McCullough, A.J. Nonalcoholic fatty liver disease: A spectrum of clinical and pathological severity. Gastroenterology 1999, 116, 1413–1419. [Google Scholar] [CrossRef]
  135. Yatsuji, S.; Hashimoto, E.; Tobari, M.; Taniai, M.; Tokushige, K.; Shiratori, K. Clinical features and outcomes of cirrhosis due to non-alcoholic steatohepatitis compared with cirrhosis caused by chronic hepatitis C. J. Gastroenterol. Hepatol. 2009, 24, 248–254. [Google Scholar] [CrossRef]
  136. Kowdley, K.V.; Belt, P.; Wilson, L.A.; Yeh, M.M.; Neuschwander-Tetri, B.A.; Chalasani, N.; Sanyal, A.J.; Nelson, J.E.; Network, N.C.R. Serum ferritin is an independent predictor of histologic severity and advanced fibrosis in patients with nonalcoholic fatty liver disease. Hepatology 2012, 55, 77–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Moon, J.H.; Park, S.H.; Oh, K.C.; Jung, J.O.; Shin, W.G.; Kim, J.P.; Kim, K.O.; Park, C.H.; Hahn, T.; Yoo, K.S.; et al. Association of hepatic iron deposition and serum iron indices with hepatic inflammation and fibrosis stage in nonalcoholic fatty liver disease. Korean J. Gastroenterol. 2006, 47, 432–439. [Google Scholar] [PubMed]
  138. Hagstrom, H.; Nasr, P.; Bottai, M.; Ekstedt, M.; Kechagias, S.; Hultcrantz, R.; Stal, P. Elevated serum ferritin is associated with increased mortality in non-alcoholic fatty liver disease after 16 years of follow-up. Liver Int. 2016, 36, 1688–1695. [Google Scholar] [CrossRef] [PubMed]
  139. Bonkovsky, H.L.; Banner, B.F.; Lambrecht, R.W.; Rubin, R.B. Iron in liver diseases other than hemochromatosis. Semin Liver Dis. 1996, 16, 65–82. [Google Scholar] [CrossRef] [PubMed]
  140. Nelson, J.E.; Wilson, L.; Brunt, E.M.; Yeh, M.M.; Kleiner, D.E.; Unalp-Arida, A.; Kowdley, K.V.; Nonalcoholic Steatohepatitis Clinical Research, N. Relationship between the pattern of hepatic iron deposition and histological severity in nonalcoholic fatty liver disease. Hepatology 2011, 53, 448–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Sorrentino, P.; D’Angelo, S.; Ferbo, U.; Micheli, P.; Bracigliano, A.; Vecchione, R. Liver iron excess in patients with hepatocellular carcinoma developed on non-alcoholic steato-hepatitis. J. Hepatol. 2009, 50, 351–357. [Google Scholar] [CrossRef]
  142. Valenti, L.; Dongiovanni, P.; Fracanzani, A.L.; Santorelli, G.; Fatta, E.; Bertelli, C.; Taioli, E.; Fiorelli, G.; Fargion, S. Increased susceptibility to nonalcoholic fatty liver disease in heterozygotes for the mutation responsible for hereditary hemochromatosis. Dig. Liver Dis. 2003, 35, 172–178. [Google Scholar] [CrossRef]
  143. Valenti, L.; Fracanzani, A.L.; Bugianesi, E.; Dongiovanni, P.; Galmozzi, E.; Vanni, E.; Canavesi, E.; Lattuada, E.; Roviaro, G.; Marchesini, G.; et al. HFE genotype, parenchymal iron accumulation, and liver fibrosis in patients with nonalcoholic fatty liver disease. Gastroenterology 2010, 138, 905–912. [Google Scholar] [CrossRef] [Green Version]
  144. Bonkovsky, H.L.; Jawaid, Q.; Tortorelli, K.; LeClair, P.; Cobb, J.; Lambrecht, R.W.; Banner, B.F. Non-alcoholic steatohepatitis and iron: Increased prevalence of mutations of the HFE gene in non-alcoholic steatohepatitis. J. Hepatol. 1999, 31, 421–429. [Google Scholar] [CrossRef]
  145. Wagner, J.; Fillebeen, C.; Haliotis, T.; Charlebois, E.; Katsarou, A.; Mui, J.; Vali, H.; Pantopoulos, K. Mouse models of hereditary hemochromatosis do not develop early liver fibrosis in response to a high fat diet. PLoS ONE 2019, 14, e0221455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Tsurusaki, S.; Tsuchiya, Y.; Koumura, T.; Nakasone, M.; Sakamoto, T.; Matsuoka, M.; Imai, H.; Yuet-Yin Kok, C.; Okochi, H.; Nakano, H.; et al. Hepatic ferroptosis plays an important role as the trigger for initiating inflammation in nonalcoholic steatohepatitis. Cell Death Dis. 2019, 10, 449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Liu, C.Y.; Feng, G.S. NCOA5, a molecular link between type 2 diabetes and liver cancer. Hepatobiliary Surg. Nutr. 2014, 3, 106–108. [Google Scholar] [CrossRef]
  149. Inoue, M.; Iwasaki, M.; Otani, T.; Sasazuki, S.; Noda, M.; Tsugane, S. Diabetes mellitus and the risk of cancer: Results from a large-scale population-based cohort study in Japan. Arch. Intern. Med. 2006, 166, 1871–1877. [Google Scholar] [CrossRef] [Green Version]
  150. Davila, J.A.; Morgan, R.O.; Shaib, Y.; McGlynn, K.A.; El-Serag, H.B. Diabetes increases the risk of hepatocellular carcinoma in the United States: A population based case control study. Gut 2005, 54, 533–539. [Google Scholar] [CrossRef]
  151. El-Serag, H.B.; Tran, T.; Everhart, J.E. Diabetes increases the risk of chronic liver disease and hepatocellular carcinoma. Gastroenterology 2004, 126, 460–468. [Google Scholar] [CrossRef]
  152. Polesel, J.; Zucchetto, A.; Montella, M.; Dal Maso, L.; Crispo, A.; La Vecchia, C.; Serraino, D.; Franceschi, S.; Talamini, R. The impact of obesity and diabetes mellitus on the risk of hepatocellular carcinoma. Ann. Oncol. 2009, 20, 353–357. [Google Scholar] [CrossRef]
  153. Wideroff, L.; Gridley, G.; Mellemkjaer, L.; Chow, W.H.; Linet, M.; Keehn, S.; Borch-Johnsen, K.; Olsen, J.H. Cancer incidence in a population-based cohort of patients hospitalized with diabetes mellitus in Denmark. J. Natl. Cancer Inst. 1997, 89, 1360–1365. [Google Scholar] [CrossRef]
  154. Noto, H.; Osame, K.; Sasazuki, T.; Noda, M. Substantially increased risk of cancer in patients with diabetes mellitus: A systematic review and meta-analysis of epidemiologic evidence in Japan. J. Diabetes Complicat. 2010, 24, 345–353. [Google Scholar] [CrossRef]
  155. Buysschaert, M.; Paris, I.; Selvais, P.; Hermans, M.P. Clinical aspects of diabetes secondary to idiopathic haemochromatosis in French-speaking Belgium. Diabetes Metab. 1997, 23, 308–313. [Google Scholar]
  156. Moirand, R.; Adams, P.C.; Bicheler, V.; Brissot, P.; Deugnier, Y. Clinical features of genetic hemochromatosis in women compared with men. Ann. Intern. Med. 1997, 127, 105–110. [Google Scholar] [CrossRef] [PubMed]
  157. Hatunic, M.; Finucane, F.M.; Brennan, A.M.; Norris, S.; Pacini, G.; Nolan, J.J. Effect of iron overload on glucose metabolism in patients with hereditary hemochromatosis. Metabolism 2010, 59, 380–384. [Google Scholar] [CrossRef] [PubMed]
  158. McClain, D.A.; Abraham, D.; Rogers, J.; Brady, R.; Gault, P.; Ajioka, R.; Kushner, J.P. High prevalence of abnormal glucose homeostasis secondary to decreased insulin secretion in individuals with hereditary haemochromatosis. Diabetologia 2006, 49, 1661–1669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Baker, K.S.; Ness, K.K.; Steinberger, J.; Carter, A.; Francisco, L.; Burns, L.J.; Sklar, C.; Forman, S.; Weisdorf, D.; Gurney, J.G.; et al. Diabetes, hypertension, and cardiovascular events in survivors of hematopoietic cell transplantation: A report from the bone marrow transplantation survivor study. Blood 2007, 109, 1765–1772. [Google Scholar] [CrossRef] [Green Version]
  160. Borgna-Pignatti, C.; Rugolotto, S.; De Stefano, P.; Zhao, H.; Cappellini, M.D.; Del Vecchio, G.C.; Romeo, M.A.; Forni, G.L.; Gamberini, M.R.; Ghilardi, R.; et al. Survival and complications in patients with thalassemia major treated with transfusion and deferoxamine. Haematologica 2004, 89, 1187–1193. [Google Scholar] [PubMed]
  161. Pappa, A.; Hausler, M.G.; Veigel, A.; Tzamouranis, K.; Pfeifer, M.W.; Schmidt, A.; Bokamp, M.; Haberland, H.; Wagner, S.; Bruckel, J.; et al. Diabetes mellitus in Friedreich Ataxia: A case series of 19 patients from the German-Austrian diabetes mellitus registry. Diabetes Res. Clin. Pract. 2018, 141, 229–236. [Google Scholar] [CrossRef]
  162. Dmochowski, K.; Finegood, D.T.; Francombe, W.; Tyler, B.; Zinman, B. Factors determining glucose tolerance in patients with thalassemia major. J. Clin. Endocrinol. Metab. 1993, 77, 478–483. [Google Scholar] [CrossRef]
  163. Merkel, P.A.; Simonson, D.C.; Amiel, S.A.; Plewe, G.; Sherwin, R.S.; Pearson, H.A.; Tamborlane, W.V. Insulin resistance and hyperinsulinemia in patients with thalassemia major treated by hypertransfusion. N. Engl. J. Med. 1988, 318, 809–814. [Google Scholar] [CrossRef]
  164. Wilson, J.G.; Lindquist, J.H.; Grambow, S.C.; Crook, E.D.; Maher, J.F. Potential role of increased iron stores in diabetes. Am. J. Med. Sci. 2003, 325, 332–339. [Google Scholar] [CrossRef]
  165. Ford, E.S.; Cogswell, M.E. Diabetes and serum ferritin concentration among U.S. adults. Diabetes Care 1999, 22, 1978–1983. [Google Scholar] [CrossRef]
  166. Brissot, P.; Pietrangelo, A.; Adams, P.C.; de Graaff, B.; McLaren, C.E.; Loreal, O. Haemochromatosis. Nat. Rev. Dis. Primers 2018, 4, 18016. [Google Scholar] [CrossRef] [PubMed]
  167. Sandhu, K.; Flintoff, K.; Chatfield, M.D.; Dixon, J.L.; Ramm, L.E.; Ramm, G.A.; Powell, L.W.; Subramaniam, V.N.; Wallace, D.F. Phenotypic analysis of hemochromatosis subtypes reveals variations in severity of iron overload and clinical disease. Blood 2018, 132, 101–110. [Google Scholar] [CrossRef] [PubMed]
  168. Ray, P.D.; Huang, B.W.; Tsuji, Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal. 2012, 24, 981–990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Mittal, M.; Siddiqui, M.R.; Tran, K.; Reddy, S.P.; Malik, A.B. Reactive oxygen species in inflammation and tissue injury. Antioxid. Redox Signal. 2014, 20, 1126–1167. [Google Scholar] [CrossRef] [Green Version]
  170. Gamberi, T.; Magherini, F.; Modesti, A.; Fiaschi, T. Adiponectin Signaling Pathways in Liver Diseases. Biomedicines 2018, 6, 52. [Google Scholar] [CrossRef] [Green Version]
  171. Cooksey, R.C.; Jones, D.; Gabrielsen, S.; Huang, J.; Simcox, J.A.; Luo, B.; Soesanto, Y.; Rienhoff, H.; Abel, E.D.; McClain, D.A. Dietary iron restriction or iron chelation protects from diabetes and loss of beta-cell function in the obese (ob/ob lep−/−) mouse. Am. J. Physiol. Endocrinol. Metab. 2010, 298, E1236–E1243. [Google Scholar] [CrossRef] [Green Version]
  172. Kim, D.K.; Kim, Y.H.; Jung, Y.S.; Kim, K.S.; Jeong, J.H.; Lee, Y.S.; Yuk, J.M.; Oh, B.C.; Choy, H.E.; Dooley, S.; et al. Orphan nuclear receptor SHP regulates iron metabolism through inhibition of BMP6-mediated hepcidin expression. Sci. Rep. 2016, 6, 34630. [Google Scholar] [CrossRef] [Green Version]
  173. Deschemin, J.C.; Foretz, M.; Viollet, B.; Vaulont, S. AMPK is not required for the effect of metformin on the inhibition of BMP6-induced hepcidin gene expression in hepatocytes. Sci. Rep. 2017, 7, 12679. [Google Scholar] [CrossRef]
  174. Kim, Y.D.; Park, K.G.; Lee, Y.S.; Park, Y.Y.; Kim, D.K.; Nedumaran, B.; Jang, W.G.; Cho, W.J.; Ha, J.; Lee, I.K.; et al. Metformin inhibits hepatic gluconeogenesis through AMP-activated protein kinase-dependent regulation of the orphan nuclear receptor SHP. Diabetes 2008, 57, 306–314. [Google Scholar] [CrossRef] [Green Version]
  175. Wang, M.; Xin, H.; Tang, W.; Li, Y.; Zhang, Z.; Fan, L.; Miao, L.; Tan, B.; Wang, X.; Zhu, Y.Z. AMPK Serves as a Therapeutic Target Against Anemia of Inflammation. Antioxid. Redox Signal. 2017, 27, 251–268. [Google Scholar] [CrossRef] [PubMed]
  176. Decensi, A.; Puntoni, M.; Goodwin, P.; Cazzaniga, M.; Gennari, A.; Bonanni, B.; Gandini, S. Metformin and cancer risk in diabetic patients: A systematic review and meta-analysis. Cancer Prev. Res. 2010, 3, 1451–1461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Paganoni, R.; Lechel, A.; Vujic Spasic, M. Iron at the Interface of Hepatocellular Carcinoma. Int. J. Mol. Sci. 2021, 22, 4097. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22084097

AMA Style

Paganoni R, Lechel A, Vujic Spasic M. Iron at the Interface of Hepatocellular Carcinoma. International Journal of Molecular Sciences. 2021; 22(8):4097. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22084097

Chicago/Turabian Style

Paganoni, Rossana, André Lechel, and Maja Vujic Spasic. 2021. "Iron at the Interface of Hepatocellular Carcinoma" International Journal of Molecular Sciences 22, no. 8: 4097. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22084097

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop