You are on page 1of 21

Received: 12 December 2018 | First decision: 8 January 2019 | Accepted: 16 January 2019

DOI: 10.1111/apt.15173

Review article: iron disturbances in chronic liver diseases


other than haemochromatosis – pathogenic, prognostic, and
therapeutic implications

Albert J. Czaja

Division of Gastroenterology and


Hepatology, Mayo Clinic College of Summary
Medicine and Science, Rochester, Background: Disturbances in iron regulation have been described in diverse chronic
Minnesota
liver diseases other than hereditary haemochromatosis, and iron toxicity may wor-
Correspondence sen liver injury and outcome.
Prof. Albert J. Czaja, Division of
Gastroenterology and Hepatology, Mayo Aims: To describe manifestations and consequences of iron dysregulation in chronic
Clinic College of Medicine and Science, liver diseases apart from hereditary haemochromatosis and to encourage investiga-
Rochester, MN.
Email: czaja.albert@mayo.edu tions that clarify pathogenic mechanisms, define risk thresholds for iron toxicity, and
direct management
Funding information
None. Methods: English abstracts were identified in PubMed by multiple search terms.
Full length articles were selected for review, and secondary and tertiary bibliogra-
phies were developed.
Results: Hyperferritinemia is present in 4%‐65% of patients with non‐alcoholic fatty
liver disease, autoimmune hepatitis, chronic viral hepatitis, or alcoholic liver disease,
and hepatic iron content is increased in 11%‐52%. Heterozygosity for the C282Y
mutation is present in 17%‐48%, but this has not uniformly distinguished patients
with adverse outcomes. An inappropriately low serum hepcidin level has charac-
terised most chronic liver diseases with the exception of non‐alcoholic fatty liver
disease, and the finding has been associated mainly with suppression of transcrip-
tional activity of the hepcidin gene. Iron overload has been associated with oxida-
tive stress, advanced fibrosis and decreased survival, and promising therapies
beyond phlebotomy and oral iron chelation have included hepcidin agonists.
Conclusions: Iron dysregulation is common in chronic liver diseases other than
hereditary haemochromatosis, and has been associated with liver toxicity and poor
prognosis. Further evaluation of iron overload as a co‐morbid factor should identify
the key pathogenic disturbances, establish the risk threshold for iron toxicity, and
promote molecular interventions.

The Handling Editor for this article was Dr Colin Howden, and this uncommissioned review
was accepted for publication after full peer‐review.

Aliment Pharmacol Ther. 2019;1–21. wileyonlinelibrary.com/journal/apt © 2019 John Wiley & Sons Ltd | 1
2 | CZAJA

1 | INTRODUCTION 3 | RESULTS

Disturbances in iron regulation have been described in diverse Hyperferritinemia has been the principal manifestation of a disturbed
chronic liver diseases other than hereditary haemochromatosis. 1–5
iron homeostasis in chronic liver disease,1–4 but other findings have
Serum ferritin levels are increased in cohorts with non‐alcoholic included inappropriate serum hepcidin levels,22,24,28–32,47 increased
fatty liver disease (NAFLD), 6–12
chronic alcoholic liver disease serum iron concentrations,11,24 stainable iron in hepatic
(ALD),2,6 chronic hepatitis B,13 chronic hepatitis C,14–19 alpha‐1 tissue,1,11,13,24–27,32,42,48,49 normal or moderately increased transfer-
anti‐trypsin deficiency,20–22 and autoimmune hepatitis.23,24 Stainable rin saturations,11,24 and mutations of the HFE gene.14,18,36 The simi-
iron has been present in 32% of cirrhotic livers of various larity of the findings across a broad spectrum of different chronic
causes,25–27 and serum hepcidin levels that regulate the duodenal liver diseases and resolution of the abnormalities during treatment
absorption and tissue storage of iron have been low in autoimmune suggest that the disturbances are consequences of liver injury.39,43,50
hepatitis,24,28 primary biliary cholangitis (PBC),28 primary sclerosing Alternatively, the association of the iron disturbances with disease
cholangitis (PSC),28 alcohol‐related cirrhosis,29 chronic hepatitis severity51–53 and the recognised hepatotoxicity of iron over-
B‐related cirrhosis, 30
and alpha‐1 anti‐trypsin deficiency. 22
In con- load45,46,54,55 compel consideration of iron as a pathogenic factor.
trast, circulating hepcidin concentrations have been increased in
NAFLD.31,32
3.1 | Normal iron metabolism
Hyerferritinemia has been associated with normal or near normal
serum transferrin saturations (≤45%) in most patients with chronic Iron is derived from dietary sources that contain heme (≤10%)
liver disease and iron dysregulation, and further testing for the point and nonheme (>90%) products.56,57 Intestinal absorption is the pri-
mutations (C282Y and H63D) of the haemochromatosis gene (HFE mary mechanism for acquiring iron, and 1‐2 mg are absorbed daily
[high Fe gene]) has not been warranted. 33,34
In those patients tested by enterocytes located mainly in the duodenum.56,57 Nonheme
for the HFE mutations, the frequency of genetic haemochromatosis iron is ingested in the oxidised ferric (Fe3+) state which renders
25
has been low; and the association of the heterozygous mutations the iron insoluble and biologically inert. Transport across the duo-
with the hyperferritinemia and liver disease has been inconsistent denal epithelium requires reduction of ferric (Fe3+) iron to ferrous
and controversial. 4,11,18,19,35–42
(Fe2+) iron by a ferric reductase in the brush border of the ente-
The disturbances in iron homeostasis in chronic liver disease may rocytes.58 Transportation of the ferrous iron into the enterocyte is
be a collateral manifestation of active inflammation and parenchymal accomplished by the transporter protein, divalent metal transporter
damage3,39,43,44 or a pathological mechanism by which iron toxicity 1 (DMT1), and the bound ferrous iron is delivered to ferroportin
worsens the liver injury and outcome. 45,46
The goals of this review on the basolateral membrane of the enterocyte.59,60 Ferroportin is
are to present the findings in diverse chronic liver diseases that sug- a transmembrane protein located on enterocytes, macrophages,
gest disturbed iron regulation, describe possible mechanisms that hepatocytes and adipocytes, and it is the main transporter of
could account for the manifestations, indicate how the dysregulation intracellular iron to the bloodstream.61–63 Hepcidin inhibits
of iron homeostasis could affect disease severity, and encourage ferroportin activity by promoting its lysosomal degradation (ubiqui-
investigations that clarify pathogenic mechanisms, identify risk tination), and it thereby modulates the intestinal absorption of
thresholds for iron toxicity, and evaluate evolving molecular iron.64–68
interventions. The extracellular ferrous iron is oxidised to ferric iron by an oxi-
dising ferroxidase (hephaestin) that is homologous to ceruloplasmin,
and the inactive ferric iron is bound to transferrin.69 Transferrin
2 | MATERIALS AND METHODS accepts iron from reticuloendothelial cells ingesting senescent ery-
throcytes, parenchymal tissue, and intestinal mucosal cells, and it
English abstracts were identified in PubMed using the primary search transports the ferric iron to the bone marrow for erythropoiesis and
words, “iron and chronic liver disease,” “iron and non‐alcoholic fatty to other sites for storage or synthesis of iron‐containing pro-
liver disease,” “iron and autoimmune hepatitis,” “iron and chronic teins.45,70,71 Transferrin can also modulate hepcidin activity as part
hepatitis C,” “iron and chronic hepatitis B,” and “iron and alcoholic of a complex system that senses plasma iron levels.71
liver disease.” Abstracts judged pertinent to the review were identi- Ferritin is the principal storage protein for iron, and the liver is
fied; key aspects were recorded; and full‐length articles were the principal storage site of ferritin.45 The 24 subunits that comprise
selected from relevant abstracts. A secondary bibliography was ferritin form a sphere containing 4500 atoms of iron that are depos-
developed from the references cited in the selected full‐length arti- ited as hydroxy‐phosphates.45,57,72 The iron is stored and detoxified
cles, and additional PubMed searches were performed to expand the in the ferritin until it is exported back to the circulation by ferro-
concepts developed in these articles. The discovery process was portin in response to signals that reflect iron deficiency.73 Ferritin is
repeated, and a tertiary bibliography was developed after reviewing an acute phase protein that can be increased during inflammation,43
selected articles from the secondary bibliography. Over 1500 whereas transferrin is down‐regulated during inflammation74 and
abstracts and 90 full length articles were reviewed. reduced in advanced liver disease.75,76
CZAJA | 3

Ferritin that has been iron‐overloaded, especially after haemoly- intake, serum ferritin level, and transferrin saturation, and the stron-
sis or blood transfusion, can undergo incomplete lysosomal degrada- gest association has been with the β‐thalassemia trait.85 The overall
tion, and the denatured ferritin can be deposited within cells as findings suggest that hepatic iron accumulation in NAFLD is often
57,77,78
haemosiderin. Haemosiderin provides storage for excess associated with genetic factors and that this association may
iron,79 but its iron stores cannot be mobilised under physiological contribute to hepatic fibrosis.
conditions.80 Transferrin that has been iron‐overloaded can release Importantly, many patients with NAFLD and hyperferritinemia
toxic nontransferrin‐bound iron (NTB1) into the plasma, and this lack an HFE mutation,39,86 and the severity of liver fibrosis42 and the
unbound ferrous iron is passively removed by the liver where it may frequency of serum ferritin levels >1.5‐fold ULN11 have been similar
5,81
accumulate and induce toxicity. There are no regulated mecha- between patients with and without an HFE mutation. Furthermore,
nisms for iron excretion, and the desquamation of intestinal epithelia, the clinical and pathological effects of iron dysregulation in NAFLD
minor bleeding, and losses in sweat, urine, and faeces account for an have been contested,87–89 especially when patients are matched for
iron loss of 1 mg daily.57 age, obesity, diabetes, and aminotransferase levels.88,89 These obser-
The key regulatory molecules that maintain iron homeostasis are vations have suggested that the HFE mutations are not the primary
transferrin receptor 1 (TfR1) and transferrin receptor 2 (TfR2) that basis for iron dysregulation in most patients with NAFLD and that
mediate the uptake of transferrin‐bound iron by the liver, the disturbances in iron homeostasis may not be directly associated
haemochromatosis protein (HFE) that regulates TfR1 activity, hep- with advanced fibrosis, progression to cirrhosis, or liver‐related mor-
cidin that modulates ferroportin activity, and ferroportin that moves tality.87
iron from storage sites within enterocytes, hepatocytes, and macro- Studies in a genetically obese mouse model suggest that iron
phages to the circulation.56,57,73 Genetic and epigenetic factors con- overload associated with dietary supplementation has diverse patho-
trol the expression of these regulatory proteins, and deficiencies in genic effects and a pathological consequence.90 Iron overloaded
gene expression or the diverse molecular regulators that maintain genetically obese mice develop manifestations of hepatic oxidative
critical signalling pathways may disrupt iron homeostasis and account stress, enhanced gene expression of inflammatory cytokines (inter-
for the iron disturbances found in some patients with chronic liver leukin‐6 [IL‐6] and tumour necrosis factor‐alpha [TNF‐α]), and
disease (Figure 1). increased production of markers associated with innate and adaptive
immune cell responses.90 Furthermore, the dietary iron overload
results in hepatocyte injury (ballooning) that has been associated
3.2 | Iron disturbances in NAFLD
with human NASH.90–93 Similar findings have also been described in
Serum ferritin levels have been increased in 58% of patients with a rat model of NASH supplemented with high dietary fat and iron.94
82 37,83
NAFLD and most patients with insulin resistance, and serum
transferrin saturations have been increased in 35%‐58% (Table 1).37,82
3.2.1 | Dysmetabolic iron overload syndrome
Serum ferritin levels greater than 1.5‐fold the upper limit of the nor-
mal range (ULN) have been associated with male gender, increased The dysmetabolic iron overload syndrome (DIOS) describes patients
serum aminotransferase concentrations, stainable iron in liver tissue, with NAFLD who have high serum ferritin levels, normal transferrin
increased NAFLD activity score, advanced hepatic fibrosis, and histo- saturations, and hepatic iron deposition.37,48,84,95 The syndrome is
11
logical features of non‐alcoholic steatohepatitis (NASH). Obesity in typically accompanied by one or more metabolic abnormalities,
association with a serum ferritin level greater than 240 ng/mL has including increased BMI, hypertension, dyslipidemia, insulin resis-
been associated with NASH in 62% of individuals with normal serum tance, and NASH, and it affects more than 30% of patients with
alanine aminotransferase (ALT) concentrations.9 Furthermore, a body NAFLD.48,96,97 The amount and distribution of iron within the liver
mass index (BMI) greater than 28.2 and serum ferritin level greater (parenchymal vs nonparenchymal deposition) have been associated
than 240 ng/mL have identified patients with hepatic fibrosis (sensi- with the frequency of hepatic fibrosis, but results have differed
tivity, 82%; specificity, 79%).9 between studies, possibly because of ethnic and environmental
Homozygosity for the C282Y mutation has been present in 3% differences.48,49,95
36
of patients with NASH, and heterozygosity for this mutation has Semi‐quantitative assessments of hepatic iron deposits have
been associated with hyperferritinemia in 17% of white North Amer- scored iron accumulations in hepatocytes and in sinusoidal and portal
ican patients with NASH36 and 45% of Italian patients.84 Further- locations in Italian patients with DIOS.48 Hepatic fibrosis has corre-
more, patients with NASH and the C282Y mutation have had higher lated with the total amount of hepatic iron deposition (P = 0.01), espe-
serum ALT levels and more hepatic fibrosis than patients without cially in the portal hepatocytes (P < 0.0001).48 In contrast, stainable
36
this mutation. Parenchymal iron deposition and moderate‐severe iron has been present in 34% of North American patients with
hepatic fibrosis have also been associated with heterozygosity for NAFLD, and advanced hepatic fibrosis, portal inflammation, hepato-
C282Y, homozygosity for H63D, and beta‐globin mutations charac- cyte ballooning, and steatohepatitis have been associated mainly with
teristic of the β‐thalassemia trait in 63% of Italian patients with stainable iron in reticuloendothelial cells.49 Despite these differences,
85
biopsy‐proven NAFLD. In the Italian population, the parenchymal each study has indicated that the presence and pattern of hepatic iron
deposition of iron has been independent of age, gender, BMI, alcohol deposition are possible factors in the progression of NAFLD.48,49
4 | CZAJA

Genetic mutations Concurrent variables Iron overload


C282Y HJV SLC40A1 ETOH
H63D HAMP HCV Fe3+ Fe3+
Hepatocyte
TfR2
Fe3+
Ferritin
+ HFE
Fe3+
BMP6 Fe3+
Ferroportin TfR2
Oxidative stress
Hypoxia
SMAD
mRNA mRNA
Hepcidin Hepcidin
HAMP
TMPRSS6
Nucleus sHemojuvelin
HGF
mRNA
STAT3 CREB3L3
Inflammation Metabolic stress
IL-1 Hepcidin Infection
IL-6 Inflammation
Erythropoiesis Nutrient loss
Anemia

F I G U R E 1 Factors contributing to iron dysregulation in chronic liver diseases outside the diagnosis of hereditary haemochromatosis.
Heterozygous genetic mutations of the HFE (High Fe) gene (C282Y and H63D) have been associated with iron overload in non‐alcoholic fatty
liver disease (NAFLD) and chronic hepatitis C, and they may impair transcriptional activity of the HAMP gene and reduce hepcidin production
(blunted arrow). Other genetic mutations, including (haemojuvelin [HJV], hepcidin [HAMP], transferrin receptor 2 [TfR2], and ferroportin
[SLC40A1]), are unstudied in these chronic liver diseases, but the genetic variants could increase iron overload by decreasing hepcidin
production or reducing ferroportin activity (blunted arrows). Concurrent host‐specific factors, especially alcohol consumption (ETOH) and
infection with the hepatitis C virus (HCV), can also suppress transcriptional activity of HAMP (blunted arrow) and increase iron overload. Iron
overload can activate bone morphogenetic proteins (BMPs), especially BMP6, which increase hepcidin production by stimulating HAMP activity
through the SMAD (sons of mothers against decapentaplegic) pathway (sharp arrow). The SMAD pathway is activated by the complex of HFE
protein, TfR2, and ferric iron (Fe3+) on the hepatocyte membrane. This stimulatory effect can be counterbalanced by disease‐associated
factors, including oxidative stress and tissue hypoxia, which can suppress hepcidin production by increasing molecular inhibitors of HAMP,
including transmembrane protease serine 6 (TMPRSS6), soluble haemojuvelin (sHaemojuvelin), and hepatocyte growth factor (HGF) (blunted
arrow). Metabolic stress on the endoplasmic reticulum can activate the transcription factor, CREB3L3 (cyclic adenosine monophosphate
response element binding protein 3‐like 3), which up‐regulates hepcidin production, and the pro‐inflammatory cytokines, interleukin (IL)‐1 and
IL‐6, can also contribute by activating the signal transducer and activator of transcription 3 (STAT3) pathway (sharp arrows). Disease‐related
up‐regulation of erythropoiesis because of an acquired anemia can contribute by dampening hepcidin production and favouring iron overload
(blunted arrow). The net effect on iron stores probably reflects the counterbalance between genetic, disease‐associated, and patient‐related
factors

Hepcidin‐induced ubiquitination of ferroportin would be


3.2.2 | Hepcidin levels in NAFLD
expected to protect against iron overload in NAFLD, but semi‐
The liver is the principal source of hepcidin,98,99 but adipose tissue quantitative histological studies suggest that parenchymal and
and macrophages also contribute.100,101 Serum hepcidin levels have nonparenchymal iron stores can be overloaded in NAFLD.48,49
been increased in patients with NAFLD with and without DIOS, and Hepcidin has been unable to limit iron absorption in patients
the amount of messenger ribonucleic acid (mRNA) for hepcidin in with DIOS following an oral challenge with iron, and this resis-
liver tissue and the level of hepcidin in serum have correlated with tance to hepcidin may contribute to the iron overload.47 A
32
hepatic iron content but not the degree of steatohepatitis. Similar similar study assessing the hepcidin response to oral iron in
findings have been reported in morbidly obese women with and patients with DIOS has suggested that the increased hepcidin
without histological features of NAFLD.31 In patients with severe level is normal but delayed.102 In this instance, a decreased sen-
obesity, the level of mRNA for hepcidin in adipose tissue has corre- sitivity to mild changes in iron load may have resulted in a
lated with indices of inflammation, including IL‐6 and C‐reactive delayed or deficient hepcidin response.103 Future investigations
protein (CRP), but it has not correlated with the presence of dia- must fully characterise the apparent disconnect between the
betes or NASH.100 These findings suggest that the excessive produc- hepcidin level and iron overload in NAFLD and continue to clar-
tion of hepcidin is associated with hepatic iron retention but not the ify the role of hepatic iron retention or excess in progressive
liver disease. hepatic fibrosis.
CZAJA | 5

T A B L E 1 Iron dysregulation in chronic liver diseases other than haemochromatosis


Chronic liver disease Manifestations of iron dysregulation Clinical associations
NAFLD Hyperferritinemia in 58%82 Male11
Increased transferrin saturation in 35%‐58%37,82 Increased serum ALT level11,36
Stainable hepatic iron in 34%‐100%49,84 Increased NAFLD activity score11
Homozygosity for C282Y in 3%36 Advanced hepatic fibrosis9,11,36
Heterozygosity for C282Y in 17%‐45%36,84 Histological features of NASH11,84
Inappropriate high serum hepcidin levels31,32 Insulin resistance37,83
Increased HAMP mRNA in liver tissue31,32 Possible hepcidin resistance47
Autoimmune hepatitis Hyperferritinemia in 65%24 Serum ferritin >2.09 ULN and serum
Increased serum iron level in 58%24 IgG <1.89 ULN associated with complete
High serum transferrin saturation in 48%24 treatment response24
Stainable hepatic iron in 38%24 Hyperferritinemia and hepatic iron load
Inappropriate low serum hepcidin level24,28 resolve during treatment24
Excess HAMP mRNA in liver tissue28 Hepcidin translation defect28
Chronic hepatitis C Hyperferritinemia in 22%‐27%19,107 Increased serum AST, ALT, GGT109
High serum transferrin saturation in 20%19 HCC in transgenic mice115
Hepatic iron deposition in 17%‐42%19,107,108 Severe hepatic steatosis19
HFE mutation in 34%‐48%14,108 Hepcidin deficiency reversible50
Inappropriate low serum hepcidin level124–126 Hepcidin transcription defect50
Decreased hepcidin mRNA in liver50,124,128
Chronic hepatitis B Abnormal iron markers in 41%13 HDV‐induced liver iron overload13
Hepatic iron deposition in 35%13 HDV‐related greater liver fibrosis13
Inappropriately low serum hepcidin level30 Early mortality (3 mo)140
Chronic alcoholic liver disease Hyperferritinemia in 63%40 Advanced liver fibrosis144
High serum transferrin saturation in 29%40 Shortened survival145
Hepatic iron deposition in 22%‐52%40,143 HCC associated with C282Y147
C282Y mutations in 8%‐16%40,142,147 Increased risk of HCC and death168
Inappropriately low serum hepcidin level154 Hepcidin transcription defect154
Decreased hepcidin mRNA in liver154

ALT, alanine aminotransferase; AST, aspartate aminotransferase; GGT, gamma‐glutamyl transferase; HAMP, hepcidin antimicrobial peptide; HCC, hepato-
cellular carcinoma; HDV, hepatitis delta virus; HFE, high Fe gene; IgG, immunoglobulin G; mRNA, messenger ribonucleic acid; NAFLD, non‐alcoholic fatty
liver disease; NASH, non‐alcoholic steatohepatitis; ULN, upper limit of the normal range.

the liver enzyme and IgG abnormalities could reflect increased iron
3.3 | Iron disturbances in autoimmune hepatitis
release from damaged hepatocytes and their remediation after suc-
Serum ferritin levels have been increased in 65% of untreated cessful treatment.43 Ferritin is an acute phase reactant, and its serum
24
patients with autoimmune hepatitis, and serum iron concentrations level does correlate with serum concentrations of CPR and the pro‐
and transferrin saturations have been abnormally increased in 58% inflammatory cytokine, IL‐6.24,100 Alternatively, the hyperferritinemia
24
and 48%, respectively (Table 1). Furthermore, the serum ferritin and increased transferrin saturation in some patients with autoim-
and immunoglobulin G (IgG) levels at presentation have predicted mune hepatitis could reflect inappropriate suppression of hepcidin
the completeness of biochemical response to standard immunosup- and increased intestinal absorption and tissue mobilisation of iron by
pressive therapy.24 Semi‐quantitative iron scores104 have demon- unfettered ferroportin activity.24
strated mild hepatic iron deposition (range, 1‐30 on scale of 0‐60) in
approximately 38% of patients, but hepatic iron content has not cor-
3.3.1 | Hepcidin levels in autoimmune hepatitis
related with the subsequent treatment response.24
A serum ferritin level >2.09‐fold ULN and serum IgG level Serum hepcidin levels are greatly reduced in untreated patients with
<1.89‐fold ULN at baseline have characterised those patients who autoimmune hepatitis (median levels, <5% of normal),24 and unlike
achieved persistent normalisation of serum aminotransferase concen- healthy individuals in whom the serum hepcidin levels parallel the
trations and IgG level after conventional glucocorticoid treatment serum ferritin levels,105 the patients with autoimmune hepatitis and
24
(complete biochemical response). Serum ferritin levels have paral- hyperferritinemia have inappropriately low serum hepcidin concen-
leled serum aminotransferase levels in those treated patients achiev- trations (Table 1).24 The laboratory indices of liver inflammation
ing laboratory resolution, and the hyperferritinemia and hepatic iron (ALT, aspartate aminotransferase [AST], alkaline phosphatase, and
deposition have resolved during treatment.24 CRP) do not correlate with the circulating hepcidin level, and the
The iron disturbances in untreated autoimmune hepatitis and homeostatic relationship between the serum hepcidin concentration
their subsequent resolution with treatment‐induced normalisation of and the standard iron parameters is not restored until induction of
6 | CZAJA

biochemical remission.24 Furthermore, the hepcidin mRNA level in patients with HCV and cirrhosis was not associated with iron
liver tissue has correlated positively with the serum ferritin level but content or the C282Y mutation.116
not with the serum hepcidin level, and the serum hepcidin concen- At least one HFE mutation has been present in 34%‐48% of
tration has remained low in some patients during a 2‐year period of patients with chronic hepatitis C,14,108 and individuals with the
28
treatment. C282Y mutation have had a higher mean hepatic iron concentration
The unexplained inappropriately low serum hepcidin level in and more hepatic fibrosis than patients without a HFE mutation
autoimmune hepatitis could reflect the severity of liver injury and (Table 1).14 Heterozygosity for the C282Y or H63D mutation has
106
impairment of the hepatic production of hepcidin. Persistence of been an independent risk factor for liver fibrosis and cirrhosis,117–119
a low hepcidin concentration despite protracted therapy in some and the frequency of HFE mutations has been higher in patients with
patients with autoimmune hepatitis suggests that the loss of hep- hepatic steatosis than in patients without this finding (P = 0.03).19
cidin production by the liver is irreversible or that other factors sup- Furthermore, serum ferritin levels and the frequency of HFE muta-
28,106
pressing hepcidin production are persistent. Furthermore, the tions have increased with the severity of hepatic steatosis.19 In Italy,
observation that the hepatic content of mRNA for hepcidin does not heterozygosity for the beta‐globin mutations associated with β‐tha-
correlate with the serum hepcidin level suggests that translation of lassemia trait have been associated with hepatic iron accumulation
the mRNA for hepcidin is impaired or that the delivery of hepcidin and fibrosis.120
to the circulation is compromised by post‐transcriptional processes.28 Not all studies have linked HFE mutations with increased hepatic
Future investigations of the iron disturbances in autoimmune hepati- iron accumulation in chronic hepatitis C,108 and the bases for the
tis should explore genetic mutations, epigenetic factors, and other associations between iron disturbances, steatosis, HFE mutations,
mechanisms that could affect transcriptional activity of the hepcidin and advanced hepatic fibrosis remain speculative.41,121 Iron absorp-
gene (HAMP, hepcidin antimicrobial peptide), the translation of mRNA tion may be favoured by heterozygous HFE mutations;63 iron‐in-
into hepcidin, and the delivery of hepcidin to the circulation. duced oxidative stress may promote hepatic steatosis and
fibrosis;115,122 and HCV may contribute by suppressing hepcidin pro-
duction and inducing metabolic abnormalities that promote hepatic
3.4 | Iron disturbances in chronic hepatitis C
steatosis.123
Hyperferritinemia has been described in 4%‐27% of patients with
chronic hepatitis C (Table 1).16,19,107 Transferrin saturation has been
3.4.1 | Hepcidin levels in chronic hepatitis C
abnormally increased in 20%,19 and hepatic iron deposition has been
present in 11%‐42%.16,19,107,108 Of 14 462 participants in a national Most studies have described low serum hepcidin levels in patients
health survey in the United States, individuals with chronic hepatitis with chronic hepatitis C, and the hepcidin response has been inap-
C have had higher mean serum levels of ferritin and iron than partic- propriate for the associated findings of elevated serum iron concen-
ipants without liver disease, and the serum levels of ferritin have tration, hyperferritinemia, and increased transferrin saturation.124–127
correlated with serum AST, ALT and γ‐glutamyl transpeptidase (GGT) The mRNA for hepcidin has been decreased in the liver tissue of
109
levels (P < 0.0001). Furthermore, 52%‐69% of patients with these patients,124–126,128 and suppression of the transcriptional activ-
chronic hepatitis C have had macrovesicular steatosis (mainly grade ity of the HAMP gene has been proposed as a basis for the iron
1 steatosis) (Table 1).19,110 The steatosis may have constituted overload.125,126,129 The defect in hepcidin production has been
a cytopathic effect associated with the hepatitis C virus (HCV), reversible with antiviral therapy and eradication of HCV.50
especially genotype 3,111–113 or it may have been part of an HCV‐ Reactive oxygen species (ROS) induced by HCV proteins have
independent, dysmetabolic syndrome associated with obesity.114 suppressed the expression of hepcidin in transgenic mice by inhibit-
Hepatic iron deposition in patients with chronic hepatitis C has ing the binding of a transcription factor to the hepcidin promoter.130
been mainly in hepatocytes, and hepatic iron accumulation in the In hepatoma cells, inhibition of gene transcription has been attribu-
sinusoidal and portal compartments has been variously associated ted to HCV‐induced ROS and to epigenetic changes associated with
with increased serum ferritin levels, histological severity, hepatic increased histone deacetylase activity.131 The hypo‐acetylation of
14–
fibrosis, cirrhosis, HFE mutations, and alcohol consumption. histones within the nuclear chromatin has inhibited the binding of
17,19,107,108
The distribution of hepatic iron, especially within the transcription regulators to the hepcidin promoter, and this inhibition
sinusoids, has also been associated with metabolic parameters, has been reversed by anti‐oxidants and by inhibitors of histone
including BMI, insulin resistance, serum low density lipoprotein (LDL) deacetylase activity.131 Oxidative damage of hepatic deoxyribonu-
19
cholesterol levels, and hepatic steatosis. cleic acid (DNA) has correlated strongly with serum ferritin levels
The association of hepatocellular carcinoma (HCC) with iron and total hepatic iron content,129 and the oxidative stress induced
overload in chronic hepatitis C remains uncertain. In male transgenic directly by HCV could result in an iron overload that in turn sustains
mice expressing the HCV polyprotein, hepatic tumours, including or enhances the liver damage.
HCC, developed in 45% of animals fed a diet enriched in iron, Hepcidin can also influence HCV replication. Hepcidin has had a
whereas none of the transgenic mice fed a normal diet manifested direct suppressive effect on HCV replication in cell culture in associ-
these changes.115 In contrast, the occurrence of HCC in 45% of 139 ation with activation of the STAT3 (signal transducer and activator
CZAJA | 7

of transcription protein 3) signalling pathway.132 It has also had an hepcidin‐to‐ferritin ratio has been significantly lower in patients with
indirect effect on HCV replication by modulating the intracellular chronic hepatitis C than in chronic hepatitis B.124 Iron overload by
iron content.133–135 Iron can inactivate HCV RNA polymerase brain imaging has been demonstrated in patients with chronic hep-
134
(NS5B) and inhibit HCV replication, and hepatic iron accumula- atitis B and cirrhosis, and the hepcidin levels have been inappropri-
tion may prevent progression of chronic hepatitis C.135 These direct ately low.30 Furthermore, hyperferritinemia has predicted 3‐month
and indirect antiviral actions of hepcidin suggest a negative feedback mortality in patients with acute‐on‐chronic liver failure associated
mechanism by which HCV replication can be modulated. with hepatitis B virus (HBV) infection.140 The findings suggest that
Hepcidin production and its antiviral action may be reduced by iron disturbances in patients with chronic hepatitis B have the same
HCV. Low serum hepcidin levels may in turn increase duodenal pattern but less severity than in chronic hepatitis C. Future studies
absorption of iron by up‐regulating duodenal ferroportin,136,137 and are needed to determine if these similarities and differences are
the increased intracellular iron content may suppress HCV replica- attributable to a common pathogenic pathway that is modulated by
134,135,138
tion in a negative feedback loop. The net effect of these virus‐specific properties and if the iron disturbances reflect or
counterbalancing actions may depend on the relative strength of the contribute to the disease severity.
inherent response to raise hepcidin levels in response to iron over-
load and the prowess of HCV to circumvent the antiviral action of
3.6 | Iron disturbances in alcoholic liver disease
hepcidin by suppressing transcriptional activity of the HAMP
gene.126 Serum ferritin levels and transferrin saturations are increased in 63%
Importantly, the effects of hepcidin on HCV replication and hep- and 29% of patients with alcoholic liver disease (ALD),40,141 and the
atic iron accumulation may vary at different stages of chronic hepati- results are significantly higher than in healthy individuals (P < 0.05)
tis C. Early in the disease, the antiviral action of hepcidin may (Table 1).142 Stainable hepatic iron in parenchymal and reticuloen-
125
directly suppress HCV replication. Later in the disease, the persis- dothelial cells has been demonstrated in 52% of patients with
tence of HCV infection may suppress hepcidin production and ALD,40 and severe (grade 3‐4) iron deposition has been present in
increase hepatic iron deposition. The iron overload may then gener- 22% of liver explants.143 Stainable hepatic iron has been one of sev-
ate an increase in hepcidin expression and counterbalance the viral eral factors associated with liver fibrosis in ALD,144 and the hepatic
125
effect. The hepcidin response to the hepatic iron concentration iron concentration has been predictive of shortened survival in alco-
has been shown to be appropriate in chronic hepatitis C,139 and holic cirrhosis.145
patients with chronic hepatitis C who achieve a sustained virological A HFE mutation has been detected more frequently in patients
response (SVR) to anti‐viral therapy correct their hepcidin defi- with ALD and hepatic iron accumulation than in patients with ALD
ciency.50 and no hepatic iron accumulation (odds ratio, 17.23; 95% CI: 2.09‐
Future investigations should evaluate serum ferritin and hepcidin 142.34; P = 0.008), but the HFE mutation has not been associated
concentrations as prognostic biomarkers in different stages of with increased hepatic fibrosis, disease severity, or inflammatory
chronic hepatitis C and determine the value of these biomarkers in activity.142 Other studies have demonstrated that the occurrence of
guiding direct‐acting antiviral interventions to protect against pro- a single HFE mutation in ALD has not increased the hepatic iron
gressive hepatic fibrosis and risk of HCC. content, frequency of fibrosis, or predisposition for severe
ALD.35,40,146 Heterozygosity for the C282Y mutation has been more
common in patients with alcoholic cirrhosis and HCC than in
3.5 | Iron disturbances in chronic hepatitis B
patients without HCC (21% vs 4%, P = 0.002),147 but the basis for
Serological evidence of iron dysregulation has been demonstrated in this association remains speculative as other contributing factors,
41% of patients with chronic hepatitis B, and hepatic iron deposits including active alcohol consumption, serum and liver hepcidin levels,
have been recognised in 35% (Table 1).13 Hepatic iron accumulation and hepatic iron concentration, have not been fully analysed.
has been mild (grade 1 deposits in 60%), and they have been associ- Alcohol down‐regulates the transcriptional activity of the hep-
ated with male gender and increased serum levels of ferritin, GGT, cidin gene by decreasing the DNA binding of CCAAT/enhancer‐
and alkaline phosphatase.13 A normal serum ferritin level has had a binding protein α (C/EBPα).148,149 C/EBPα is a transcription factor
negative predictive value of 90% for excluding mild‐moderate hep- that interacts with regulatory sequences in the promoter and
atic iron accumulation, and the major factor associated with higher enhancer regions of diverse genes to promote transcriptional activ-
grades of hepatic iron deposition and more advanced stages of hep- ity,150 and its effect can be diminished by pro‐inflammatory cytoki-
13
atic fibrosis has been co‐infection with hepatitis D virus (HDV). nes (interferon‐γ, IL‐1, IL‐6, TNF‐α),150–152 bacterial
152,153
These findings have supported speculation that the iron dysregula- lipopolysaccharides, and certain toxins, including alco-
tion and hepatic iron deposition in chronic hepatitis B are manifesta- hol.148,150 The hepatic content of mRNA for hepcidin is diminished
tions of active inflammation or advanced stage disease. in ALD,154 and serum hepcidin levels are inappropriately low in
Comparisons between chronic hepatitis C and chronic hepatitis B patients with manifestations of iron overload.154,155 The duodenal
have indicated more hepatic iron deposition and lower hepatic hep- iron transporters are up‐regulated,148,156 and the duodenal absorp-
128
cidin mRNA levels in patients with chronic hepatitis C, and the tion of iron is increased.149,155
8 | CZAJA

The deposition of iron within the liver has been proposed as an transferrin receptor 2 (TfR2), and ferroportin (SLC40A1).34,63,173 Most
145
independent risk factor for survival in ALD (P = 0.007), and the individuals homozygous for C282Y (as many as 99%) do not develop
hepatic accumulation of ferrous iron may generate reactive oxygen haemochromatosis,169,174 and the occurrence of haemochromatosis
species (ROS) that promote progressive tissue injury, liver fibrosis, in individuals homozygous for C282Y probably relates to genetic
and malignant transformation.157–159 In a murine model of combined modifiers or environmental factors.63,175
liver injury by iron overload, alcohol ingestion, and high fat diet, the Hereditary haemochromatosis has been excluded in most
iron overload induced an unfolded protein response within the liver patients with chronic liver disease and iron overload, but heterozy-
and protein changes indicative of stress within the endoplasmic gosity for the HFE mutations has been present in some experiences.
reticulum that were accompanied by impaired degradation and recy- The impact of simple heterozygosity on iron accumulation in patients
cling of cellular components (autophagy).160 The findings suggested with chronic liver disease is unclear. Simple heterozygosity for the
that the iron toxicity increased hepatic stress by impairing adaptive HFE mutations in healthy individuals has not been associated with
compensatory mechanisms. iron excess,176 but undefined factors associated with the liver dis-
Alcohol is also a strong promoter of tissue hypoxia which in turn ease (aetiology, severity, duration) or individual (genetic modifiers,
can increase oxidative stress within the liver.159,161,162 Alcohol can alcohol abuse, nutritional status) may be critical for the clinical
reduce oxygen tensions in the liver possibly by increasing hepatic expression of a heterozygous predisposition. Studies that suggest an
159,161,162
metabolic activity and altering hepatic blood flow. Hypoxia association between simple heterozygosity and hyperferritinemia in
markedly reduces the liver expression of HAMP and inhibits the tran- chronic liver disease36,38,42,117,118 are counterbalanced by studies
163–165
scription of hepcidin. Alcohol can also enhance the production that have found no association.11,42 Disease‐related or patient‐speci-
of ROS by inducing the activity of cytochrome P450 2E1 fic factors may account for these discrepant findings, and future
(CYP2E1)159,166 and by impairing the anti‐oxidant response of investigations must continue to define these risk factors in well‐
167
mitochondria. characterised patient populations.
The risks of HCC (hazard ratio, 1.76; 95% CI: 1.01‐3.06;
P = 0.03) and death (hazard ratio, 1.63; 95% CI: 1.07‐2.44; P = 0.02)
3.7.2 | Disruptions in hepcidin transcription
have been greater in patients with alcoholic cirrhosis and low circu-
lating levels of hepcidin.168 The lack of a strong correlation between Hepcidin production by the liver can be affected by factors that
serum hepcidin levels and hepatic iron accumulation suggests that affect the transcription of the hepcidin gene (HAMP) and the transla-
alcohol consumption (resulting in low serum hepcidin levels) rather tion of the hepcidin mRNA. Transcription of the hepcidin gene is
than iron overload is the major pathogenic factor. regulated by the serum iron concentration which is sensed by two
Hepatic iron accumulation and chronic alcohol consumption are transferrin receptors (TfR1 and TfR2) and the haemochromatosis
each potential risk factors for progressive liver damage, HCC, and protein (HFE) (Figure 1).177 Holotransferrin (diferric transferrin) dis-
diminished survival in ALD mainly through pathways of oxidative places HFE from TfR1 as its concentration increases, and the dis-
stress. Future studies must continue to evaluate distinctions and placed HFE can interact with TfR2 creating a complex that activates
synergisms between these factors. pathways involving bone morphogenetic protein (BMP) and mitogen
activated protein kinase (MAPK).178–180
The binding of BMPs to BMP receptors activates the SMAD
3.7 | Mechanisms of disrupted iron homeostasis in
(Sons of Mothers Against Decapentaplegic) pathway within the cyto-
chronic liver disease
sol of hepatocytes (Figure 1).177,181–183 The hepcidin gene (HAMP) is
Genetic and molecular factors are involved in maintaining total body up‐regulated, and hepcidin production is increased.184,185 BMP6 is
iron content, and one or more of these factors has been implicated an iron‐specific ligand whose expression reflects the iron content of
in the iron disturbances found in some patients with NAFLD, hepatocytes, and it is the principal BMP that induces hepcidin pro-
autoimmune hepatitis, chronic hepatitis B, chronic hepatitis C, and duction.186,187 Haemojuvelin is a co‐receptor for BMP, and it can
ALD. enhance the hepcidin response by binding to BMP2, BMP4, and
BMP9.184,188–190
Hepcidin is an acute‐phase reactant with antimicrobial properties,
3.7.1 | Genetic mutations
and pre‐treatment with hepcidin has protected mice from lethal
C282Y is the principal point mutation of the HFE gene associated doses of lipopolysaccharides.191 The regulation of hepcidin produc-
with hereditary haemochromatosis, and 90% of patients with heredi- tion by multiple intra‐ and extra‐cellular stress signals (iron stores,
tary haemochromatosis are homozygous for C282Y (Figure 1).169 inflammation, hypoxia, and erythropoiesis) may help modulate acute
H63D and S65C are also polymorphisms of HFE, but they have not inflammatory responses (Figure 1).192
been associated with iron overload unless heterozygous with C282Y Inflammation and infection promote hepcidin production by
(compound heterozygosity).63,170–172 Other point mutations that can increasing the secretion of IL‐1 and IL‐6193 and activating the pro-
cause iron overload are outside the HFE gene, and they are muta- moter region within the signal transducer and activator of transcrip-
tions of the genes for haemojuvelin (HJV), hepcidin (HAMP), tion 3 (STAT3) gene.194–197 CREB3L3 (cyclic adenosine
CZAJA | 9

monophosphate response element binding protein 3‐like 3) is a Maintenance of the homeostatic axis regulating hepcidin produc-
stress‐associated transcription factor in the endoplasmic reticulum of tion is a pivotal management objective, and the salutary effects may
cells that can also up‐regulate hepcidin production in response to include prevention of hepatic fibrosis and protection from hepatic
198,199 200
inflammation or metabolic stress. The CREB3L3 transcrip- steatosis. Hepcidin has protected against hepatic fibrosis in murine
tion factor has been associated with hepcidin production and iron models of toxic liver disease and bile duct ligation by suppressing
retention in chronic inflammation and nutrient deprivation (diabetes, phosphorylation of the SMAD3 pathway,212 and it has protected
insulin resistance, starvation) (Figure 1).37,201 mice from diet‐induced obesity and hepatic steatosis by promoting
Key factors that can inhibit hepcidin production are matriptase‐2, glucose tolerance, insulin sensitivity, lipolysis, and weight reduc-
soluble haemojuvelin, missense mutations of various genes involved tion.213 Future investigations in chronic liver disease must continue
in hepcidin transcription and activity, and hepatocyte growth factor to clarify the nature of the interactions between genetic predisposi-
(HGF) (Figure 1). Matriptase‐2, also named transmembrane protease tions, the diverse factors for iron overload (viruses, oxidative stress,
serine 6 (TMPRSS6), is a membrane protease whose overexpression alcohol consumption, and toxins), and the deleterious effects of
can cleave haemojuvelin, impair BMP binding, and suppress activa- hepcidin deficiency.
tion of the HAMP promoter.189,202 Soluble haemojuvelin is the extra-
cellular component of membrane haemojuvelin, and it can bind BMP,
3.7.3 | Disruption in the translation and post‐
competitively antagonise membrane haemojuvelin, and suppress
transcriptional processing of hepcidin mRNA
expression of hepcidin mRNA.185,203 Mutations in the HJV gene can
contribute by reducing the transcription of haemojuvelin, and muta- Disruption in the translation of hepcidin mRNA into hepcidin or
tions in the transferrin receptor‐2 gene, ferroportin gene, and cerulo- delivery of hepcidin to the circulation is suggested by hepatic con-
plasmin gene could alter the sensing of iron levels, efflux of iron from centrations of mRNA for hepcidin that exceed the serum level of
4,173,204,205
cells, and iron absorption. functional hepcidin (Figure 1).28 This disassociation has been
Hepatocyte growth factor (HGF) can inhibit the transcription of described in autoimmune hepatitis, PBC, and PSC,28 and the finding
206
hepcidin by countering the stimulatory effects of BMP6 and IL‐6, may be a feature of iron overload in immune‐mediated liver disease.
and HGF may be a factor in reducing serum hepcidin concentrations The pathogenic mechanisms that disrupt the translation, trafficking,
24
in autoimmune hepatitis (Figure 1). HGF is a plasminogen that is binding, and delivery of hepcidin in certain chronic liver diseases
secreted mainly by mesenchymal cells,207–210 and it may protect with iron overload are unknown, and the clarification of these mech-
against inflammatory and immune‐mediated responses by inhibiting anisms and determination of their disease specificity might improve
activation of the nuclear factor kappa‐light‐chain enhancer of acti- diagnostic and therapeutic algorithms.
vated B cells (NF‐κB), reducing the production of pro‐inflammatory
cytokines, increasing the secretion of IL‐10, and inducing the prolif-
3.7.4 | Liver inflammation and damage
eration of regulatory T cells (Tregs).210,211 Increased expression of
HGF could be a mechanism by which hepcidin production is The laboratory and histological manifestations of iron overload in
inhibited, iron absorption is increased, and hepatic iron toxicity chronic liver disease could be consequences of the liver injury. The
occurs.206 HGF may be a key modulatory factor altered by disease‐ intensity of inflammatory activity (resulting in hepatocyte and macro-
related elements (cytokines, oxidative stress, hypoxia), and its role in phage release of stored iron), the severity of liver injury (resulting in
the suppression of hepcidin production in chronic liver diseases impaired hepatic production of hepcidin), and the presence of ane-
24
must be clarified. mia and tissue hypoxia (resulting in ineffective erythropoiesis, sup-
Disruption in the transcription of hepcidin has been suggested pression of hepcidin production, and enhanced duodenal absorption
as a mechanism for low serum hepcidin levels and iron overload of iron) could erroneously suggest iron overload as a pathogenic fac-
in chronic hepatitis C. Low levels of mRNA for hepcidin in liver tor.3,39,43,159,163
tissue have justified this consideration,128 and ROS and epige- Multiple clinical observations support the possibility that the
netic factors (increased histone deacetylase activity) in association iron disturbances in chronic liver disease are collateral manifesta-
with the HCV infection may suppress the transcriptional activity tions of liver injury. The pattern and nature of the iron abnormali-
of the HAMP gene.124–126,131 The hepatic content of mRNA for ties lack disease‐specificity,1,4,204,214 and serum ferritin levels have
hepcidin is also reduced in patients with ALD, and suppression correlated with markers of liver inflammation (serum AST, ALT
of the transcriptional activity of the hepcidin gene by alcohol, and GGT levels) in autoimmune hepatitis and chronic hepatitis
the reactive products of oxidative stress, and hypoxia within the C.24,109 The serum ferritin level can parallel the circulating level of
148,149,159
hepatic microenvironment have been implicated. Genetic CRP,24 daily alcohol consumption,142,215 and severity of liver injury
predispositions outside the HFE mutations may also influence the in the absence of iron overload.44 The iron abnormalities can
mechanisms by which the serum iron level is sensed and moni- rapidly resolve with improvement of the liver disease24,50 or with-
tored in certain liver diseases, and they may contribute to fail- drawal from alcohol,6 and the manifestations of iron overload
ures in the proper physiological response of the HAMP gene to (HFE mutations, serum ferritin level, hepatic iron deposition, and
iron excess. serum hepcidin concentrations) and features of liver injury
10 | CZAJA

(steatosis, fibrosis, HCC, and survival) have lacked a consistent it is the ferrous iron that reacts with hydrogen peroxide and induces
relationship. the formation of reactive hydroxyl radicals (Table 2).54,55,226,227 The
Future investigations must validate that iron overload in chronic generation of ROS (superoxide, hydrogen peroxide, hydroxyl radicals,
liver diseases outside of hereditary haemochromatosis has a patho- and singlet oxygen) can damage cell membranes, affect gene expres-
genic effect that warrants clinical assessment and management. Cur- sion, impair mitochondrial function, induce hepatocyte injury, and
rent surrogate markers of iron overload can overestimate the hepatic promote hepatic fibrosis.158 Oxidative stress has been implicated in
iron content and misrepresent its pathogenic significance.44 Hepatic the pathogenesis of NAFLD,228–230 ALD,231,232 autoimmune hepati-
iron deposition must supersede the surrogate markers as the pivotal tis,233 and chronic hepatitis C,234,235 and the iron overload that can
finding in future studies, and it should be determined uniformly in all occur in each of these diseases could be a deleterious factor that
study participants by tissue staining,216,217 direct measure- warrants assessment and management.
ment,218,219 magnetic resonance imaging (MRI),220–222 or dual‐energy
computerised tomography (CT).223 The hepatic iron content can then
3.8.2 | Ferritin as a cytoprotective agent and
be correlated with clinical features and outcome irrespective of the
pathogenic factor
surrogate markers. Thresholds of clinical relevance can be deter-
mined, diagnostic algorithms refined, and management options con- Ferritin is an acute phase reactant whose production is stimulated
sidered. by the pro‐inflammatory cytokines, TNF‐α236–238 and IL‐1β.238
Magnetic susceptometry is an emerging non‐invasive mechanism Oxidative stress increases the synthesis of ferritin,239–244 and the
by which to assess hepatic iron concentration, and preliminary stud- NFκB pathway can increase cytokine production and up‐regulate the
ies with a room temperature susceptometer have demonstrated a expression of ferritin (Table 2).240,244 Under these circumstances,
highly linear correlation (r2 = 0.998) between its signal and hepatic hyperferritinemia could be a consequence of liver damage and oxida-
224
iron concentration. The performance parameters of the suscep- tive stress in the absence of hepatic iron overload.39 Ferritin also
tometer for detection of hepatic iron overload (grade ≥2) have been detoxifies iron,45 and it may modulate inflammatory and immune
equal to those of atomic absorption spectroscopy and superior to all responses by suppressing the proliferation and differentiation of pro-
serum iron markers. Furthermore, iron removal by phlebotomy could genitor cells within the bone marrow.245–247 Under these circum-
224
be accurately monitored. stances, hyperferritinemia could have a protective role in mitigating
Magnetic susceptibility measures the attraction or repulsion of a iron‐induced tissue injury.
given material by a magnetic field, and it originally required a highly The ferritin molecule consists of heavy or heart (H) and light or
complex and expensive device based on superconductor technology liver (L) subunits,57 and the H subunit has ferroxidase activity that can
and liquid helium for cooling. Magnetic susceptibility can now be oxidise ferrous iron to ferric iron and protect mitochondria from oxida-
determined at the bedside by a helium‐free, room temperature sus- tive damage.72,73,248–251 The hyperferritinemia in some patients with
224
ceptometer that could be cost‐effective compared to MRI. Room chronic liver disease may constitute an anti‐oxidant response that
temperature susceptometry at the bedside has the potential to facili- attenuates oxidative stress (Table 2).45,72,252–255 The H subunit of fer-
tate future investigations of the pathogenic role of iron overload and ritin can also modulate the response to apoptotic stimuli,255 and the
its removal in the chronic liver diseases that are not hereditary over‐expression of the H subunit in rat livers protects against ische-
haemochromatosis. mia‐reperfusion injury by inhibiting endothelial cell and hepatocyte
apoptosis.256 Furthermore, ferritin activates the promoters of genes
that transcribe proteins regulating haematopoiesis in cell lines, and this
3.8 | Pathogenic consequences of iron overload
direct up‐regulation of haemoglobin production could reduce the gen-
Most iron is tightly complexed with proteins that prevent its toxicity eration of ferrous iron.257 Acidic isoferritins also suppress the prolifer-
45
by rendering it inactive and insoluble (Table 2). Iron resides mainly ation of granulocytes and macrophages in normal individuals,245 and
in haemoglobin (67%), liver, spleen, and bone marrow (15%), iron‐ this myelosuppressive action in a murine model246,247 suggests that
containing cytochromes and enzymes (10%), and muscle myoglobin hyperferritinemia might reduce the inflammatory and immune
(8%).45,225 The conversion of stored iron from its bound, inactive, fer- responses that contribute to certain forms of tissue injury.
ric state to its unbound, reactive, ferrous state can trigger oxidative Hyperferritinemia is also an important prognostic marker that has
stress,45,54 and it is this concern that has generated interest in evaluat- been associated with dire outcomes in advanced chronic liver dis-
ing and treating iron dysregulation in the chronic liver diseases that ease,51–53,258,259 and it may have a nefarious role in some forms of
are outside the diagnosis of hereditary haemochromatosis. liver disease that counters its anti‐oxidant and cytoprotective
actions. Ferritin can promote Fas‐mediated apoptosis,260 activate
hepatic stellate cells (HSCs),261 increase hepatic fibrosis,11,261 and
3.8.1 | Iron overload and oxidative stress
behave as a pro‐inflammatory cytokine (Table 2).261 The association
The release of iron from its binding complex and its conversion from of hyperferritinemia with low serum hepcidin levels and poor treat-
inactive ferric iron to reactive ferrous iron is triggered by iron over- ment response in autoimmune hepatitis,24 increased early mortality
226,227 54
load. The unbound ferric iron is reduced to ferrous iron, and in decompensated cirrhosis,51–53 shortened survival in acute‐on‐
CZAJA | 11

T A B L E 2 Mechanisms of iron toxicity in chronic liver diseases with iron overload


Mechanism Pathogenic actions Pathological consequences
Oxidative stress Generates unbound reactive ferrous iron54 Disrupts cell membranes158
Ferrous iron reacts with hydrogen peroxide54 Affects gene expression158
Superoxide, hydroxyl radicals develop158 Impairs mitochondrial function158
NFκB and cytokines stimulated240 Induces hepatocyte loss158
Ferritin synthesis increased239,242,243 Promotes hepatic fibrosis158
Hyperferritinemia Responds as acute phase reactant43 Promotes Fas‐mediated apoptosis260
(detrimental effects) Increased by inflammation, TNF‐α, IL‐1β238 Activates hepatic stellate cells261
Fails to retain inactive ferric iron226,227 Increases hepatic fibrosis11,261
Allows transformation to active ferrous iron54 Behaves as pro‐inflammatory cytokine261
Promotes generation of ROS54,227 Associated with poor outcome51–53,258,259
Hyperferritinemia Detoxifies and stores iron in cells45 Reduces oxidative stress242,243
(beneficial effects) Ferroxidase on heavy chain of ferritin248,249,251 Modulates inflammatory responses245–247
Transforms ferrous to ferric iron254 Mitigates iron‐induced tissue injury256
Anti‐oxidant effect242,243 Anti‐apoptotic in ischemia‐reperfusion256
Activates genes for erythropoiesis257
Suppresses granulocytes and macrophages246

IL‐1β, interleukin‐1 beta; NFκB, nuclear factor kappa‐light‐chain enhancer of activated B cells; ROS, reactive oxygen species; TNF‐α, transforming growth
factor‐alpha.

chronic liver failure,140 adverse pre‐ and post‐liver transplant out- iron chelation therapy has been a substitute for phlebotomy in indi-
comes,259,262 and advanced fibrosis in NAFLD9,11 bespeak a patho- viduals intolerant of the procedure.34,274,275 Iron removal by phle-
genic role that requires clarification. Future investigations that clarify botomy has decreased the surrogate markers of oxidative stress
the actions of ferritin outside its roles as an intracellular iron storage (modified serum proteins) and reduced the serum level of the pro‐
protein and iron detoxifier may elucidate pathogenic mechanisms fibrotic cytokine, transforming growth factor‐beta 1 (TGF‐β1).276
that can predict outcome and be favourably manipulated. Phlebotomy has also been performed in patients with iron overload
and NAFLD263–265 or chronic hepatitis C.266–268

3.9 | Management strategies


3.10.1 | Phlebotomy in NAFLD
Management of chronic liver diseases with iron dysregulation out-
side of hereditary haemochromatosis is challenging. The pathogenic Phlebotomy has improved fasting and glucose‐stimulated plasma
bases for the iron excess are still being evaluated; the threshold cri- insulin levels by 40%‐55% and decreased serum ALT levels in 17
teria for instituting treatment are incompletely defined; and the ideal patients with NAFLD (Table 3).263 Phlebotomy has reduced insulin
method for improving the iron disturbance is still under study. Inter- resistance in 64 patients with NAFLD and hyperferritinemia com-
ventions can be directed at reducing the iron excess by restricting pared to patients managed by lifestyle modifications,264 and it has
84
iron consumption (dietary measures), removing iron (phlebotomy normalised serum ALT levels in patients maintained on lifestyle mod-
or oral iron chelation),263–270 correcting the underlying mechanisms ifications.277 The NAFLD activity score and individual histological
for the iron overload (low serum hepcidin levels),99,103 or treating features (lobular inflammation, steatosis, and hepatocyte ballooning)
the underlying liver disease (NAFLD, ALD, autoimmune hepatitis, have improved during a phase II clinical trial of phlebotomy in
chronic viral hepatitis) and associated risk factors (alcohol consump- NAFLD, but the size of the positive effect has been insufficient to
tion, obesity, metabolic syndrome, diabetes) (Table 3).44 justify a phase III clinical trial.265 Iron depletion by deferoxamine in a
The appropriate approach depends in part on the nature and hepatoma cell line and murine model of fatty liver has increased the
severity of the iron disturbance. A serum ferritin level >1000 ng/mL activity of the insulin receptor and up‐regulated the glucose trans-
has been proposed as a threshold indication for iron reduction ther- porter, but oral iron chelation therapy has not been evaluated in
apy as this level has been associated with reduced survival.258,271 patients with NAFLD.269
Confirmation of hepatic iron overload by tissue sampling or imaging
is a requisite to support the treatment decision.44 Iron removal by
3.10.2 | Phlebotomy in chronic hepatitis C
phlebotomy and stimulation of hepcidin production by hepcidin ago-
nists are being actively evaluated or considered. Biweekly phlebotomies for 3 months have improved serum ALT
levels in 33 patients with chronic hepatitis C,266 and a phlebotomy
regimen for 44‐144 months combined with a low iron diet has been
3.10 | Phlebotomy
associated with laboratory improvement and reduced risk of HCC in
Phlebotomy has been the standard method of treating iron overload 35 patients with moderate‐severe hepatic fibrosis and failure or
in patients with hereditary haemochromatosis,34,63,216,272,273 and oral intolerance of interferon therapy (Table 3).267 Serum ALT levels
12 | CZAJA

T A B L E 3 Evolving management strategies for iron overload in the absence of haemochromatosis


Strategy Rationale NonHaemochromatotic Experience
Phlebotomy Standard treatment for HH216,272,275 Experience in NAFLD patients:
Reduces hepatic iron content in HH34,63 ! Improves plasma insulin levels
263

Decreases oxidative stress in HH276 ! Reduces insulin resistance


264

Reduced profibrotic TGF‐β1 in HH276 ! Decreases serum ALT levels


263,277

! Improves histological features


265

! Small positive effect in phase II trial


265

Experience in chronic hepatitis C patients:


! Improves serum ALT level
266,267

! Reduces risk of HCC


267

! Improves frequency of SVR


268

! Antedates direct‐acting antiviral therapy


268

Oral iron chelation Substitute for phlebotomy in HH34,274,275 Deferoxamine experience in NAFLD model:
Deferasirox reduces iron burden in HH274 ! Up‐regulated insulin receptor
269

! Increased glucose transporter


269

Deferiprone in murine model of thalassemia:


! Removes iron with hepcidin agonists
286,287

279
Minihepcidins Truncated bioactive hepcidin derivative Murine models of β‐thalassemia and PV:
N‐terminus motif inhibits ferroportin279 ! Increased erythropoiesis
282

Reduced iron overload in HH model283 ! Improved anemia


282

Used in hepcidin deficiency models283 ! Decreased iron overload


282

TMRPSS6 inhibitors siRNA and ASOs inhibit TMPRSS6280,285 Increased hepcidin in thalassemia model280
siRNA delivered in lipid nanoparticles280 Decreased iron load in thalassemia model280
Designed to up‐regulate hepcidin gene189 Titratable, durable, reversible effect280
ASOs decreased iron in HH model285
Recombinant BMP6 Stimulates SMAD signalling182,184 Unassessed outside haemochromatosis289
Promotes hecidin gene expression184 Causes injection site calcification289
Increases hepcidin in murine HH289
Small hepcidin agonists Identified by molecular screening278,290 Unassessed278
Progesterone and genistein identified291,292 Lack specificity and proven efficacy278

ALT, alanine aminotransferase; ASOs, anti‐sense oligonucleotides; BMP6, bone morphogenetic protein 6; HH, hereditary haemochromatosis;
NAFLD, non‐alcoholic fatty liver disease; PV, polycythemia vera; siRNA, small interfering ribonucleic acids; SMAD, sons of mothers against
decapentaplegic pathway; SVR, sustained virological response; TGF‐β1, transforming growth factor‐beta 1; TMPRSS6, transmembrane protease
serine 6.

decreased in all patients on the long‐term phlebotomy regimen (re-


3.11 | Hepcidin replacement or stimulation
turning to normal in 69%), and the frequency of HCC was 5.7% in
the phlebotomised patients at 5 years and 8.6% at 10 years com- Most chronic liver diseases with iron overload outside the diagnosis
pared to 17.5% at 5 years and 39% at 10 years in the control of hereditary haemochromatosis have low serum hepcidin levels, and
patients. Multivariate analysis demonstrated that the risk of HCC correction of the hepcidin deficiency by replacement or stimulation
was significantly reduced by the long‐term iron reduction therapy is an evolving management strategy.99,103,278 The objective is to
267
(P = 0.03). Furthermore, a meta‐analysis of six prospective ran- restore iron homeostasis by decreasing duodenal absorption of iron
domised clinical trials has concluded that phlebotomy improves the and diminishing the release of ferric iron from intracellular storage
frequency of a SVR during interferon therapy (Odds ratio, 2.7; 95% sites. The hepcidin molecule is difficult to synthesise because of its
268
CI: 1.6‐4.5; P < 0.0001). complex secondary structure, and management by replacement of
The findings of phlebotomy in NAFLD and chronic hepatitis C the hepcidin molecule can be compromised by its rapid turn-
have been encouraging, but they have been underwhelming in a over.279,280 Small, biomimetic molecules that have hepcidin activity
phase II clinical trial of NAFLD265 and obtained prior to the availabil- and the development of hepcidin agonists promise to improve man-
ity of direct‐acting antiviral therapy for chronic hepatitis C. 268
agement opportunities.278
Results may be improved in NAFLD by better selection criteria for
phlebotomy, and current direct‐acting antiviral regimens may render
3.11.1 | Minihepcidins
phlebotomy unnecessary for chronic hepatitis C. Future investiga-
tions must establish the appropriate therapeutic target in chronic The N‐terminus of the hepcidin molecule is critical for binding with
281
liver diseases with iron overload outside of hereditary haemochro- ferroportin, and stable, truncated, bioactive derivatives of hep-
matosis and direct interventions to the iron overload, the primary cidin containing the critical 7‐9 amino acid motif at the N‐terminus
liver disease, or both. can inhibit ferroportin.279 These engineered peptides that have been
CZAJA | 13

designed to limit iron efflux into the circulation have been named
3.11.4 | Small molecule hepcidin agonists
minihepcidins. They have increased erythropoiesis, improved anemia,
and decreased iron overload in murine models of β‐thalassemia and Multiple small molecules that activate hepcidin pathways and increase
282
polycythemia vera (Table 3). In a hepcidin‐deficient murine model hepcidin production have been identified by molecular screen-
of severe haemochromatosis, minihepcidin has prevented hepatic ing.278,290 Progesterone and two other steroid molecules (epitiostanol
iron deposition, reduced myocardial iron content, increased duodenal and mifepristone) have been identified in zebrafish that increase hep-
iron retention, and redistributed iron from the liver to the spleen.283 cidin production outside the BMP and STAT3 pathways
278,291
Proprietary preparations of synthetic human hepcidin are also being (Table 3). These steroid molecules require progesterone recep-
assessed in a similar fashion.278 tor membrane component‐1 (PRMC‐1) to stimulate hepcidin produc-
tion. Genistein, the phytoestrogen in soybeans, is another small
molecule hepcidin agonist, and it can increase STAT3 phosphorylation
3.11.2 | TMPRSS6 Inhibitors
and up‐regulate hepcidin production in hepatoma cells.292 Small mole-
Transmembrane protease serine 6 (TMPRSS6), also named matriptase‐ cule hepcidin agonists lack specificity for hepcidin expression, and
2, is a membrane‐bound protease in hepatocytes that cleaves their ability to correct pathological deficiencies of hepcidin is unclear.
haemojuvelin from the hepatocyte surface, decreases activation of the Nevertheless, they underscore the diversity of hepcidin agonists in the
SMAD signalling pathway, blocks HAMP gene expression, and reduces environment, and the possibility that multiple factors outside the indi-
hepcidin production.189,284 The actions of TMPRSS6 can be inhibited vidual can modulate the hepcidin response in certain disease states.
by small interfering RNA (siRNA)280 and anti‐sense oligonucleotides Hepcidin agonists, especially the minihepcidins and TMPRSS6
(ASOs).285 The siRNAs are designed to have target‐specific inhibitors, constitute an evolving armamentarium for the hepcidin‐
complementary nucleic acids that silence the intended gene by binding deficient clinical syndromes,278 and iron‐overloaded patients with
to its mRNA and inducing cleavage and degradation of the chronic liver diseases other than hereditary haemochromatosis may
transcript.280 ASOs act similarly by targeting the mRNA of the become beneficiaries of the investigations now driven by the heredi-
TMPRSS6 gene.285 tary anemias and genetic haemochromatosis. Investigations to
TMPRSS6 siRNA that has been formulated in lipid nanoparti- improve the manifestations of oxidative stress in the liver158 and
cles can be cleared by the liver, and the administration of this studies that directly target the synthesis or activation of ferro-
preparation to a murine model of β‐thalassemia has increased hep- portin278 may also yield results that have applicability to highly
280
cidin production and decreased iron loading (Table 3). Further- selected cases of iron‐overloaded chronic liver disease.
more, the effect has been reversible, titratable, durable (80% gene
silencing after 14 days), relevant (hepatic hepcidin mRNA increased
3.12 | Overview
by 2‐3 fold through 7 days), and appropriate (serum iron level and
transferrin saturation decreased by 50% through 14 days).280 Sec- Manifestations of iron dysregulation are common in NAFLD,11,82
ond generation ASOs in murine haemochromatosis has also autoimmune hepatitis,24 chronic viral hepatitis,13,19,30,107,113 and
decreased serum iron, transferrin saturation, and hepatic iron accu- ALD.40,141,142 Hyperferritinemia,1–4 increased serum iron concentra-
285
mulation through a similar mechanism of TPMRSS6 inhibition. tions,11,24 elevated serum transferrin levels,11,24 stainable iron in liver
Both siRNA and ASO inhibitors of TMPRSS6 have been combined tissue,13,25,42 inappropriate serum hepcidin levels,24,28,30,31,124,154 and
with an iron chelator (deferiprone) to prevent secondary iron over- mutations of the HFE gene14,18,36 are findings that justify concern that
load in a murine model of β‐thalassemia, and the combination has iron overload is a pathogenic factor that could worsen outcome.
the prospect of removing iron already deposited and preventing This concern has been supported by clinical experiences associat-
further accumulation.286,287 ing the manifestations of iron overload with histological severity and
hepatic fibrosis (NAFLD),9,11,49 poor response to glucocorticoid treat-
ment (autoimmune hepatitis),24 development of HCC (ALD),116,147
3.11.3 | Recombinant BMP6
and reduced survival (ALD, chronic viral hepatitis).140,145,168 Studies
BMP6 stimulates the SMAD signalling pathway and increases hep- linking iron excess with oxidative stress and liver toxicity have incen-
cidin gene expression in hepatocytes.182,184 In a murine model of tivised investigative efforts to better diagnose and manage the iron
haemochromatosis, the proteins participating in the SMAD pathway disturbance.45,226,227
for hepcidin signalling are inappropriately low for the iron burden The pathogenic basis for the iron dysregulation in chronic liver
and the level of BMP6 mRNA, suggesting a block in BMP6 sig- diseases other than hereditary haemochromatosis is unclear, but it
nalling.288 Supra‐physiological doses of recombinant BMP6 can over- may relate to genetic (HFE and non‐HFE genes),63,173 epigenetic (tox-
come impaired BMP6‐SMAD signalling in murine haemochromatosis ins, nutrition),293,294 and disease‐related (cytokine responses, tissue
and increase hepcidin production (Table 3). 289
The effect can be hypoxia, hepatocyte damage)192,193,201 factors that disrupt iron
blocked by antibodies to BMP6. The major drawback has been the homeostasis. The serum hepcidin response to the liver injury may be
propensity for bone production and calcification at injection sites the pivotal factor in understanding the basis for the iron disturbance
after long‐term use.289 and directing management.
14 | CZAJA

Inappropriately low serum hepcidin levels have been recognised 4. Fargion S, Valenti L, Fracanzani AL. Beyond hereditary hemochro-
in most chronic liver diseases with iron overload,24,30,124–126,154 and matosis: new insights into the relationship between iron overload
and chronic liver diseases. Dig Liver Dis. 2011;43:89‐95.
the hepcidin agonists alone (minihepcidins279,282,283 and TMPRSS6
5. Siddique A, Kowdley KV. Review article: the iron overload syn-
inhibitors280,285) or in combination with an oral iron chelator286,287 dromes. Aliment Pharmacol Ther. 2012;35:876‐893.
are emerging as promising interventions in the hereditary anemias 6. Bell H, Skinningsrud A, Raknerud N, Try K. Serum ferritin and trans-
and genetic haemochromatosis. Therapies that stimulate or replace ferrin saturation in patients with chronic alcoholic and non‐alcoholic
liver diseases. J Intern Med. 1994;236:315‐322.
hepcidin production may have application in the hepcidin‐deficient
7. Moirand R, Mendler MH, Guillygomarc'h A, Brissot P, Deugnier Y.
chronic liver diseases that are outside the diagnosis of hereditary Non‐alcoholic steatohepatitis with iron: part of insulin resistance‐as-
haemochromatosis. Importantly, the iron disturbances in these sociated hepatic iron overload? J Hepatol. 2000;33:1024‐1026.
chronic liver diseases are probably a consequence rather than a 8. Aigner E, Theurl I, Haufe H, et al. Copper availability contributes to
iron perturbations in human nonalcoholic fatty liver disease. Gas-
cause of the liver disease, and successful management of the
troenterology. 2008;135:680‐688.
primary liver disease remains a necessary, and possibly sufficient, 9. Manousou P, Kalambokis G, Grillo F, et al. Serum ferritin is a dis-
strategy for correcting the iron disturbance.44 criminant marker for both fibrosis and inflammation in histologically
Future investigations must continue to understand the proven non‐alcoholic fatty liver disease patients. Liver Int.
2011;31:730‐739.
pathogenic association between the iron overload and liver
10. Valenti L, Swinkels DW, Burdick L, et al. Serum ferritin levels are
disease and develop confident, measurable thresholds of hepatic
associated with vascular damage in patients with nonalcoholic fatty
iron toxicity that compel intervention. Studies based on hepatic liver disease. Nutr Metab Cardiovasc Dis. 2011;21:568‐575.
iron content must refine correlations with surrogate markers 11. Kowdley KV, Belt P, Wilson LA, et al. Elevated serum ferritin is an
of iron overload to eliminate overestimations of risk,44 and cost‐ independent predictor of histologic severity and advanced fibrosis
in patients with nonalcoholic fatty liver disease. Hepatology.
effective non‐invasive evaluations of hepatic iron content, such as
2012;55:77‐85.
the room temperature susceptometer,224 must continue to be 12. Aigner E, Weiss G, Datz C. Dysregulation of iron and copper home-
developed and applied. ostasis in nonalcoholic fatty liver. World J Hepatol. 2015;7:177‐188.
13. Sebastiani G, Tempesta D, Alberti A. Hepatic iron overload is com-
mon in chronic hepatitis B and is more severe in patients coin-
ACKNOWLEDGEMENTS fected with hepatitis D virus. J Viral Hepat. 2012;19:e170‐176.
14. Bonkovsky HL, Troy N, McNeal K, et al. Iron and HFE or TfR1
Declaration of personal interests: None. mutations as comorbid factors for development and progression of
chronic hepatitis C. J Hepatol. 2002;37:848‐854.
15. Corengia C, Galimberti S, Bovo G, et al. Iron accumulation in
AUTHORSHIP chronic hepatitis C: relation of hepatic iron distribution, HFE geno-
type, and disease course. Am J Clin Pathol. 2005;124:846‐853.
Guarantor of the article: Albert J. Czaja, MD. 16. D'Souza RF, Feakins R, Mears L, Sabin CA, Foster GR. Relationship
Author contributions: Dr Albert J. Czaja conceived, researched, between serum ferritin, hepatic iron staining, diabetes mellitus and
fibrosis progression in patients with chronic hepatitis C. Aliment
designed, and wrote the manuscript without other writing assistance,
Pharmacol Ther. 2005;21:519‐524.
research support or financial aid from a funding agency or institu- 17. Rigamonti C, Andorno S, Maduli E, Capelli F, Boldorini R, Sartori M.
tion. The three fully referenced tables were developed by Albert J. Gender and liver fibrosis in chronic hepatitis: the role of iron status.
Czaja, MD exclusively for this review. The one colour figure is origi- Aliment Pharmacol Ther. 2005;21:1445‐1451.
18. Bonkovsky HL, Naishadham D, Lambrecht RW, et al. Roles of iron
nal and previously unpublished. The figure was created by Dr Albert
and HFE mutations on severity and response to therapy during
Czaja exclusively for this review. Dr Albert J. Czaja has no conflicts retreatment of advanced chronic hepatitis C. Gastroenterology.
of interests to declare, and he has reviewed and approved the final 2006;131:1440‐1451.
version of the manuscript prior to its submission, including the 19. Valenti L, Pulixi EA, Arosio P, et al. Relative contribution of iron
genes, dysmetabolism and hepatitis C virus (HCV) in the pathogen-
authorship list.
esis of altered iron regulation in HCV chronic hepatitis. Haematolog-
ica. 2007;92:1037‐1042.
20. Valenti L, Dongiovanni P, Piperno A, et al. Alpha 1‐antitrypsin
ORCID mutations in NAFLD: high prevalence and association with altered
iron metabolism but not with liver damage. Hepatology.
Albert J. Czaja https://orcid.org/0000-0002-5024-3065
2006;44:857‐864.
21. Ghio AJ, Soukup JM, Richards JH, Fischer BM, Voynow JA, Sch-
mechel DE. Deficiency of alpha‐1‐antitrypsin influences systemic
REFERENCES iron homeostasis. Int J Chron Obstruct Pulmon Dis. 2013;8:45‐51.
22. Schaefer B, Haschka D, Finkenstedt A, et al. Impaired hepcidin
1. Alla V, Bonkovsky HL. Iron in nonhemochromatotic liver disorders. expression in alpha‐1‐antitrypsin deficiency associated with iron
Semin Liver Dis. 2005;25:461‐472. overload and progressive liver disease. Hum Mol Genet.
2. Hearnshaw S, Thompson NP, McGill A. The epidemiology of hyper- 2015;24:6254‐6263.
ferritinaemia. World J Gastroenterol. 2006;12:5866‐5869. 23. An IC, Tiwari AK, Ameda S, Laird‐Fick HS. Autoimmune hepatitis:
3. Camaschella C, Poggiali E. Towards explaining "unexplained hyper- diagnostic dilemma in the setting of suspected iron overload. Case
ferritinemia". Haematologica. 2009;94:307‐309. Rep Gastrointest Med. 2013;2013:872987.
CZAJA | 15

24. Taubert R, Hardtke‐Wolenski M, Noyan F, et al. Hyperferritinemia 46. Ganz T. Systemic iron homeostasis. Physiol Rev. 2013;93:1721–
and hypergammaglobulinemia predict the treatment response to 1741.
standard therapy in autoimmune hepatitis. PLoS ONE. 2017;12: 47. Rametta R, Dongiovanni P, Pelusi S, et al. Hepcidin resistance in
e0179074. dysmetabolic iron overload. Liver Int. 2016;36:1540–1548.
25. Ludwig J, Hashimoto E, Porayko MK, Moyer TP, Baldus WP. 48. Riva A, Trombini P, Mariani R, et al. Revaluation of clinical and his-
Hemosiderosis in cirrhosis: a study of 447 native livers. Gastroen- tological criteria for diagnosis of dysmetabolic iron overload syn-
terology. 1997;112:882‐888. drome. World J Gastroenterol. 2008;14:4745–4752.
26. Abu Rajab M, Guerin L, Lee P, Brown KE. Iron overload secondary 49. Nelson JE, Wilson L, Brunt EM, et al; Nonalcoholic Steatohepatitis
to cirrhosis: a mimic of hereditary haemochromatosis? Histopathol- Clinical Research N. Relationship between the pattern of hepatic
ogy. 2014;65:561‐569. iron deposition and histological severity in nonalcoholic fatty liver
27. Geramizadeh B, Ghazanfari Y, Nikeghbalian S, Malekhosseini SA. A disease. Hepatology. 2011;53:448–457.
single center study comparing the stainable iron depositions in 50. Fujita N, Sugimoto R, Motonishi S, et al. Patients with chronic hep-
1000 explanted cirrhotic livers of different causes. Hepat Mon. atitis C achieving a sustained virological response to peginterferon
2015;15:e33710. and ribavirin therapy recover from impaired hepcidin secretion. J
28. Lyberopoulou A, Chachami G, Gatselis NK, et al. Low serum hep- Hepatol. 2008;49:702–710.
cidin in patients with autoimmune liver diseases. PLoS ONE. 51. Maiwall R, Kumar S, Chaudhary AK, et al. Serum ferritin predicts
2015;10:e0135486. early mortality in patients with decompensated cirrhosis. J Hepatol.
29. Iqbal T, Diab A, Ward DG, et al. Is iron overload in alcohol‐related 2014;61:43–50.
cirrhosis mediated by hepcidin? World J Gastroenterol. 52. Oikonomou T, Goulis I, Soulaidopoulos S, et al. High serum ferritin
2009;15:5864‐5866. is associated with worse outcome of patients with decompensated
30. Lin D, Ding J, Liu JY, et al. Decreased serum hepcidin concentration cirrhosis. Ann Gastroenterol. 2017;30:217–224.
correlates with brain iron deposition in patients with HBV‐related 53. Umer N, Makki MU, Kiran SK, Jadoon NA. Serum ferritin as a pre-
cirrhosis. PLoS ONE. 2013;8:e65551. dictor of 30 days mortality in patients of decompensated chronic
31. Auguet T, Aragones G, Berlanga A, et al. Hepcidin in morbidly liver disease. J Ayub Med Coll Abbottabad. 2017;29:415–418.
obese women with non‐alcoholic fatty liver disease. PLoS ONE. 54. Halliwell B, Gutteridge JM. Oxygen toxicity, oxygen radicals, transi-
2017;12:e0187065. tion metals and disease. Biochem J. 1984;219:1–14.
32. Marmur J, Beshara S, Eggertsen G, et al. Hepcidin levels correlate 55. Halliwell B, Gutteridge JM, Cross CE. Free radicals, antioxidants,
to liver iron content, but not steatohepatitis, in non‐alcoholic fatty and human disease: where are we now? J Lab Clin Med.
liver disease. BMC Gastroenterol. 2018;18:78. 1992;119:598–620.
33. Adams P, Brissot P, Powell LW. EASL International Consensus Con- 56. Siah CW, Ombiga J, Adams LA, Trinder D, Olynyk JK. Normal iron
ference on Haemochromatosis. J Hepatol. 2000;33:485–504. metabolism and the pathophysiology of iron overload disorders. Clin
34. Pietrangelo A. Hereditary hemochromatosis: pathogenesis, diagno- Biochem Rev. 2006;27:5–16.
sis, and treatment. Gastroenterology. 2010;139:393–408 e391–392. 57. Milto IV, Suhodolo IV, Prokopieva VD, Klimenteva TK. Molecular
35. Grove J, Daly AK, Burt AD, et al. Heterozygotes for HFE mutations and cellular bases of Iron metabolism in humans. Biochemistry
have no increased risk of advanced alcoholic liver disease. Gut. (Mosc). 2016;81:549–564.
1998;43:262–266. 58. McKie AT, Barrow D, Latunde‐Dada GO, et al. An iron‐regulated
36. Bonkovsky HL, Jawaid Q, Tortorelli K, et al. Non‐alcoholic steato- ferric reductase associated with the absorption of dietary iron.
hepatitis and iron: increased prevalence of mutations of the HFE Science. 2001;291:1755–1759.
gene in non‐alcoholic steatohepatitis. J Hepatol. 1999;31:421– 59. Gunshin H, Mackenzie B, Berger UV, et al. Cloning and characteri-
429. zation of a mammalian proton‐coupled metal‐ion transporter. Nat-
37. Mendler MH, Turlin B, Moirand R, et al. Insulin resistance‐associ- ure. 1997;388:482–488.
ated hepatic iron overload. Gastroenterology. 1999;117:1155–1163. 60. Conrad ME, Umbreit JN. Pathways of iron absorption. Blood Cells
38. Hohler T, Leininger S, Kohler HH, Schirmacher P, Galle PR. Mol Dis. 2002;29:336–355.
Heterozygosity for the hemochromatosis gene in liver diseases– 61. McKie AT, Marciani P, Rolfs A, et al. A novel duodenal iron‐regu-
prevalence and effects on liver histology. Liver. 2000;20:482–486. lated transporter, IREG1, implicated in the basolateral transfer of
39. Bugianesi E, Manzini P, D'Antico S, et al. Relative contribution of iron to the circulation. Mol Cell. 2000;5:299–309.
iron burden, HFE mutations, and insulin resistance to fibrosis in 62. Donovan A, Lima CA, Pinkus JL, et al. The iron exporter ferro-
nonalcoholic fatty liver. Hepatology. 2004;39:179–187. portin/Slc40a1 is essential for iron homeostasis. Cell Metab.
40. Gleeson D, Evans S, Bradley M, et al. HFE genotypes in decompen- 2005;1:191–200.
sated alcoholic liver disease: phenotypic expression and comparison 63. Pietrangelo A. Genetics, genetic testing, and management of
with heavy drinking and with normal controls. Am J Gastroenterol. hemochromatosis: 15 years since hepcidin. Gastroenterology.
2006;101:304–310. 2015;149:1240–1251.
41. Gattoni A, Parlato A, Vangieri B, Bresciani M, Derna R, Baldassarre 64. Nemeth E, Tuttle MS, Powelson J, et al. Hepcidin regulates cellular
R. Role of hemochromatosis genes in chronic hepatitis C. Clin Ter. iron efflux by binding to ferroportin and inducing its internalization.
2006;157:61–68. Science. 2004;306:2090–2093.
42. Valenti L, Fracanzani AL, Bugianesi E, et al. HFE genotype, parenchy- 65. Rossi E. Hepcidin–the iron regulatory hormone. Clin Biochem Rev.
mal iron accumulation, and liver fibrosis in patients with nonalcoholic 2005;26:47–49.
fatty liver disease. Gastroenterology. 2010;138:905–912. 66. Hentze MW, Muckenthaler MU, Galy B, Camaschella C. Two to tango:
43. Bhagat CI, Fletcher S, Joseph J, Beilby JP. Plasma ferritin in acute regulation of Mammalian iron metabolism. Cell. 2010;142:24–38.
hepatocellular damage. Clin Chem. 2000;46:885–886. 67. Qiao B, Sugianto P, Fung E, et al. Hepcidin‐induced endocytosis of
44. Beaton MD, Adams PC. Treatment of hyperferritinemia. Ann Hepa- ferroportin is dependent on ferroportin ubiquitination. Cell Metab.
tol. 2012;11:294–300. 2012;15:918–924.
45. Harrison PM, Arosio P. The ferritins: molecular properties, iron stor- 68. Gulec S, Anderson GJ, Collins JF. Mechanistic and regulatory
age function and cellular regulation. Biochim Biophys Acta. aspects of intestinal iron absorption. Am J Physiol Gastrointest Liver
1996;1275:161–203. Physiol. 2014;307:G397–409.
16 | CZAJA

69. Vulpe CD, Kuo YM, Murphy TL, et al. Hephaestin, a ceruloplasmin 90. Handa P, Morgan‐Stevenson V, Maliken BD, et al. Iron overload
homologue implicated in intestinal iron transport, is defective in the results in hepatic oxidative stress, immune cell activation, and hepa-
sla mouse. Nat Genet. 1999;21:195–199. tocellular ballooning injury, leading to nonalcoholic steatohepatitis
70. Hemmaplardh D, Morgan EH. The role of endocytosis in transferrin in genetically obese mice. Am J Physiol Gastrointest Liver Physiol.
uptake by reticulocytes and bone marrow cells. Br J Haematol. 2016;310:G117–127.
1977;36:85–96. 91. Yeh MM, Brunt EM. Pathology of nonalcoholic fatty liver disease.
71. Gkouvatsos K, Papanikolaou G, Pantopoulos K. Regulation of iron Am J Clin Pathol. 2007;128:837–847.
transport and the role of transferrin. Biochim Biophys Acta. 92. Caldwell S, Ikura Y, Dias D, et al. Hepatocellular ballooning in
2012;1820:188–202. NASH. J Hepatol. 2010;53:719–723.
72. Arosio P, Ingrassia R, Cavadini P. Ferritins: a family of molecules for 93. Yeh MM, Brunt EM. Pathological features of fatty liver disease.
iron storage, antioxidation and more. Biochim Biophys Acta. Gastroenterology. 2014;147:754–764.
2009;1790:589–599. 94. Atarashi M, Izawa T, Miyagi R, et al. Dietary iron supplementa-
73. Paul BT, Manz DH, Torti FM, Torti SV. Mitochondria and Iron: cur- tion alters hepatic inflammation in a rat model of nonalcoholic
rent questions. Expert Rev Hematol. 2017;10:65–79. steatohepatitis. Nutrients. 2018;10:175.
74. Ritchie RF, Palomaki GE, Neveux LM, Navolotskaia O, Ledue TB, 95. Dongiovanni P, Fracanzani AL, Fargion S, Valenti L. Iron in fatty
Craig WY. Reference distributions for the negative acute‐phase liver and in the metabolic syndrome: a promising therapeutic target.
serum proteins, albumin, transferrin and transthyretin: a practical, J Hepatol. 2011;55:920–932.
simple and clinically relevant approach in a large cohort. J Clin Lab 96. Deugnier Y, Bardou‐Jacquet E, Laine F. Dysmetabolic iron overload
Anal. 1999;13:273–279. syndrome (DIOS). Presse Med. 2017;46:e306–e311.
75. Potter BJ, Chapman RW, Nunes RM, Sorrentino D, Sherlock S. 97. Datz C, Muller E, Aigner E. Iron overload and non‐alcoholic fatty
Transferrin metabolism in alcoholic liver disease. Hepatology. liver disease. Minerva Endocrinol. 2017;42:173–183.
1985;5:714–721. 98. Pietrangelo A. Hereditary hemochromatosis. Biochim Biophys Acta.
76. Bruns T, Nuraldeen R, Mai M, et al. Low serum transferrin corre- 2006;1763:700–710.
lates with acute‐on‐chronic organ failure and indicates short‐term 99. Pietrangelo A. Hepcidin in human iron disorders: therapeutic impli-
mortality in decompensated cirrhosis. Liver Int. 2017;37:232–241. cations. J Hepatol. 2011;54:173–181.
77. Fischbach FA, Gregory DW, Harrison PM, Hoy TG, Williams JM. 100. Bekri S, Gual P, Anty R, et al. Increased adipose tissue expression
On the structure of hemosiderin and its relationship to ferritin. J of hepcidin in severe obesity is independent from diabetes and
Ultrastruct Res. 1971;37:495–503. NASH. Gastroenterology. 2006;131:788–796.
78. Iancu TC. Ferritin and hemosiderin in pathological tissues. Electron 101. Peyssonnaux C, Zinkernagel AS, Datta V, Lauth X, Johnson RS,
Microsc Rev. 1992;5:209–229. Nizet V. TLR4‐dependent hepcidin expression by myeloid cells in
79. O'Connell M, Halliwell B, Moorhouse CP, Aruoma OI, Baum H. response to bacterial pathogens. Blood. 2006;107:3727–3732.
Peters TJ. Formation of hydroxyl radicals in the presence of ferritin 102. Trombini P, Paolini V, Pelucchi S, et al. Hepcidin response to acute
and haemosiderin. Is haemosiderin formation a biological protective iron intake and chronic iron loading in dysmetabolic iron overload
mechanism? Biochem J. 1986;234:727–731. syndrome. Liver Int. 2011;31:994–1000.
80. Ozaki M, Kawabata T, Awai M. Iron release from haemosiderin and 103. Swinkels DW, Drenth JP. Hepcidin in the management of patients
production of iron‐catalysed hydroxyl radicals in vitro. Biochem J. with mild non‐hemochromatotic iron overload: fact or fiction? J
1988;250:589–595. Hepatol. 2008;49:680–685.
81. Wright TL, Brissot P, Ma WL, Weisiger RA. Characterization of 104. Deugnier YM, Charalambous P, Le Quilleuc D, et al. Preneoplastic
non‐transferrin‐bound iron clearance by rat liver. J Biol Chem. significance of hepatic iron‐free foci in genetic hemochromatosis: a
1986;261:10909–10914. study of 185 patients. Hepatology. 1993;18:1363–1369.
82. Bacon BR, Farahvash MJ, Janney CG, Neuschwander‐Tetri BA. 105. Bohne F, Martinez‐Llordella M, Lozano JJ, et al. Intra‐graft expres-
Nonalcoholic steatohepatitis: an expanded clinical entity. Gastroen- sion of genes involved in iron homeostasis predicts the develop-
terology. 1994;107:1103–1109. ment of operational tolerance in human liver transplantation. J Clin
83. Brudevold R, Hole T, Hammerstrom J. Hyperferritinemia is associ- Invest. 2012;122:368–382.
ated with insulin resistance and fatty liver in patients without iron 106. Vela D. Low hepcidin in liver fibrosis and cirrhosis; a tale of pro-
overload. PLoS ONE. 2008;3:e3547. gressive disorder and a case for a new biochemical marker. Mol
84. Fargion S, Mattioli M, Fracanzani AL, et al. Hyperferritinemia, iron Med. 2018;24:5.
overload, and multiple metabolic alterations identify patients at risk for 107. Guyader D, Thirouard AS, Erdtmann L, et al. Liver iron is a surro-
nonalcoholic steatohepatitis. Am J Gastroenterol. 2001;96:2448–2455. gate marker of severe fibrosis in chronic hepatitis C. J Hepatol.
85. Valenti L, Canavesi E, Galmozzi E, et al. Beta‐globin mutations are 2007;46:587–595.
associated with parenchymal siderosis and fibrosis in patients with 108. Hezode C, Cazeneuve C, Coue O, et al. Liver iron accumulation in
non‐alcoholic fatty liver disease. J Hepatol. 2010;53:927–933. patients with chronic active hepatitis C: prevalence and role of
86. Yoneda M, Nozaki Y, Endo H, et al. Serum ferritin is a clinical bio- hemochromatosis gene mutations and relationship with hepatic his-
marker in Japanese patients with nonalcoholic steatohepatitis tological lesions. J Hepatol. 1999;31:979–984.
(NASH) independent of HFE gene mutation. Dig Dis Sci. 109. Shan Y, Lambrecht RW, Bonkovsky HL. Association of hepatitis C
2010;55:808–814. virus infection with serum iron status: analysis of data from the
87. Younossi ZM, Gramlich T, Bacon BR, et al. Hepatic iron and nonal- third National Health and Nutrition Examination Survey. Clin Infect
coholic fatty liver disease. Hepatology. 1999;30:847–850. Dis. 2005;40:834–841.
88. Angulo P, Keach JC, Batts KP, Lindor KD. Independent predictors 110. Czaja AJ, Carpenter HA, Santrach PJ, Moore SB. Host‐ and disease‐
of liver fibrosis in patients with nonalcoholic steatohepatitis. Hepa- specific factors affecting steatosis in chronic hepatitis C. J Hepatol.
tology. 1999;30:1356–1362. 1998;29:198–206.
89. Angulo P, George J, Day CP, et al. Serum ferritin levels lack diag- 111. Rubbia‐Brandt L, Quadri R, Abid K, et al. Hepatocyte steatosis is a
nostic accuracy for liver fibrosis in patients with nonalcoholic fatty cytopathic effect of hepatitis C virus genotype 3. J Hepatol.
liver disease. Clin Gastroenterol Hepatol. 2014;12:1163‐1169 2000;33:106–115.
e1161.
CZAJA | 17

112. Kumar D, Farrell GC, Fung C, George J. Hepatitis C virus genotype 133. Bartolomei G, Cevik RE, Marcello A. Modulation of hepatitis C virus
3 is cytopathic to hepatocytes: reversal of hepatic steatosis after replication by iron and hepcidin in Huh7 hepatocytes. J Gen Virol.
sustained therapeutic response. Hepatology. 2002;36:1266–1272. 2011;92:2072–2081.
113. Sebastiani G, Vario A, Ferrari A, Pistis R, Noventa F, Alberti A. 134. Fillebeen C, Rivas‐Estilla AM, Bisaillon M, et al. Iron inactivates the
Hepatic iron, liver steatosis and viral genotypes in patients with RNA polymerase NS5B and suppresses subgenomic replication of
chronic hepatitis C. J Viral Hepat. 2006;13:199–205. hepatitis C Virus. J Biol Chem. 2005;280:9049–9057.
114. Monto A, Alonzo J, Watson JJ, Grunfeld C, Wright TL. Steatosis in 135. Fillebeen C, Pantopoulos K. Iron inhibits replication of infectious
chronic hepatitis C: relative contributions of obesity, diabetes melli- hepatitis C virus in permissive Huh7.5.1 cells. J Hepatol.
tus, and alcohol. Hepatology. 2002;36:729–736. 2010;53:995–999.
115. Furutani T, Hino K, Okuda M, et al. Hepatic iron overload induces 136. Ma L, Zou T, Yuan Y, et al. Duodenal ferroportin is up‐regulated in
hepatocellular carcinoma in transgenic mice expressing the hepatitis patients with chronic hepatitis C. PLoS ONE. 2014;9:e110658.
C virus polyprotein. Gastroenterology. 2006;130:2087–2098. 137. Sato M, Miyanishi K, Tanaka S, et al. Increased duodenal iron
116. Nahon P, Sutton A, Rufat P, et al. Liver iron, HFE gene mutations, absorption through upregulation of ferroportin 1 due to decrement
and hepatocellular carcinoma occurrence in patients with cirrhosis. in serum hepcidin in patients with chronic hepatitis C. Can J Gas-
Gastroenterology. 2008;134:102–110. troenterol Hepatol. 2018;2018:2154361.
117. Smith BC, Gorve J, Guzail MA, et al. Heterozygosity for hereditary 138. Tai AW, Benita Y, Peng LF, et al. A functional genomic screen iden-
hemochromatosis is associated with more fibrosis in chronic hepati- tifies cellular cofactors of hepatitis C virus replication. Cell Host
tis C. Hepatology. 1998;27:1695–1699. Microbe. 2009;5:298–307.
118. Erhardt A, Maschner‐Olberg A, Mellenthin C, et al. HFE mutations 139. Aoki CA, Rossaro L, Ramsamooj R, Brandhagen D, Burritt MF,
and chronic hepatitis C: H63D and C282Y heterozygosity are inde- Bowlus CL. Liver hepcidin mRNA correlates with iron stores, but
pendent risk factors for liver fibrosis and cirrhosis. J Hepatol. not inflammation, in patients with chronic hepatitis C. J Clin Gas-
2003;38:335–342. troenterol. 2005;39:71–74.
119. Tung BY, Emond MJ, Bronner MP, Raaka SD, Cotler SJ, Kowdley 140. Wu J, Chen L, Chen Y, Yang J, Wu D. Serum ferritin concentration
KV. Hepatitis C, iron status, and disease severity: relationship with predicts mortality in patients with hepatitis B virus‐related acute on
HFE mutations. Gastroenterology. 2003;124:318–326. chronic liver failure. Arch Med Res. 2014;45:251–256.
120. Sartori M, Andorno S, Pagliarulo M, et al. Heterozygous beta‐globin 141. Machado MV, Ravasco P, Martins A, Almeida MR, Camilo ME, Cor-
gene mutations as a risk factor for iron accumulation and liver tez‐Pinto H. Iron homeostasis and H63D mutations in alcoholics
fibrosis in chronic hepatitis C. Gut. 2007;56:693–698. with and without liver disease. World J Gastroenterol. 2009;15:106–
121. Thorburn D, Curry G, Spooner R, et al. The role of iron and 111.
haemochromatosis gene mutations in the progression of liver dis- 142. Costa‐Matos L, Batista P, Monteiro N, Henriques P, Girao F, Car-
ease in chronic hepatitis C. Gut. 2002;50:248–252. valho A. Hfe mutations and iron overload in patients with alcoholic
122. Hevi S, Chuck SL. Ferritins can regulate the secretion of liver disease. Arq Gastroenterol. 2013;50:35–41.
apolipoprotein B. J Biol Chem. 2003;278:31924–31929. 143. Pascoe A, Kerlin P, Steadman C, et al. Spur cell anaemia and hep-
123. Bugianesi E, Marchesini G, Gentilcore E, et al. Fibrosis in genotype 3 atic iron stores in patients with alcoholic liver disease undergoing
chronic hepatitis C and nonalcoholic fatty liver disease: Role of insulin orthotopic liver transplantation. Gut. 1999;45:301–305.
resistance and hepatic steatosis. Hepatology. 2006;44:1648–1655. 144. Raynard B, Balian A, Fallik D, et al. Risk factors of fibrosis in alco-
124. Fujita N, Sugimoto R, Takeo M, et al. Hepcidin expression in the hol‐induced liver disease. Hepatology. 2002;35:635–638.
liver: relatively low level in patients with chronic hepatitis C. Mol 145. Ganne‐Carrie N, Christidis C, Chastang C, et al. Liver iron is predic-
Med. 2007;13:97–104. tive of death in alcoholic cirrhosis: a multivariate study of 229 con-
125. Girelli D, Pasino M, Goodnough JB, et al. Reduced serum hepcidin secutive patients with alcoholic and/or hepatitis C virus cirrhosis: a
levels in patients with chronic hepatitis C. J Hepatol. 2009;51:845– prospective follow up study. Gut. 2000;46:277–282.
852. 146. Frenzer A, Rudzki Z, Norton ID, Butler WJ, Roberts‐Thomson IC.
126. Horl WH, Schmidt A. Low hepcidin triggers hepatic iron accumula- Heterozygosity of the haemochromatosis mutation, C282Y, does
tion in patients with hepatitis C. Nephrol Dial Transplant. not influence susceptibility to alcoholic cirrhosis. Scand J Gastroen-
2014;29:1141–1144. terol. 1998;33:1324.
127. Radicheva MP, Andonova AN, Milcheva HT, et al. Serum markers 147. Lauret E, Rodriguez M, Gonzalez S, et al. HFE gene mutations in
of iron metabolism in chronic liver diseases. Open Access Maced J alcoholic and virus‐related cirrhotic patients with hepatocellular car-
Med Sci. 2018;6:1010–1016. cinoma. Am J Gastroenterol. 2002;97:1016–1021.
128. Sikorska K, Romanowski T, Stalke P, Izycka Swieszewska E, Bie- 148. Harrison‐Findik DD, Schafer D, Klein E, et al. Alcohol metabolism‐
lawski KP. Association of hepcidin mRNA expression with hepato- mediated oxidative stress down‐regulates hepcidin transcription
cyte iron accumulation and effects of antiviral therapy in chronic and leads to increased duodenal iron transporter expression. J Biol
hepatitis C infection. Hepat Mon. 2014;14:e21184. Chem. 2006;281:22974–22982.
129. Fujita N, Sugimoto R, Ma N, et al. Comparison of hepatic oxidative 149. Bridle K, Cheung TK, Murphy T, et al. Hepcidin is down‐regulated
DNA damage in patients with chronic hepatitis B and C. J Viral in alcoholic liver injury: implications for the pathogenesis of alco-
Hepat. 2008;15:498–507. holic liver disease. Alcohol Clin Exp Res. 2006;30:106–112.
130. Nishina S, Hino K, Korenaga M, et al. Hepatitis C virus‐induced 150. Ramji DP, Foka P. CCAAT/enhancer‐binding proteins: structure,
reactive oxygen species raise hepatic iron level in mice by reducing function and regulation. Biochem J. 2002;365:561–575.
hepcidin transcription. Gastroenterology. 2008;134:226–238. 151. Stephens JM, Pekala PH. Transcriptional repression of the C/EBP‐
131. Miura K, Taura K, Kodama Y, Schnabl B, Brenner DA. Hepatitis C alpha and GLUT4 genes in 3T3‐L1 adipocytes by tumor necrosis
virus‐induced oxidative stress suppresses hepcidin expression factor‐alpha. Regulations is coordinate and independent of protein
through increased histone deacetylase activity. Hepatology. synthesis. J Biol Chem. 1992;267:13580–13584.
2008;48:1420–1429. 152. Tengku‐Muhammad TS, Hughes TR, Ranki H, Cryer A, Ramji DP.
132. Liu H, Trinh TL, Dong H, Keith R, Nelson D, Liu C. Iron regulator Differential regulation of macrophage CCAAT‐enhancer binding
hepcidin exhibits antiviral activity against hepatitis C virus. PLoS protein isoforms by lipopolysaccharide and cytokines. Cytokine.
ONE. 2012;7:e46631. 2000;12:1430–1436.
18 | CZAJA

153. Alam T, An MR, Papaconstantinou J. Differential expression of 175. Fletcher LM, Dixon JL, Purdie DM, Powell LW, Crawford DH.
three C/EBP isoforms in multiple tissues during the acute phase Excess alcohol greatly increases the prevalence of cirrhosis in
response. J Biol Chem. 1992;267:5021–5024. hereditary hemochromatosis. Gastroenterology. 2002;122:281–
154. Costa‐Matos L, Batista P, Monteiro N, et al. Liver hepcidin mRNA 289.
expression is inappropriately low in alcoholic patients compared 176. Zaloumis SG, Allen KJ, Bertalli NA, et al; HealthIron Study I. Natural
with healthy controls. Eur J Gastroenterol Hepatol. 2012;24:1158– history of HFE simple heterozygosity for C282Y and H63D: a
1165. prospective 12‐year study. J Gastroenterol Hepatol. 2015;30:719–725.
155. Milic S, Mikolasevic I, Orlic L, et al. The role of iron and iron over- 177. Ganz T, Nemeth E. Hepcidin and disorders of iron metabolism. Ann
load in chronic liver disease. Med Sci Monit. 2016;22:2144–2151. Rev Med. 2011;62:347–360.
156. Dostalikova‐Cimburova M, Balusikova K, Kratka K, et al. Role of 178. Schmidt PJ, Toran PT, Giannetti AM, Bjorkman PJ, Andrews NC.
duodenal iron transporters and hepcidin in patients with alcoholic The transferrin receptor modulates Hfe‐dependent regulation of
liver disease. J Cell Mol Med. 2014;18:1840–1850. hepcidin expression. Cell Metab. 2008;7:205–214.
157. Mueller S, Rausch V. The role of iron in alcohol‐mediated hepato- 179. Gao J, Chen J, Kramer M, Tsukamoto H, Zhang AS, Enns CA. Inter-
carcinogenesis. Adv Exp Med Biol. 2015;815:89–112. action of the hereditary hemochromatosis protein HFE with trans-
158. Czaja AJ. Nature and implications of oxidative and nitrosative stres- ferrin receptor 2 is required for transferrin‐induced hepcidin
ses in autoimmune hepatitis. Dig Dis Sci. 2016;61:2784–2803. expression. Cell Metab. 2009;9:217–227.
159. Silva I, Rausch V, Seitz HK, Mueller S. Does hypoxia cause carcino- 180. Ramey G, Deschemin JC, Vaulont S. Cross‐talk between the mito-
genic iron accumulation in alcoholic liver disease (ALD)? Cancers gen activated protein kinase and bone morphogenetic protein/he-
(Basel). 2017;9:145. mojuvelin pathways is required for the induction of hepcidin by
160. Tan TC, Crawford DH, Jaskowski LA, et al. Excess iron modulates holotransferrin in primary mouse hepatocytes. Haematologica.
endoplasmic reticulum stress‐associated pathways in a mouse 2009;94:765–772.
model of alcohol and high‐fat diet‐induced liver injury. Lab Invest. 181. Babitt JL, Huang FW, Xia Y, Sidis Y, Andrews NC, Lin HY. Modula-
2013;93:1295–1312. tion of bone morphogenetic protein signaling in vivo regulates sys-
161. Arteel GE, Raleigh JA, Bradford BU, Thurman RG. Acute alcohol temic iron balance. J Clin Invest. 2007;117:1933–1939.
produces hypoxia directly in rat liver tissue in vivo: role of Kupffer 182. Wang RH, Li C, Xu X, et al. A role of SMAD4 in iron metabolism
cells. Am J Physiol. 1996;271:G494–500. through the positive regulation of hepcidin expression. Cell Metab.
162. Arteel GE, Iimuro Y, Yin M, Raleigh JA, Thurman RG. Chronic ent- 2005;2:399–409.
eral ethanol treatment causes hypoxia in rat liver tissue in vivo. 183. Kautz L, Meynard D, Monnier A, et al. Iron regulates phosphoryla-
Hepatology. 1997;25:920–926. tion of Smad1/5/8 and gene expression of Bmp6, Smad 7, Id1, and
163. Nicolas G, Chauvet C, Viatte L, et al. The gene encoding the iron Atoh8 in the mouse liver. Blood. 2008;112:1503–1509.
regulatory peptide hepcidin is regulated by anemia, hypoxia, and 184. Babitt JL, Huang FW, Wrighting DM, et al. Bone morphogenetic
inflammation. J Clin Invest. 2002;110:1037–1044. protein signaling by hemojuvelin regulates hepcidin expression. Nat
164. Leung PS, Srai SK, Mascarenhas M, Churchill LJ, Debnam ES. Genet. 2006;38:531–539.
Increased duodenal iron uptake and transfer in a rat model of 185. Piperno A, Mariani R, Trombini P, Girelli D. Hepcidin modulation in
chronic hypoxia is accompanied by reduced hepcidin expression. human diseases: from research to clinic. World J Gastroenterol.
Gut. 2005;54:1391–1395. 2009;15:538–551.
165. Pantopoulos K, Porwal SK, Tartakoff A, Devireddy L. Mechanisms 186. Meynard D, Kautz L, Darnaud V, Canonne‐Hergaux F, Coppin H,
of mammalian iron homeostasis. Biochemistry. 2012;51:5705–5724. Roth MP. Lack of the bone morphogenetic protein BMP6 induces
166. Seitz HK, Stickel F. Risk factors and mechanisms of hepatocarcino- massive iron overload. Nat Genet. 2009;41:478–481.
genesis with special emphasis on alcohol and oxidative stress. Biol 187. Andriopoulos B Jr, Corradini E, Xia Y, et al. BMP6 is a key endoge-
Chem. 2006;387:349–360. nous regulator of hepcidin expression and iron metabolism. Nat
167. Albano E. Alcohol, oxidative stress and free radical damage. Proc Genet. 2009;41:482–487.
Nutr Soc. 2006;65:278–290. 188. Truksa J, Peng H, Lee P, Beutler E. Bone morphogenetic proteins
168. Nahon P, Nuraldeen R, Rufat P, Sutton A, Trautwein C, Strnad P. In 2, 4, and 9 stimulate murine hepcidin 1 expression independently
alcoholic cirrhosis, low‐serum hepcidin levels associate with poor of Hfe, transferrin receptor 2 (Tfr2), and IL‐6. Proc Natl Acad Sci
long‐term survival. Liver Int. 2016;36:185–188. USA. 2006;103:10289–10293.
169. Adams PC. Nonexpressing homozygotes for C282Y hemochro- 189. Silvestri L, Pagani A, Nai A, De Domenico I, Kaplan J, Camaschella
matosis: minority or majority of cases? Mol Genet Metab. C. The serine protease matriptase‐2 (TMPRSS6) inhibits hepcidin
2000;71:81–86. activation by cleaving membrane hemojuvelin. Cell Metab.
170. Rochette J, Pointon JJ, Fisher CA, et al. Multicentric origin of 2008;8:502–511.
hemochromatosis gene (HFE) mutations. Am J Hum Genet. 190. Xia Y, Babitt JL, Sidis Y, Chung RT, Lin HY. Hemojuvelin regulates
1999;64:1056–1062. hepcidin expression via a selective subset of BMP ligands and
171. Gochee PA, Powell LW, Cullen DJ, Du Sart D, Rossi E, Olynyk JK. receptors independently of neogenin. Blood. 2008;111:5195–5204.
A population‐based study of the biochemical and clinical expression 191. De Domenico I, Zhang TY, Koening CL, et al. Hepcidin mediates
of the H63D hemochromatosis mutation. Gastroenterology. transcriptional changes that modulate acute cytokine‐induced
2002;122:646–651. inflammatory responses in mice. J Clin Invest. 2010;120:2395–
172. Gurrin LC, Bertalli NA, Dalton GW, et al; HealthIron Study I. HFE 2405.
C282Y/H63D compound heterozygotes are at low risk of 192. Zhao N, Zhang AS, Enns CA. Iron regulation by hepcidin. J Clin
hemochromatosis‐related morbidity. Hepatology. 2009;50:94–101. Invest. 2013;123:2337–2343.
173. Santos PC, Dinardo CL, Cancado RD, Schettert IT, Krieger JE, Per- 193. Lee P, Peng H, Gelbart T, Wang L, Beutler E. Regulation of hep-
eira AC. Non‐HFE hemochromatosis. Rev Bras Hematol Hemoter. cidin transcription by interleukin‐1 and interleukin‐6. Proc Natl Acad
2012;34:311–316. Sci USA. 2005;102:1906–1910.
174. Beutler E, Felitti VJ, Koziol JA, Ho NJ, Gelbart T. Penetrance of 194. Nemeth E, Rivera S, Gabayan V, et al. IL‐6 mediates hypoferremia
845G‐‐> A (C282Y) HFE hereditary haemochromatosis mutation in of inflammation by inducing the synthesis of the iron regulatory
the USA. Lancet. 2002;359:211–218. hormone hepcidin. J Clin Invest. 2004;113:1271–1276.
CZAJA | 19

195. Wrighting DM, Andrews NC. Interleukin‐6 induces hepcidin expres- 216. European Association for the Study of the L. EASL clinical prac-
sion through STAT3. Blood. 2006;108:3204–3209. tice guidelines for HFE hemochromatosis. J Hepatol. 2010;53:3–
196. Verga Falzacappa MV, Vujic Spasic M, Kessler R, Stolte J, Hentze 22.
MW, Muckenthaler MU. STAT3 mediates hepatic hepcidin 217. Bacon BR, Adams PC, Kowdley KV, Powell LW, Tavill AS. Diagnosis
expression and its inflammatory stimulation. Blood. and management of hemochromatosis: 2011 Practice Guideline by
2007;109:353–358. the American Association for the Study of Liver Diseases. Hepatol-
197. Pietrangelo A, Dierssen U, Valli L, et al. STAT3 is required for IL‐6‐ ogy. 2011;54:328–343.
gp130‐dependent activation of hepcidin in vivo. Gastroenterology. 218. Deter RL. The measurement of iron in rat liver tissue and subcellu-
2007;132:294–300. lar fractions using atomic absorption spectroscopy. Tex Rep Biol
198. Zhang K, Shen X, Wu J, et al. Endoplasmic reticulum stress acti- Med. 1975;33:407–413.
vates cleavage of CREBH to induce a systemic inflammatory 219. Olynyk JK, O'Neill R, Britton RS, Bacon BR. Determination of hep-
response. Cell. 2006;124:587–599. atic iron concentration in fresh and paraffin‐embedded tissue: diag-
199. Shin DY, Chung J, Joe Y, et al. Pretreatment with CO‐releasing nostic implications. Gastroenterology. 1994;106:674–677.
molecules suppresses hepcidin expression during inflammation and 220. Gandon Y, Olivie D, Guyader D, et al. Non‐invasive assessment of
endoplasmic reticulum stress through inhibition of the STAT3 and hepatic iron stores by MRI. Lancet. 2004;363:357–362.
CREBH pathways. Blood. 2012;119:2523–2532. 221. St Pierre TG, Clark PR, Chua‐Anusorn W. Measurement and map-
200. Zhang C, Wang G, Zheng Z, et al. Endoplasmic reticulum‐tethered ping of liver iron concentrations using magnetic resonance imaging.
transcription factor cAMP responsive element‐binding protein, hep- Ann N Y Acad Sci. 2005;1054:379–385.
atocyte specific, regulates hepatic lipogenesis, fatty acid oxidation, 222. Hankins JS, McCarville MB, Loeffler RB, et al. R2* magnetic reso-
and lipolysis upon metabolic stress in mice. Hepatology. nance imaging of the liver in patients with iron overload. Blood.
2012;55:1070–1082. 2009;113:4853–4855.
201. Vecchi C, Montosi G, Zhang K, et al. ER stress controls iron meta- 223. Joe E, Kim SH, Lee KB, et al. Feasibility and accuracy of dual‐
bolism through induction of hepcidin. Science. 2009;325:877–880. source dual‐energy CT for noninvasive determination of hepatic
202. Du X, She E, Gelbart T, et al. The serine protease TMPRSS6 is iron accumulation. Radiology. 2012;262:126–135.
required to sense iron deficiency. Science. 2008;320:1088–1092. 224. Mueller J, Raisi H, Rausch V, et al. Sensitive and non‐invasive
203. Lin L, Goldberg YP, Ganz T. Competitive regulation of hepcidin assessment of hepatocellular iron using a novel room‐temperature
mRNA by soluble and cell‐associated hemojuvelin. Blood. susceptometer. J Hepatol. 2017;67:535–542.
2005;106:2884–2889. 225. Tipton IH, Cook MJ. Trace elements in human tissue. II. Adult sub-
204. Camaschella C. Understanding iron homeostasis through genetic jects from the United States. Health Phys. 1963;9:103–145.
analysis of hemochromatosis and related disorders. Blood. 226. Brissot P, Ropert M, Le Lan C, Loreal O. Non‐transferrin bound
2005;106:3710–3717. iron: a key role in iron overload and iron toxicity. Biochim Biophys
205. Pietrangelo A. Hemochromatosis: an endocrine liver disease. Hepa- Acta. 2012;1820:403–410.
tology. 2007;46:1291–1301. 227. Koskenkorva‐Frank TS, Weiss G, Koppenol WH, Burckhardt S. The
206. Goodnough JB, Ramos E, Nemeth E, Ganz T. Inhibition of hepcidin complex interplay of iron metabolism, reactive oxygen species, and
transcription by growth factors. Hepatology. 2012;56:291–299. reactive nitrogen species: insights into the potential of various iron
207. Stoker M, Gherardi E, Perryman M, Gray J. Scatter factor is a therapies to induce oxidative and nitrosative stress. Free Radic Biol
fibroblast‐derived modulator of epithelial cell mobility. Nature. Med. 2013;65:1174–1194.
1987;327:239–242. 228. Koek GH, Liedorp PR, Bast A. The role of oxidative stress in non‐
208. Montesano R, Matsumoto K, Nakamura T, Orci L. Identification of alcoholic steatohepatitis. Clin Chim Acta. 2011;412:1297–1305.
a fibroblast‐derived epithelial morphogen as hepatocyte growth 229. Sumida Y, Niki E, Naito Y, Yoshikawa T. Involvement of free radi-
factor. Cell. 1991;67:901–908. cals and oxidative stress in NAFLD/NASH. Free Radic Res.
209. Sonnenberg E, Meyer D, Weidner KM, Birchmeier C. Scatter fac- 2013;47:869–880.
tor/hepatocyte growth factor and its receptor, the c‐met tyrosine 230. Ciftci A, Yilmaz B, Koklu S, et al. Serum levels of nitrate, nitrite and
kinase, can mediate a signal exchange between mesenchyme and advanced oxidation protein products (AOPP) in patients with nonal-
epithelia during mouse development. J Cell Biol. 1993;123:223– coholic fatty liver disease. Acta Gastroenterol Belg. 2015;78:201–
235. 205.
210. Molnarfi N, Benkhoucha M, Funakoshi H, Nakamura T, Lalive PH. 231. Galicia‐Moreno M, Gutierrez‐Reyes G. The role of oxidative stress
Hepatocyte growth factor: a regulator of inflammation and autoim- in the development of alcoholic liver disease. Rev Gastroenterol
munity. Autoimmun Rev. 2015;14:293–303. Mex. 2014;79:135–144.
211. Singhal E, Sen P. Hepatocyte growth factor‐induced c‐Src‐phos- 232. Louvet A, Mathurin P. Alcoholic liver disease: mechanisms of injury
phatidylinositol 3‐kinase‐AKT‐mammalian target of rapamycin path- and targeted treatment. Nat Rev Gastroenterol Hepatol.
way inhibits dendritic cell activation by blocking IkappaB kinase 2015;12:231–242.
activity. Int J Biochem Cell Biol. 2011;43:1134–1146. 233. Pemberton PW, Aboutwerat A, Smith A, Burrows PC, McMahon
212. Han CY, Koo JH, Kim SH, et al. Hepcidin inhibits Smad3 phospho- RF, Warnes TW. Oxidant stress in type I autoimmune hepatitis: the
rylation in hepatic stellate cells by impeding ferroportin‐mediated link between necroinflammation and fibrogenesis? Biochim Biophys
regulation of Akt. Nat Commun. 2016;7:13817. Acta. 2004;1689:182–189.
213. Folgueras AR, Freitas‐Rodriguez S, Ramsay AJ, et al. Matriptase‐2 234. Choi J, Ou JH. Mechanisms of liver injury. III. Oxidative stress in
deficiency protects from obesity by modulating iron homeostasis. the pathogenesis of hepatitis C virus. Am J Physiol Gastrointest Liver
Nat Commun. 2018;9:1350. Physiol. 2006;290:G847–851.
214. Adams PC, Passmore L, Chakrabarti S, et al. Liver diseases in the 235. Choi J, Corder NL, Koduru B, Wang Y. Oxidative stress and hepatic
hemochromatosis and iron overload screening study. Clin Gastroen- Nox proteins in chronic hepatitis C and hepatocellular carcinoma.
terol Hepatol. 2006;4:918–923. Free Radic Biol Med. 2014;72:267–284.
215. Whitfield JB, Zhu G, Heath AC, Powell LW, Martin NG. Effects of 236. Torti SV, Kwak EL, Miller SC, et al. The molecular cloning and char-
alcohol consumption on indices of iron stores and of iron stores on acterization of murine ferritin heavy chain, a tumor necrosis factor‐
alcohol intake markers. Alcohol Clin Exp Res. 2001;25:1037–1045. inducible gene. J Biol Chem. 1988;263:12638–12644.
20 | CZAJA

237. Miller LL, Miller SC, Torti SV, Tsuji Y, Torti FM. Iron‐independent 257. Wu YJ, Noguchi CT. Activation of globin gene expression by cDNAs
induction of ferritin H chain by tumor necrosis factor. Proc Natl from induced K562 cells. Evidence for involvement of ferritin in globin
Acad Sci USA. 1991;88:4946–4950. gene expression. J Biol Chem. 1991;266:17566–17572.
238. Smirnov IM, Bailey K, Flowers CH, Garrigues NW, Wesselius LJ. 258. Adams PC, Barton JC, Guo H, Alter D, Speechley M. Serum ferritin
Effects of TNF‐alpha and IL‐1beta on iron metabolism by A549 is a biomarker for liver mortality in the Hemochromatosis and Iron
cells and influence on cytotoxicity. Am J Physiol. 1999;277:L257– Overload Screening Study. Ann Hepatol. 2015;14:348–353.
263. 259. Walker NM, Stuart KA, Ryan RJ, et al. Serum ferritin concentration
239. Cairo G, Tacchini L, Pogliaghi G, Anzon E, Tomasi A, Bernelli‐Zaz- predicts mortality in patients awaiting liver transplantation. Hepatol-
zera A. Induction of ferritin synthesis by oxidative stress. Transcrip- ogy. 2010;51:1683–1691.
tional and post‐transcriptional regulation by expansion of the "free" 260. Bresgen N, Ohlenschlager I, Fiedler B, et al. Ferritin–a mediator of
iron pool. J Biol Chem. 1995;270:700–703. apoptosis? J Cell Physiol. 2007;212:157–164.
240. Kwak EL, Larochelle DA, Beaumont C, Torti SV, Torti FM. Role for 261. Ruddell RG, Hoang‐Le D, Barwood JM, et al. Ferritin functions as a
NF‐kappa B in the regulation of ferritin H by tumor necrosis fac- proinflammatory cytokine via iron‐independent protein kinase C
tor‐alpha. J Biol Chem. 1995;270:15285–15293. zeta/nuclear factor kappaB‐regulated signaling in rat hepatic stellate
241. Jang MK, Choi MS, Park YB. Regulation of ferritin light chain gene cells. Hepatology. 2009;49:887–900.
expression by oxidized low‐density lipoproteins in human monocytic 262. Weismuller TJ, Kirchner GI, Scherer MN, et al. Serum ferritin con-
THP‐1 cells. Biochem Biophys Res Commun. 1999;265:577–583. centration and transferrin saturation before liver transplantation
242. Tsuji Y, Ayaki H, Whitman SP, Morrow CS, Torti SV, Torti FM. predict decreased long‐term recipient survival. Hepatology.
Coordinate transcriptional and translational regulation of ferritin in 2011;54:2114–2124.
response to oxidative stress. Mol Cell Biol. 2000;20:5818–5827. 263. Facchini FS, Hua NW, Stoohs RA. Effect of iron depletion in carbo-
243. Orino K, Lehman L, Tsuji Y, Ayaki H, Torti SV, Torti FM. Ferritin and hydrate‐intolerant patients with clinical evidence of nonalcoholic
the response to oxidative stress. Biochem J. 2001;357:241–247. fatty liver disease. Gastroenterology. 2002;122:931–939.
244. Pham CG, Bubici C, Zazzeroni F, et al. Ferritin heavy chain upregu- 264. Valenti L, Fracanzani AL, Dongiovanni P, et al. Iron depletion by
lation by NF‐kappaB inhibits TNFalpha‐induced apoptosis by sup- phlebotomy improves insulin resistance in patients with nonalco-
pressing reactive oxygen species. Cell. 2004;119:529–542. holic fatty liver disease and hyperferritinemia: evidence from a
245. Broxmeyer HE, Bognacki J, Dorner MH, de Sousa M. Identification case‐control study. Am J Gastroenterol. 2007;102:1251–1258.
of leukemia‐associated inhibitory activity as acidic isoferritins. A 265. Beaton MD, Chakrabarti S, Levstik M, Speechley M, Marotta P,
regulatory role for acidic isoferritins in the production of granulo- Adams P. Phase II clinical trial of phlebotomy for non‐alcoholic
cytes and macrophages. J Exp Med. 1981;153:1426–1444. fatty liver disease. Aliment Pharmacol Ther. 2013;37:720–729.
246. Broxmeyer HE, Williams DE, Geissler K, et al. Suppressive effects 266. Yano M, Hayashi H, Yoshioka K, et al. A significant reduction in
in vivo of purified recombinant human H‐subunit (acidic) ferritin on serum alanine aminotransferase levels after 3‐month iron reduction
murine myelopoiesis. Blood. 1989;73:74–79. therapy for chronic hepatitis C: a multicenter, prospective, random-
247. Broxmeyer HE, Cooper S, Levi S, Arosio P. Mutated recombinant ized, controlled trial in Japan. J Gastroenterol. 2004;39:570–574.
human heavy‐chain ferritins and myelosuppression in vitro and 267. Kato J, Miyanishi K, Kobune M, et al. Long‐term phlebotomy with low‐
in vivo: a link between ferritin ferroxidase activity and biological iron diet therapy lowers risk of development of hepatocellular carci-
function. Proc Natl Acad Sci USA. 1991;88:770–774. noma from chronic hepatitis C. J Gastroenterol. 2007;42:830–836.
248. Levi S, Luzzago A, Cesareni G, et al. Mechanism of ferritin iron 268. Desai TK, Jamil LH, Balasubramaniam M, Koff R, Bonkovsky HL.
uptake: activity of the H‐chain and deletion mapping of the ferro‐ Phlebotomy improves therapeutic response to interferon in
oxidase site. A study of iron uptake and ferro‐oxidase activity of patients with chronic hepatitis C: a meta‐analysis of six prospective
human liver, recombinant H‐chain ferritins, and of two H‐chain randomized controlled trials. Dig Dis Sci. 2008;53:815–822.
deletion mutants. J Biol Chem. 1988;263:18086–18092. 269. Dongiovanni P, Valenti L, Ludovica Fracanzani A, Gatti S, Cairo G,
249. Cozzi A, Santambrogio P, Levi S, Arosio P. Iron detoxifying activity Fargion S. Iron depletion by deferoxamine up‐regulates glucose
of ferritin. Effects of H and L human apoferritins on lipid peroxida- uptake and insulin signaling in hepatoma cells and in rat liver. Am J
tion in vitro. FEBS Lett. 1990;277:119–122. Pathol. 2008;172:738–747.
250. Epsztejn S, Glickstein H, Picard V, et al. H‐ferritin subunit overex- 270. Poggiali E, Cassinerio E, Zanaboni L, Cappellini MD. An update on
pression in erythroid cells reduces the oxidative stress response iron chelation therapy. Blood Transfus. 2012;10:411–422.
and induces multidrug resistance properties. Blood. 1999;94:3593– 271. Ellervik C, Marott JL, Tybjaerg‐Hansen A, Schnohr P, Nordestgaard
3603. BG. Total and cause‐specific mortality by moderately and markedly
251. Cozzi A, Corsi B, Levi S, Santambrogio P, Albertini A, Arosio P. increased ferritin concentrations: general population study and
Overexpression of wild type and mutated human ferritin H‐chain in metaanalysis. Clin Chem. 2014;60:1419–1428.
HeLa cells: in vivo role of ferritin ferroxidase activity. J Biol Chem. 272. Davis WD Jr, Arrowsmith WR. The effect of repeated bleeding in
2000;275:25122–25129. hemochromatosis. J Lab Clin Med. 1950;36:814–815.
252. Cazzola M, Bergamaschi G, Dezza L, Arosio P. Manipulations of cel- 273. George DK, Powell LW. Review article: the screening, diagnosis
lular iron metabolism for modulating normal and malignant cell pro- and optimal management of haemochromatosis. Aliment Pharmacol
liferation: achievements and prospects. Blood. 1990;75:1903–1919. Ther. 1997;11:631–639.
253. Reif DW. Ferritin as a source of iron for oxidative damage. Free 274. Phatak P, Brissot P, Wurster M, et al. A phase 1/2, dose‐escalation trial
Radic Biol Med. 1992;12:417–427. of deferasirox for the treatment of iron overload in HFE‐related hered-
254. Balla G, Jacob HS, Balla J, et al. Ferritin: a cytoprotective antioxi- itary hemochromatosis. Hepatology. 2010;52:1671–1779.
dant strategem of endothelium. J Biol Chem. 1992;267:18148– 275. Adams P, Altes A, Brissot P, et al. Therapeutic recommendations in
18153. HFE hemochromatosis for p.Cys282Tyr (C282Y/C282Y) homozy-
255. Arosio P, Levi S. Ferritin, iron homeostasis, and oxidative damage. gous genotype. Hepatol Int. 2018;12:83–86.
Free Radic Biol Med. 2002;33:457–463. 276. Houglum K, Ramm GA, Crawford DH, Witztum JL, Powell LW,
256. Berberat PO, Katori M, Kaczmarek E, et al. Heavy chain ferritin Chojkier M. Excess iron induces hepatic oxidative stress and trans-
acts as an antiapoptotic gene that protects livers from ischemia forming growth factor beta1 in genetic hemochromatosis. Hepatol-
reperfusion injury. FASEB J. 2003;17:1724–1726. ogy. 1997;26:605–610.
CZAJA | 21

277. Valenti L, Moscatiello S, Vanni E, et al. Venesection for non‐alcoholic improves erythropoiesis and reduces iron overload in a mouse
fatty liver disease unresponsive to lifestyle counselling—a propen- model of beta‐thalassemia intermedia. Haematologica. 2016;101:
sity score‐adjusted observational study. QJM. 2011;104:141–149. e8–e11.
278. Katsarou A, Pantopoulos K. Hepcidin therapeutics. Pharmaceuticals 288. Corradini E, Garuti C, Montosi G, et al. Bone morphogenetic pro-
(Basel). 2018;11:127. tein signaling is impaired in an HFE knockout mouse model of
279. Preza GC, Ruchala P, Pinon R, et al. Minihepcidins are rationally hemochromatosis. Gastroenterology. 2009;137:1489–1497.
designed small peptides that mimic hepcidin activity in mice and 289. Corradini E, Schmidt PJ, Meynard D, et al. BMP6 treatment
may be useful for the treatment of iron overload. J Clin Invest. compensates for the molecular defect and ameliorates hemochro-
2011;121:4880–4888. matosis in Hfe knockout mice. Gastroenterology. 2010;139:1721–
280. Schmidt PJ, Toudjarska I, Sendamarai AK, et al. An RNAi therapeu- 1729.
tic targeting Tmprss6 decreases iron overload in Hfe(‐/‐) mice and 290. Gaun V, Patchen B, Volovetz J, et al. A chemical screen identifies
ameliorates anemia and iron overload in murine beta‐thalassemia small molecules that regulate hepcidin expression. Blood Cells Mol
intermedia. Blood. 2013;121:1200–1208. Dis. 2014;53:231–240.
281. Nemeth E, Preza GC, Jung CL, Kaplan J, Waring AJ, Ganz T. The 291. Li X, Rhee DK, Malhotra R, et al. Progesterone receptor membrane
N‐terminus of hepcidin is essential for its interaction with ferro- component‐1 regulates hepcidin biosynthesis. J Clin Invest.
portin: structure‐function study. Blood. 2006;107:328–333. 2016;126:389–401.
282. Casu C, Oikonomidou PR, Chen H, et al. Minihepcidin peptides as 292. Zhen AW, Nguyen NH, Gibert Y, et al. The small molecule, genis-
disease modifiers in mice affected by beta‐thalassemia and poly- tein, increases hepcidin expression in human hepatocytes. Hepatol-
cythemia vera. Blood. 2016;128:265–276. ogy. 2013;58:1315–1325.
283. Ramos E, Ruchala P, Goodnough JB, et al. Minihepcidins prevent 293. Czaja AJ. Epigenetic changes and their implications in autoimmune
iron overload in a hepcidin‐deficient mouse model of severe hepatitis. Eur J Clin Invest. 2018;48:e12899.
hemochromatosis. Blood. 2012;120:3829–3836. 294. Czaja AJ. Under‐evaluated or unassessed pathogenic pathways in
284. Park TJ, Lee YJ, Kim HJ, Park HG, Park WJ. Cloning and characteri- autoimmune hepatitis and implications for future management. Dig
zation of TMPRSS6, a novel type 2 transmembrane serine protease. Dis Sci. 2018;63:1706–1725.
Mol Cells. 2005;19:223–227.
285. Guo S, Casu C, Gardenghi S, et al. Reducing TMPRSS6 ameliorates
hemochromatosis and beta‐thalassemia in mice. J Clin Invest.
2013;123:1531–1541.
How to cite this article: Czaja AJ. Review article: iron
286. Schmidt PJ, Racie T, Westerman M, Fitzgerald K, Butler JS, Fleming
MD. Combination therapy with a Tmprss6 RNAi‐therapeutic and disturbances in chronic liver diseases other than
the oral iron chelator deferiprone additively diminishes secondary haemochromatosis – pathogenic, prognostic, and therapeutic
iron overload in a mouse model of beta‐thalassemia intermedia. Am implications. Aliment Pharmacol Ther. 2019;00:
J Hematol. 2015;90:310–313.
1–21. https://doi.org/10.1111/apt.15173
287. Casu C, Aghajan M, Oikonomidou PR, Guo S, Monia BP, Rivella S.
Combination of Tmprss6‐ ASO and the iron chelator deferiprone

You might also like