Skip to content
Publicly Available Published by De Gruyter August 7, 2023

Serum hepcidin levels in chronic liver disease: a systematic review and meta-analysis

  • Ruchi Sharma ORCID logo , Weidan Zhao , Yousaf Zafar , Arvind R. Murali and Kyle E. Brown EMAIL logo

Abstract

Objectives

Dysregulation of hepcidin-iron axis is presumed to account for abnormal iron status in patients with chronic liver disease (CLD). Our aim is to determine the effect of specific etiologies of CLD and of cirrhosis on serum hepcidin levels.

Methods

PubMed, Embase, Web of Science were searched for studies comparing serum hepcidin levels in patients with CLD to that in controls using enzyme-linked immunosorbent assay. The study was conducted in accordance with the Preferred Reporting Items for Systematic Review and Meta-Analysis Guidelines. Statistical analysis was carried out with STATA using random effects model to calculate the mean difference (MD) between two groups.

Results

Hepcidin levels were significantly lower in subjects with hepatitis C virus (16 studies) [MD −1.6 (95 % CI: −2.66 to −0.54), p<0.01] and alcoholic liver disease (3 studies) [MD −0.84 (95 % CI: −1.6 to −0.07), p=0.03] than controls. Serum hepcidin was significantly higher in subjects with non-alcoholic fatty liver disease (12 studies) [MD 0.62 (95 % CI: 0.21 to 1.03), p<0.01], but did not differ in subjects with hepatitis B and controls (eight studies) [MD −0.65 (95 % CI: −1.47 to 0.16), p=0.12]. Hepcidin levels were significantly lower in patients with cirrhosis of any etiology (four studies) [MD −1.02 (CI: −1.59 to −0.45), p<0.01] vs. controls (CI: confidence interval).

Conclusions

Serum hepcidin levels are altered in common forms of CLD albeit not in a consistent direction. Additional study is needed to determine how changes in hepcidin levels are related to dysregulation of iron metabolism in CLD.

Introduction

Hepcidin is a peptide hormone synthesized primarily in the liver. It was discovered simultaneously as a regulator of iron homeostasis in the body and an antimicrobial peptide that is excreted in the urine [1, 2]. Its discovery established a central role for the liver in the control of iron metabolism. Hepcidin exerts its regulatory effect by controlling the release of iron from macrophages, enterocytes, and hepatocytes. It does so by means of its interaction with ferroportin, a transmembrane protein that is the only known cellular iron exporter in the body. A number of mechanisms account for hepcidin’s effects on iron transit, including its ability to cause internalization and degradation of ferroportin, as well as binding to and occluding the central cavity of ferroportin, thereby preventing iron export [3, 4]. Elevated iron levels, blood transfusions, and iron supplements [5, 6] all stimulate increased hepcidin production, reducing levels of ferroportin, leading to sequestration of iron derived from erythrophagocytosis in macrophages and reduced transfer of dietary iron into the plasma [7]. Patients with TMPRSS6 variants exhibit elevated hepcidin levels [8]. Under conditions of high iron demand such as anemia or expanded erythropoiesis or erythropoiesis stimulating agents, or in patients with hemochromatosis, the opposite occurs: hepcidin levels decrease, ferroportin is stabilized and release of iron from macrophages and enterocytes to the systemic circulation is enhanced (Figure 1) [9, 10].

Figure 1: 
Overview of role of hepcidin in maintaining iron homeostasis. (A) Conditions with increased hepcidin resulting in degradation of ferroportin and intra-cellular sequestration of iron. (B) Conditions with reduced hepcidin resulting increased bioavailable iron.
Figure 1:

Overview of role of hepcidin in maintaining iron homeostasis. (A) Conditions with increased hepcidin resulting in degradation of ferroportin and intra-cellular sequestration of iron. (B) Conditions with reduced hepcidin resulting increased bioavailable iron.

Hepcidin is also a type II acute phase reactant. Consistent with this role, hepcidin gene expression is transcriptionally activated by inflammatory cytokines, the most important of which is interleukin-6 [11]. This results in functional iron deficiency despite adequate body iron stores. Hepcidin is thus the key mediator of anemia of inflammation. In addition to iron status and inflammation, factors such as hypoxia, increased levels of reactive oxygen species (ROS), and endoplasmic reticulum stress have been shown to modulate hepcidin expression in animal and cell culture models [12], [13], [14], [15]. Some of these factors are stimulatory while others are inhibitory, and the interplay between them is complex [16, 17]. In the context of a chronic pathological process, there may be multiple, opposing inputs with the potential to modulate hepcidin levels, rendering the ultimate outcome in terms of iron metabolism difficult to predict.

Dysregulation of iron metabolism in chronic liver disease (CLD) has been a subject of intense interest over the past several decades [12]. Abnormal iron status has been linked to accelerated progression of liver fibrosis [18, 19], increased risk of hepatocellular carcinoma (HCC) [20, 21] and was associated with decreased responsiveness to hepatitis C virus (HCV) treatment in the interferon era [22]. Numerous trials of iron reduction reported decreased aminotransferase levels in HCV and non-alcoholic fatty liver disease (NAFLD) [23], [24], [25], [26], [27]. A long-term trial of iron reduction in patients with cirrhosis due to HCV was associated with a lowered risk of HCC [28], while a meta-analysis of phlebotomy trials in HCV concluded that this intervention improved responses to interferon treatment [29]. These observations indicate that modulation of iron status may alter the course of CLD, supporting the hypothesis that there is a feedback loop between CLD and dysregulation of iron metabolism, with the latter driving the progression of CLD to cirrhosis and HCC. However, claims that hepcidin levels are affected by CLD remain unconfirmed, as studies that have compared serum hepcidin levels in patients with CLD to controls are fraught with discrepancies.

In an attempt to resolve these conflicting results, we carried out a meta-analysis and systematic review of studies comparing serum hepcidin levels in patients with CLD due to various etiologies including HCV, hepatitis B virus (HBV), NAFLD, and alcoholic liver disease (ALD) to healthy controls. In addition, we analyzed whether the presence of cirrhosis, irrespective of underlying etiology, is associated with altered serum hepcidin levels.

Materials and methods

Literature search

Search strategies were developed by the authors with the assistance of a health sciences librarian with expertise in systematic reviews. Comprehensive strategies, including both index and keyword methods were devised for the following databases: PubMed, Web of Science, and Embase, which were searched from inception to April 2022 using the terms “Hepatitis, Autoimmune” or “Non-alcoholic Fatty Liver Disease” or “Hepatitis, Chronic” or “Liver Diseases, Alcoholic” or “Liver Failure” or “Iron Metabolism Disorders” or “Liver Cirrhosis” and “Hepcidins” or “Antimicrobial Cationic Peptides” or “HAMP protein, human” (Figure 2). The full PubMed search strategy was adapted for the other databases. In addition, for included studies, a citing paper and reference list search was conducted using SCOPUS to capture any relevant records.

Figure 2: 
Database search strategies.
Figure 2:

Database search strategies.

Study selection and data extraction

Studies that compared serum hepcidin levels in patients with CLD due to etiologies other than hemochromatosis to those in healthy controls were included. For consistency, we included only studies which used enzyme-linked immunosorbent assay (ELISA) methodology to measure serum hepcidin levels. We also restricted our meta-analysis to studies that reported serum hepcidin values as nanogram/deciliter or units that could be converted to nanogram/deciliter (Figure 2). Most of the studies did not specify the form of hepcidin measured, i.e., bioactive form, hepcidin-25, or total serum hepcidin. We therefore included all studies that otherwise met our selection criteria. We excluded studies that used methods other than ELISA to measure serum hepcidin levels [30], studies of patients on hemodialysis, animal studies, studies with incomplete data points, non-English language articles, abstracts, review articles, letters to the editor, comments, and opinion papers (Figure 2). The study was conducted in accordance with the Preferred Reporting Items for Systematic Review and Meta-Analysis guidelines.

R.S. and Y.Z. independently reviewed the database and agreed on final articles to be included in the meta-analysis. Data were extracted using a standardized template and included the number of patients/controls and serum hepcidin values. Conflicts regarding study selection or data extraction were resolved by mutual discussion and final database was approved by both authors. Data were expressed as mean and standard deviations after being converted if necessary. In addition, study year, patient characteristics, and inclusion/exclusion criteria for individual studies were recorded.

Statistical analysis

Meta-analysis was performed to determine the pooled weighted mean difference of serum hepcidin in patients with CLD vs. controls. Meta-analysis was carried out and forest plots were generated using Stata Corp. Stat version 17 (College Station, TX) using the random effects model. p-Value<0.05 was considered statistically significant. Heterogeneity was calculated by means of a χ2 test (Cochran Q statistic) and quantified with the I2 statistic. If I2>50 %, then there was significant heterogeneity. Funnel plots were obtained to assess the risk of bias. Sensitivity analysis was done using the leave-one-out method on Stata.

Results

After removing duplicates, a total of 2,467 records were identified by the search strategy. Thirty-four studies met inclusion criteria comprising a total of 3,839 subjects which included 2,221 patients and 1,618 controls (Figure 3).

Figure 3: 
Prisma flow diagram for selection of studies.
Figure 3:

Prisma flow diagram for selection of studies.

Hepatitis C virus

Meta-analysis of 16 studies (1,322 subjects) concerning patients with HCV showed that serum hepcidin levels were significantly lower in HCV compared with controls with a mean difference of −1.6 (95 % CI: −2.66 to −0.54), p<0.01; I2=98.51 % (Figure 4) [31], [32], [33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46] (CI: confidence interval).

Figure 4: 
Meta-analysis of studies comparing serum hepcidin levels in patients with hepatitis C virus to controls.
Figure 4:

Meta-analysis of studies comparing serum hepcidin levels in patients with hepatitis C virus to controls.

Alcoholic liver disease

Meta-analysis of three studies (150 subjects) for ALD showed significantly lower serum hepcidin levels in patients with ALD compared to controls with a mean difference of −0.84 (95 % CI: −1.6 to −0.07), p=0.03; I2=76.87 % (Figure 5) [47], [48], [49].

Figure 5: 
Meta-analysis of studies comparing serum hepcidin levels in patients with alcoholic liver disease to controls.
Figure 5:

Meta-analysis of studies comparing serum hepcidin levels in patients with alcoholic liver disease to controls.

Non-alcoholic fatty liver disease

A total of 12 studies (1,875 subjects) were found for NAFLD and meta-analysis showed higher serum hepcidin levels in patients compared to controls with a mean difference of 0.62 (0.21–1.03), p<0.01; I2=92.44 % (Figure 6) [35, 38, 50], [51], [52], [53], [54], [55], [56], [57], [58], [59].

Figure 6: 
Meta-analysis of studies comparing serum hepcidin levels in patients with non-alcoholic fatty liver disease to controls.
Figure 6:

Meta-analysis of studies comparing serum hepcidin levels in patients with non-alcoholic fatty liver disease to controls.

Hepatitis B virus

For HBV, meta-analysis of eight studies (626 subjects) revealed that there was no difference in serum hepcidin levels between patients and controls with a mean difference of −0.65 (95 % CI: −1.47 to 0.16), p=0.12; I2=94.1 % (Figure 7) [35, 38, 48, 60], [61], [62], [63], [64].

Figure 7: 
Meta-analysis of studies comparing serum hepcidin levels in patients with hepatitis B virus to controls.
Figure 7:

Meta-analysis of studies comparing serum hepcidin levels in patients with hepatitis B virus to controls.

Cirrhosis irrespective of underlying etiology

We found four studies (287 subjects) that compared hepcidin levels in patients with cirrhosis to levels in controls. Three of these studies included patients with cirrhosis due to HBV and one with cirrhosis due to ALD. Serum hepcidin levels were significantly reduced in patients with cirrhosis compared to controls, mean difference of −1.02 (95 % CI: −1.59 to −0.45), p<0.01, I2=76.45 % (Figure 8) [49, 60], [61], [62].

Figure 8: 
Meta-analysis of studies comparing serum hepcidin levels in patients with cirrhosis of any etiology to controls.
Figure 8:

Meta-analysis of studies comparing serum hepcidin levels in patients with cirrhosis of any etiology to controls.

The results are summarized in Table 1. Data for study characteristics and quality assessment of studies are presented in Tables 2 and 3, respectively. Funnel plots were obtained to assess the risk of publication bias (Supplementary Figure 1). Sensitivity analysis confirmed the findings for all groups except for ALD, where exclusion of certain studies from the meta-analysis resulted in no significant difference in serum hepcidin levels between patients and healthy controls (Supplementary Figure 2).

Table 1:

Summary of results comparing serum hepcidin level in patients with liver disease with controls.

Comparison Number of studies Mean difference Confidence interval p-Value Heterogeneity
Hepatitis C virus vs. controls 16 −1.6 −2.66 to −0.54 <0.01 98.5 %
Alcoholic liver disease vs. controls 3 −0.84 −1.6 to −0.07 0.03 76.87 %
Non-alcoholic fatty liver disease vs. controls 12 0.62 0.21 to 1.03 <0.01 92.44 %
Hepatitis B virus vs. controls 8 −0.65 −1.47 to 0.16 0.12 94.1 %
Cirrhosis from any etiology vs. controls 4 −1.02 −1.59 to −0.45 <0.01 76.45 %
Table 2:

Overall characteristics of studies included for meta-analysis.

Study Year Country Subgroup n %Female Age, years
Study Control Study Control Study Control
Abbas et al. [31] 2020 Egypt HCV 20 20 55.00 35 48.45 49.4
Mohsen et al. [41] 2021 Iraq HCV 54 50
Ali et al. [42] 2020 Egypt HCV 50 20 40
Amer et al. [43] 2020 Egypt HCV 35 15 41–76 33–63
Auguet et al. [50] 2017 Spain NAFLD 46 49 100 100 48.3 43.9
Boga et al. [51] 2015 Turkey NAFLD 66 35 65.2 57.1 44.4 43
Cam et al. [62] 2020 Turkey HBV, cirrhosis 126 23 35.7 47.8 39 33
Darwich et al. [32] 2013 Spain HCV 31 52 32.2 36.4 56.3 49.8
Demircioglu et al. [58] 2014 Turkey NAFLD 30 30 36.7 56.7 12.1 12.7
Dostalikova-Cimburova et al. [47] 2014 Czech Republic ALD 16 8 57.38 57.4
Gao et al. [60] 2018 China HBV, cirrhosis 163 78 32.1 38.5 46 44
Giannini et al. [59] 2022 Italy NAFLD 18 33 55.6 36.4 9.7 6.3
Gill et al. [44] 2021 Pakistan HCV 22 22 0 0 50.35 50.35
Girelli et al. [33] 2009 Italy HCV 81 57 35.8 36.8 42.2 35
Hou et al. [63] 2019 China HBV 41 20 22 50 36.83 35.7
Jamali et al. [52] 2016 Iran NAFLD, cirrhosis 18 18 27.8 27.8 34.5 30.44
Lin et al. [61] 2013 China HBV, cirrhosis 30 19 5 10.5 50.37 49.74
Liu et al. [34] 2014 China HCV 129 84
Lyberopoulou et al. [35] 2015 Greece HCV, HBV, NAFLD 21 17 52.4 41.2 40.6 45.2
Maras et al. [49] 2015 India Cirrhosis 20 20 25 20 34 33
Marmur et al. [53] 2018 Sweden NAFLD 38 34 34.2 55.9 54.42 40
Mohamed et al. [36] 2019 Egypt HCV 50 50 52 54 10 9.5
Nanba et al. [45] 2016 Japan HCV 60 20 51.7 57
Pan et al. [54] 2019 China NAFLD 482 490 32 32.4 48 48
Rogalska-Taranta et al. [37] 2017 Poland HCV 19 15 21.1 46.7 44 40
Ryan et al. [38] 2018 United Kingdom HCV, HBV 81 20 30.9 60 50–55 58
Schwarz et al. [39] 2011 Germany HCV 17 39 0 43.6 38 36
Senates et al. [55] 2011 Turkey NAFLD 88 88 36.4 21.8 44 43
Tsochatzis et al. [40] 2010 Greece HCV 96 30 54.2 56.7 44 44
Uysal et al. [56] 2011 Turkey NAFLD 60 28 33.3 64.3 47.7 48.1
Varghese et al. [48] 2020 India HBV, ALD, cirrhosis 53 46 0 0 45.5–47.4 46.6
Vuppalanchi et al. [57] 2014 United States of America NAFLD, cirrhosis 44 60 88.6 23.3 47 51
Wahab et al. [46] 2020 Egypt HCV 45 15 53.3 46.7 40.7 36.2
Wang et al. [64] 2013 China HBV 71 13 38 38.5 40.7 36.3
  1. HCV, hepatitis C virus; ALD, alcoholic liver disease; NAFLD, non-alcoholic fatty liver disease; HBV, hepatitis B virus.

Table 3:

The Newcastle-Ottawa scale is used to assess the quality of case control studies. The scale assigns up to four points for comparability, two points for selection, and three points for outcome.

Study Type of study Blinding Selection Comparability Outcome Overall quality score
Abbas et al. [31] Case control N/A 3 2 3 8
Mohsen et al. [41] Case control N/A 3 3 6
Ali et al. [42] Case control N/A 3 1 3 7
Amer et al. [43] Case control N/A 2 3 5
Auguet et al. [50] Case control N/A 3 1 3 7
Boga et al. [51] Case control N/A 3 1 3 7
Cam et al. [62] Case control N/A 3 1 3 7
Darwich et al. [32] Case control N/A 3 1 3 7
Demircioglu et al. [58] Case control N/A 3 1 3 7
Dostalikova-Cimburova et al. [47] Case control N/A 3 1 3 7
Gao et al. [60] Case control N/A 3 1 3 7
Giannini et al. [59] Case control N/A 3 1 3 7
Gill et al. [44] Case control N/A 3 1 3 7
Girelli et al. [33] Case control N/A 3 1 3 7
Hou et al. [63] Case control N/A 3 1 3 7
Jamali et al. [52] Case control N/A 3 1 3 7
Lin et al. [61] Case control N/A 3 2 3 8
Liu et al. [34] Case control N/A 3 1 3 7
Lyberopoulou et al. [35] Case control N/A 3 1 3 7
Maras et al. [49] Case control N/A 3 1 3 7
Marmur et al. [53] Case control N/A 3 1 3 7
Mohamed et al. [36] Case control N/A 3 1 3 7
Nanba et al. [45] Case control N/A 3 3 6
Pan et al. [54] Case control N/A 3 2 3 8
Rogalska-Taranta et al. [37] Case control N/A 3 1 3 7
Ryan et al. [38] Case control N/A 3 1 3 7
Schwarz et al. [39] Case control N/A 2 3 5
Senates et al. [55] Case control N/A 3 1 3 7
Tsochatzis et al. [40] Case control N/A 3 1 3 7
Uysal et al. [56] Case control N/A 3 1 3 7
Varghese et al. [48] Case control N/A 3 1 3 7
Vuppalanchi et al. [57] Case control N/A 3 1 3 7
Wahab et al. [46] Case control N/A 3 1 3 7
Wang et al. [64] Case control N/A 3 1 3 7

We did not perform a meta-analysis of the two studies that compared hepcidin levels in patients with cirrhosis to patients with liver disease without cirrhosis due to the small number of studies.

Similarly, we did not carry out a meta-analysis on studies of patients with autoimmune hepatitis (AIH), primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC) as we found only two studies for patients with these conditions that met our inclusion criteria. Lyberopoulou et al. reported low serum hepcidin levels compared with controls for patients with AIH and PBC/PSC [35]. Radicheva et al. reported combined results for patients with AIH and PBC/PSC and found that serum hepcidin levels in patients were not significantly different from that in controls [65].

Discussion

Some patients with CLD demonstrate evidence of aberrant iron metabolism, which has been implicated in fibrosis progression and increased risk of HCC [18, 22, 66, 67]. Given the primacy of hepcidin in the control of iron metabolism, it is reasonable to hypothesize that the fundamental cause of altered iron metabolism in CLD is dysregulation of hepcidin expression. Many investigations have addressed this question by comparing hepcidin levels in patients with CLD to that in controls. Because of the considerable variability in the results, we performed a meta-analysis in an attempt to bring clarity to this topic.

Our results show that serum hepcidin levels are significantly lower in subjects with HCV compared to controls. This finding is consistent with research in cell culture and in transgenic mice demonstrating that hepcidin gene expression is suppressed by HCV [68, 69]. These approaches implicated virus-mediated increases in ROS production as the main factor driving reductions in hepcidin, with differing downstream mechanisms proposed to account for the downregulation of hepcidin expression by ROS, i.e., increased histone deacetylase activity in one model, and decreased DNA-binding activity of CCAT/enhancer binding proteinα (C/EBPα, the main transcription factor controlling hepcidin gene expression) in another [68, 69]. The concept that abnormalities of serum iron studies in HCV-infected individuals arise as a consequence of viral effects on hepcidin expression is consistent with the finding that those abnormalities normalize promptly with eradication of HCV infection [70].

Our meta-analysis found that serum hepcidin is also significantly reduced in patients with ALD, though sensitivity analysis did not confirm this finding. This result is broadly consistent with studies showing that ethanol down-regulates hepcidin expression in rodent models [71]. Similar to HCV, ethanol appears to modulate hepcidin expression via increased levels of ROS that constrain the transcriptional activity of C/EBPα [72].

By contrast, our results indicate that serum hepcidin levels in subjects with NAFLD are elevated compared to controls. The tendency for patients with NAFLD to have relatively higher levels of serum hepcidin than other forms of CLD can perhaps be understood in the context of the multiple factors involved in the pathogenesis of NAFLD. Obesity itself is associated with increased hepcidin levels, perhaps augmented by small amounts of hepcidin originating from adipose tissue [73], and the presence of metabolic syndrome and its components engender a chronic inflammatory state, further stimulating hepcidin expression. In contrast to these results, animal models of fatty liver have generally shown downregulation of hepcidin, even in models that are characterized by insulin resistance and inflammation, apparently due to lowered hepatic iron stores resulting from increased iron utilization or impairment of intestinal iron uptake, depending on the model [71]. However, with few exceptions, these experiments examined hepcidin mRNA, not serum hepcidin levels.

The findings above suggest that some forms of CLD affect hepcidin levels independent of the presence of cirrhosis and impaired hepatic synthetic function. The levels of many proteins synthesized by the liver are diminished in advanced liver disease, so it is reasonable to predict that the same might be true of hepcidin. Our finding of a significant difference between serum hepcidin levels in patients with cirrhosis and controls supports this prediction, but there are reasons to interpret these results cautiously. Three of the four studies examined only patients with cirrhosis due to HBV. Since we did not find a significant difference in serum hepcidin levels between subjects with HBV and controls, this would suggest that HBV patients develop low serum hepcidin levels only after the disease has progressed to cirrhosis. However, two of the studies found no statistically significant difference in serum hepcidin levels in HBV patients with or without cirrhosis [60, 62]. This raises concern that the results may be spurious, i.e., resulting from low study numbers or skewed data. Unfortunately, we did not find any studies on serum hepcidin levels in cirrhosis due to other etiologies, hence we are unable to comment on the effect of cirrhosis on serum hepcidin levels in general.

In extrapolating our results to predicted downstream effects on iron metabolism, it is important to recognize that much of our current knowledge concerning the regulation of hepcidin is based on measurements of hepcidin transcripts derived from animal models or cell culture. There has been relatively little investigation into the existence of post-transcriptional mechanisms that might modulate the abundance or activity of secreted hepcidin. However, there are reports of inconsistencies between hepatic mRNA levels, levels of hepcidin in the circulation or urine, and markers of iron status, suggesting that such mechanisms may exist. For example, in a study of patients with dysmetabolic iron overload, Barisani et al. found no correlation between urinary hepcidin measurements and hepcidin mRNA levels and also noted that hepatic iron concentrations were more strongly correlated with urinary hepcidin levels than with hepatic hepcidin transcripts [74]. More recently, Rametta and colleagues provided evidence for a state of mild hepcidin resistance in patients with this condition, based on comparisons of serum hepcidin concentrations and changes in transferrin saturation following a dose of oral iron [75]. These data suggest that a model of iron homeostasis that presumes the existence of a one-to-one correspondence between the abundance of hepatic hepcidin transcripts and the quantity and biological activity of the resulting peptide, generating predictable effects on iron absorption (and ultimately body iron stores) may be overly simplistic.

A few comments are needed regarding the very high levels of heterogeneity observed in our analysis. Several authors have emphasized the importance of understanding the sources of heterogeneity in meta-analyses [76, 77]. Some portion of the heterogeneity may be the result of clinical heterogeneity due to differences in the baseline characteristics of the patients included in some of these analyses. For example, some studies included patients with cirrhosis, some without, while others included a mix of both. Hepcidin levels are also known to be affected by fasting and circadian rhythms [78], and may be modulated by factors specific to liver disease, such as the proportion of patients with advanced liver disease in a given study population.

Another important source of heterogeneity may be the methodology by which hepcidin was quantitated. In order to facilitate comparisons, we limited our analysis to studies that used ELISA-based assays. However, not all of the studies used the same ELISA. In many cases, a commercially sourced assay was chosen, while others were developed in-house. This is noteworthy because the generation of antibodies to hepcidin poses particular challenges due to its small size, conserved amino acid sequence and intramolecular disulfide bonds [78]. Depending on the antibodies used in an ELISA, a given assay may measure not only the biologically active 25-amino acid peptide, but also the 22- and 20-amino acid isoforms of hepcidin, whose functions, if any, are unknown [79]. A large number of studies did not specify the form of hepcidin measured by their assay. Another concern is the fact that the majority of hepcidin in the circulation is bound to α2-macroglobulin; the extent to which the various ELISAs measure total vs. bound vs. unbound hepcidin is unclear (and the biological significance of the binding to α2-macroglobulin remains undefined) [80]. These factors may explain the fact that hepcidin levels (in both patients and controls) differed by as much as three orders of magnitude across the various studies. The technical challenges involved in hepcidin assays have been highlighted by an international group of experts, who reported that measurements of hepcidin performed on a panel of standard samples varied widely between methods, including different immunochemical methods [79]. Hence, methodological differences likely also contributed to the heterogeneity we observed and are a limitation of our study, despite the presence of healthy controls. The subjects also belonged to different demographics hence factors such as nutritional habits may influence serum hepcidin levels.

In conclusion, our meta-analysis indicates that hepcidin levels are altered in some common forms of CLD. However, the direction of the alteration is inconsistent, with decreased hepcidin levels observed in HCV and ALD and increased levels in NAFLD. Since disturbances in iron metabolism are frequently observed in patients with these conditions, further study is needed to establish how changes in hepcidin levels are related to dysregulation of iron metabolism in these forms of CLD. Studies using rigorously validated assays for hepcidin in well-characterized groups of patients with and without CLD will be essential to this effort.


Corresponding author: Kyle E. Brown, MD, Department of Internal Medicine, Division of Gastroenterology-Hepatology, University of Iowa Carver College of Medicine, 4553 JCP, 200 Hawkins Drive, Iowa City, IA 52242, USA; Iowa City Veterans Administration Medical Center, 601 US-6 W, Iowa City, IA 52246, USA; and Department of Radiation Oncology, Program in Free Radical and Radiation Biology, University of Iowa Carver College of Medicine, Iowa City, IA, USA, Phone: (319) 384 6579, Fax: (319) 356 7918, E-mail:

Acknowledgments

Jennifer Deberg, MLS, Hardin Library for the Health Sciences.

  1. Research ethics: The local Institutional Review Board deemed the study exempt from review.

  2. Informed consent: Not applicable.

  3. Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  4. Competing interests: Authors state no conflict of interest.

  5. Research funding: None declared.

References

1. Park, CH, Valore, EV, Waring, AJ, Ganz, T. Hepcidin, a urinary antimicrobial peptide synthesized in the liver. J Biol Chem 2001;276:7806–10. https://doi.org/10.1074/jbc.m008922200.Search in Google Scholar PubMed

2. Pigeon, C, Ilyin, G, Courselaud, B, Leroyer, P, Turlin, B, Brissot, P, et al.. A new mouse liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload. J Biol Chem 2001;276:7811–9. https://doi.org/10.1074/jbc.m008923200.Search in Google Scholar

3. Nemeth, E, Tuttle, MS, Powelson, J, Vaughn, MB, Donovan, A, Ward, DM, et al.. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 2004;306:2090–3. https://doi.org/10.1126/science.1104742.Search in Google Scholar PubMed

4. Aschemeyer, S, Qiao, B, Stefanova, D, Valore, EV, Sek, AC, Ruwe, TA, et al.. Structure-function analysis of ferroportin defines the binding site and an alternative mechanism of action of hepcidin. Blood 2018;131:899–910. https://doi.org/10.1182/blood-2017-05-786590.Search in Google Scholar PubMed PubMed Central

5. Origa, R, Galanello, R, Ganz, T, Giagu, N, Maccioni, L, Faa, G, et al.. Liver iron concentrations and urinary hepcidin in beta-thalassemia. Haematologica 2007;92:583–8. https://doi.org/10.3324/haematol.10842.Search in Google Scholar PubMed

6. Moretti, D, Goede, JS, Zeder, C, Jiskra, M, Chatzinakou, V, Tjalsma, H, et al.. Oral iron supplements increase hepcidin and decrease iron absorption from daily or twice-daily doses in iron-depleted young women. Blood 2015;126:1981–9. https://doi.org/10.1182/blood-2015-05-642223.Search in Google Scholar PubMed

7. Ganz, T. Hepcidin and iron regulation, 10 years later. Blood 2011;117:4425–33. https://doi.org/10.1182/blood-2011-01-258467.Search in Google Scholar PubMed PubMed Central

8. Nai, A, Pagani, A, Silvestri, L, Campostrini, N, Corbella, M, Girelli, D, et al.. TMPRSS6 rs855791 modulates hepcidin transcription in vitro and serum hepcidin levels in normal individuals. Blood 2011;118:4459–62. https://doi.org/10.1182/blood-2011-06-364034.Search in Google Scholar PubMed

9. Robach, P, Recalcati, S, Girelli, D, Campostrini, N, Kempf, T, Wollert, KC, et al.. Serum hepcidin levels and muscle iron proteins in humans injected with low- or high-dose erythropoietin. Eur J Haematol 2013;91:74–84. https://doi.org/10.1111/ejh.12122.Search in Google Scholar PubMed

10. Papanikolaou, G, Tzilianos, M, Christakis, JI, Bogdanos, D, Tsimirika, K, MacFarlane, J, et al.. Hepcidin in iron overload disorders. Blood 2005;105:4103–5. https://doi.org/10.1182/blood-2004-12-4844.Search in Google Scholar PubMed PubMed Central

11. Nemeth, E, Rivera, S, Gabayan, V, Keller, C, Taudorf, S, Pedersen, BK, et al.. IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J Clin Invest 2004;113:1271–6. https://doi.org/10.1172/jci200420945.Search in Google Scholar

12. Nicolas, G, Chauvet, C, Viatte, L, Danan, JL, Bigard, X, Devaux, I, et al.. The gene encoding the iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and inflammation. J Clin Invest 2002;110:1037–44. https://doi.org/10.1172/jci0215686.Search in Google Scholar

13. Choi, SO, Cho, YS, Kim, HL, Park, JW. ROS mediate the hypoxic repression of the hepcidin gene by inhibiting C/EBPalpha and STAT-3. Biochem Biophys Res Commun 2007;356:312–7. https://doi.org/10.1016/j.bbrc.2007.02.137.Search in Google Scholar PubMed

14. Liu, Q, Davidoff, O, Niss, K, Haase, VH. Hypoxia-inducible factor regulates hepcidin via erythropoietin-induced erythropoiesis. J Clin Invest 2012;122:4635–44. https://doi.org/10.1172/jci63924.Search in Google Scholar

15. Vecchi, C, Montosi, G, Zhang, K, Lamberti, I, Duncan, SA, Kaufman, RJ, et al.. ER stress controls iron metabolism through induction of hepcidin. Science 2009;325:877–80. https://doi.org/10.1126/science.1176639.Search in Google Scholar PubMed PubMed Central

16. Huang, H, Constante, M, Layoun, A, Santos, MM. Contribution of STAT3 and SMAD4 pathways to the regulation of hepcidin by opposing stimuli. Blood 2009;113:3593–9. https://doi.org/10.1182/blood-2008-08-173641.Search in Google Scholar PubMed PubMed Central

17. Darshan, D, Frazer, DM, Wilkins, SJ, Anderson, GJ. Severe iron deficiency blunts the response of the iron regulatory gene Hamp and pro-inflammatory cytokines to lipopolysaccharide. Haematologica 2010;95:1660–7. https://doi.org/10.3324/haematol.2010.022426.Search in Google Scholar PubMed PubMed Central

18. Beinker, NK, Voigt, MD, Arendse, M, Smit, J, Stander, IA, Kirsch, RE. Threshold effect of liver iron content on hepatic inflammation and fibrosis in hepatitis B and C. J Hepatol 1996;25:633–8. https://doi.org/10.1016/s0168-8278(96)80231-5.Search in Google Scholar PubMed

19. George, DK, Goldwurm, S, Macdonald, GA, Cowley, LL, Walker, NI, Ward, PJ, et al.. Increased hepatic iron concentration in nonalcoholic steatohepatitis is associated with increased fibrosis. Gastroenterology 1998;114:311–8. https://doi.org/10.1016/s0016-5085(98)70482-2.Search in Google Scholar PubMed

20. Ko, C, Siddaiah, N, Berger, J, Gish, R, Brandhagen, D, Sterling, RK, et al.. Prevalence of hepatic iron overload and association with hepatocellular cancer in end-stage liver disease: results from the National Hemochromatosis Transplant Registry. Liver Int 2007;27:1394–401. https://doi.org/10.1111/j.1478-3231.2007.01596.x.Search in Google Scholar PubMed

21. Kowdley, KV. Iron, hemochromatosis, and hepatocellular carcinoma. Gastroenterology 2004;127:S79–86. https://doi.org/10.1016/j.gastro.2004.09.019.Search in Google Scholar PubMed

22. Olynyk, JK, Reddy, KR, di Bisceglie, AM, Jeffers, LJ, Parker, TI, Radick, JL, et al.. Hepatic iron concentration as a predictor of response to interferon alfa therapy in chronic hepatitis C. Gastroenterology 1995;108:1104–9. https://doi.org/10.1016/0016-5085(95)90209-0.Search in Google Scholar PubMed

23. Piperno, A, Sampietro, M, D’Alba, R, Roffi, L, Fargion, S, Parma, S, et al.. Iron stores, response to alpha-interferon therapy, and effects of iron depletion in chronic hepatitis C. Liver 1996;16:248–54. https://doi.org/10.1111/j.1600-0676.1996.tb00737.x.Search in Google Scholar PubMed

24. Fong, TL, Han, SH, Tsai, NCS, Morgan, TR, Mizokami, M, Qianp, D, et al.. A pilot randomized, controlled trial of the effect of iron depletion on long-term response to α-interferon in patients with chronic hepatitis C. J Hepatol 1998;28:369–74. https://doi.org/10.1016/s0168-8278(98)80308-5.Search in Google Scholar PubMed

25. Di Bisceglie, AM, Bonkovsky, HL, Chopra, S, Flamm, S, Reddy, RK, Grace, N, et al.. Iron reduction as an adjuvant to interferon therapy in patients with chronic hepatitis C who have previously not responded to interferon: a multicenter, prospective, randomized, controlled trial. Hepatology 2000;32:135–8. https://doi.org/10.1053/jhep.2000.8700.Search in Google Scholar PubMed

26. Fontana, RJ, Israel, J, LeClair, P, Banner, BF, Tortorelli, K, Grace, N, et al.. Iron reduction before and during interferon therapy of chronic hepatitis C: results of a multicenter, randomized, controlled trial. Hepatology 2000;31:730–6. https://doi.org/10.1002/hep.510310325.Search in Google Scholar PubMed

27. Murali, AR, Gupta, A, Brown, K. Systematic review and meta-analysis to determine the impact of iron depletion in dysmetabolic iron overload syndrome and non-alcoholic fatty liver disease. Hepatol Res 2018;48:E30–41. https://doi.org/10.1111/hepr.12921.Search in Google Scholar PubMed

28. Kato, J, Miyanishi, K, Kobune, M, Nakamura, T, Takada, K, Takimoto, R, et al.. Long-term phlebotomy with low-iron diet therapy lowers risk of development of hepatocellular carcinoma from chronic hepatitis C. J Gastroenterol 2007;42:830–6. https://doi.org/10.1007/s00535-007-2095-z.Search in Google Scholar PubMed

29. Desai, TK, Jamil, LH, Balasubramaniam, M, Koff, R, Bonkovsky, HL. Phlebotomy improves therapeutic response to interferon in patients with chronic hepatitis C: a meta-analysis of six prospective randomized controlled trials. Dig Dis Sci 2008;53:815–22. https://doi.org/10.1007/s10620-007-9945-7.Search in Google Scholar PubMed

30. Tan, TC, Crawford, DH, Franklin, ME, Jaskowski, LA, Macdonald, GA, Jonsson, JR, et al.. The serum hepcidin:ferritin ratio is a potential biomarker for cirrhosis. Liver Int 2012;32:1391–9. https://doi.org/10.1111/j.1478-3231.2012.02828.x.Search in Google Scholar PubMed

31. Abbas, OM, Khalifa, KAE, Makhlouf, MM, Osman, NF, Abdel Razek, WM, Atta, AS. Influence of esophageal variceal bleeding on iron status in chronic hepatitis C patients. Eur J Gastroenterol Hepatol 2020;32:616–22. https://doi.org/10.1097/meg.0000000000001547.Search in Google Scholar

32. Darwich, E, To-Figueras, J, Molina-López, RA, Deulofeu, R, Olbina, G, Westerman, M, et al.. Increased serum hepcidin levels in patients with porphyria cutanea tarda. J Eur Acad Dermatol Venereol 2013;27:e68–74. https://doi.org/10.1111/j.1468-3083.2012.04511.x.Search in Google Scholar PubMed

33. Girelli, D, Pasino, M, Goodnough, JB, Nemeth, E, Guido, M, Castagna, A, et al.. Reduced serum hepcidin levels in patients with chronic hepatitis C. J Hepatol 2009;51:845–52. https://doi.org/10.1016/j.jhep.2009.06.027.Search in Google Scholar PubMed PubMed Central

34. Liu, Y, Lv, Q, Gao, J, Long, L, Duan, Z, Liang, H, et al.. Coinfection with HIV-1 alleviates iron accumulation in patients with chronic hepatitis C virus infection. PLoS One 2014;9:e98039. https://doi.org/10.1371/journal.pone.0098039.Search in Google Scholar PubMed PubMed Central

35. Lyberopoulou, A, Chachami, G, Gatselis, NK, Kyratzopoulou, E, Saitis, A, Gabeta, S, et al.. Low serum hepcidin in patients with autoimmune liver diseases. PLoS One 2015;10:e0135486. https://doi.org/10.1371/journal.pone.0135486.Search in Google Scholar PubMed PubMed Central

36. Mohamed, AA, Abd Almonaem, ER, Mansour, AI, Algebaly, HF, Khattab, RA, El Abd, YS. Importance of studying the levels of hepcidin and vitamin D in Egyptian children with chronic hepatitis C. J Transl Intern Med 2019;7:15–21. https://doi.org/10.2478/jtim-2019-0004.Search in Google Scholar PubMed PubMed Central

37. Rogalska-Taranta, M, Jaroszewicz, J, Flisiak, R. Normalizing serum hepcidin but not alpha-1-antitrypsin level during effective treatment of chronic hepatitis C. Clin Exp Hepatol 2017;3:203–8. https://doi.org/10.5114/ceh.2017.71573.Search in Google Scholar PubMed PubMed Central

38. Ryan, JD, Armitage, AE, Cobbold, JF, Banerjee, R, Borsani, O, Dongiovanni, P, et al.. Hepatic iron is the major determinant of serum ferritin in NAFLD patients. Liver Int 2018;38:164–73. https://doi.org/10.1111/liv.13513.Search in Google Scholar PubMed

39. Schwarz, P, Strnad, P, von Figura, G, Janetzko, A, Krayenbühl, P, Adler, G, et al.. A novel monoclonal antibody immunoassay for the detection of human serum hepcidin. J Gastroenterol 2011;46:648–56. https://doi.org/10.1007/s00535-010-0344-z.Search in Google Scholar PubMed

40. Tsochatzis, E, Papatheodoridis, GV, Koliaraki, V, Hadziyannis, E, Kafiri, G, Manesis, EK, et al.. Serum hepcidin levels are related to the severity of liver histological lesions in chronic hepatitis C. J Viral Hepat 2010;17:800–6. https://doi.org/10.1111/j.1365-2893.2009.01244.x.Search in Google Scholar PubMed

41. Mohsen Al-Abedy, NM, Salman, ED, Faraj, SA. Evaluation of serum hepcidin level in Iraqi patients with chronic hepatitis C and relation to serum iron status. Indian J Forensic Med Toxicol 2021;15:1425–30.10.37506/ijfmt.v15i3.15505Search in Google Scholar

42. Ali, M, Sayed, A, Elhawary, M, Assem, A. Serum hepcidin in patients with chronic hepatitis C and its relation to treatment with antiviral drugs. Al-Azhar Int Med J 2020;1:4. https://doi.org/10.21608/aimj.2020.28247.1201.Search in Google Scholar

43. Amer, A, Aboelmagd, Y, Sami, M. Study of hepcidin level in Egyptian HCV infected patients and its correlation to ferritin. Plant Arch 2020;20:2105–9.Search in Google Scholar

44. Gill, M, Sharafat, J, Ikram, F, Ul Qamar, M, Rehman, I, Saleem, M, et al.. Correlation between serum ferritin and hepcidin levels in chronic hepatitis C patients. Cureus 2021;13:e17484. https://doi.org/10.7759/cureus.17484.Search in Google Scholar PubMed PubMed Central

45. Nanba, S, Ikeda, F, Baba, N, Takaguchi, K, Senoh, T, Nagano, T, et al.. Association of hepatic oxidative stress and iron dysregulation with HCC development after interferon therapy in chronic hepatitis C. J Clin Pathol 2016;69:226–33. https://doi.org/10.1136/jclinpath-2015-203215.Search in Google Scholar PubMed

46. Wahab, SEA, Ibrahim, IM, El-Gebaly, AM, Sediq, AM, Youssef, AMF. Serum hepcidin level in different stages of hepatitis C induced chronic liver disease. Egypt J Hosp Med 2020;81:1634–8. https://doi.org/10.21608/ejhm.2020.116818.Search in Google Scholar

47. Dostalikova-Cimburova, M, Balusikova, K, Kratka, K, Chmelikova, J, Hejda, V, Hnanicek, J, et al.. Role of duodenal iron transporters and hepcidin in patients with alcoholic liver disease. J Cell Mol Med 2014;18:1840–50. https://doi.org/10.1111/jcmm.12310.Search in Google Scholar PubMed PubMed Central

48. Varghese, J, Varghese James, J, Karthikeyan, M, Rasalkar, K, Raghavan, R, Sukumaran, A, et al.. Iron homeostasis is dysregulated, but the iron-hepcidin axis is functional, in chronic liver disease. J Trace Elem Med Biol 2020;58:126442. https://doi.org/10.1016/j.jtemb.2019.126442.Search in Google Scholar PubMed

49. Maras, JS, Maiwall, R, Harsha, HC, Das, S, Hussain, MS, Kumar, C, et al.. Dysregulated iron homeostasis is strongly associated with multiorgan failure and early mortality in acute-on-chronic liver failure. Hepatology 2015;61:1306–20. https://doi.org/10.1002/hep.27636.Search in Google Scholar PubMed

50. Auguet, T, Aragonès, G, Berlanga, A, Martínez, S, Sabench, F, Binetti, J, et al.. Hepcidin in morbidly obese women with non-alcoholic fatty liver disease. PLoS One 2017;12:e0187065. https://doi.org/10.1371/journal.pone.0187065.Search in Google Scholar PubMed PubMed Central

51. Boga, S, Alkim, H, Alkim, C, Koksal, AR, Bayram, M, Yilmaz Ozguven, MB, et al.. The relationship of serum hemojuvelin and hepcidin levels with iron overload in nonalcoholic fatty liver disease. J Gastrointest Liver Dis 2015;24:293–300. https://doi.org/10.15403/jgld.2014.1121.243.hak.Search in Google Scholar PubMed

52. Jamali, R, Hatami, N, Kosari, F. The correlation between serum adipokines and liver cell damage in non-alcoholic fatty liver disease. Hepat Mon 2016;16:e37412. https://doi.org/10.5812/hepatmon.37412.Search in Google Scholar PubMed PubMed Central

53. Marmur, J, Beshara, S, Eggertsen, G, Onelöv, L, Albiin, N, Danielsson, O, et al.. Hepcidin levels correlate to liver iron content, but not steatohepatitis, in non-alcoholic fatty liver disease. BMC Gastroenterol 2018;18:78. https://doi.org/10.1186/s12876-018-0804-0.Search in Google Scholar PubMed PubMed Central

54. Pan, X, Chen, B, Liu, W, Li, Y, Hu, Z, Lin, X, et al.. Circulating iron levels interaction with central obesity on the risk of nonalcoholic fatty liver disease: a case-control study in Southeast China. Ann Nutr Metab 2019;74:207–14. https://doi.org/10.1159/000497228.Search in Google Scholar PubMed

55. Senates, E, Yilmaz, Y, Colak, Y, Ozturk, O, Altunoz, ME, Kurt, R, et al.. Serum levels of hepcidin in patients with biopsy-proven nonalcoholic fatty liver disease. Metab Syndr Relat Disord 2011;9:287–90. https://doi.org/10.1089/met.2010.0121.Search in Google Scholar PubMed

56. Uysal, S, Armutcu, F, Aydogan, T, Akin, K, Ikizek, M, Yigitoglu, MR. Some inflammatory cytokine levels, iron metabolism and oxidan stress markers in subjects with nonalcoholic steatohepatitis. Clin Biochem 2011;44:1375–9. https://doi.org/10.1016/j.clinbiochem.2011.09.017.Search in Google Scholar PubMed

57. Vuppalanchi, R, Troutt, JS, Konrad, RJ, Ghabril, M, Saxena, R, Bell, LN, et al.. Serum hepcidin levels are associated with obesity but not liver disease. Obesity (Silver Spring) 2014;22:836–41. https://doi.org/10.1002/oby.20403.Search in Google Scholar PubMed PubMed Central

58. Demircioglu, F, Gorunmez, G, Dagistan, E, Goksugur, SB, Bekdas, M, Tosun, M, et al.. Serum hepcidin levels and iron metabolism in obese children with and without fatty liver: case-control study. Eur J Pediatr 2014;173:947–51. https://doi.org/10.1007/s00431-014-2268-8.Search in Google Scholar PubMed

59. Giannini, C, Polidori, N, Saltarelli, MA, Chiarelli, F, Basilico, R, Mohn, A. Increased hepcidin levels and non-alcoholic fatty liver disease in obese prepubertal children: a further piece to the complex puzzle of metabolic derangements. J Pediatr Endocrinol Metab 2022;35:39–47. https://doi.org/10.1515/jpem-2021-0070.Search in Google Scholar PubMed

60. Gao, YH, Wang, JY, Liu, PY, Sun, J, Wang, XM, Wu, RH, et al.. Iron metabolism disorders in patients with hepatitis B-related liver diseases. World J Clin Cases 2018;6:600–10. https://doi.org/10.12998/wjcc.v6.i13.600.Search in Google Scholar PubMed PubMed Central

61. Lin, D, Ding, J, Liu, JY, He, YF, Dai, Z, Chen, CZ, et al.. Decreased serum hepcidin concentration correlates with brain iron deposition in patients with HBV-related cirrhosis. PLoS One 2013;8:e65551. https://doi.org/10.1371/journal.pone.0065551.Search in Google Scholar PubMed PubMed Central

62. Cam, H, Yilmaz, N. Serum hepcidin levels are related to serum markers for iron metabolism and fibrosis stage in patients with chronic hepatitis B: a cross-sectional study. Arab Journal Gastroenterol 2020;21:85–90. https://doi.org/10.1016/j.ajg.2020.04.013.Search in Google Scholar PubMed

63. Hou, SW, Wang, ZF, Guan, SH, Yang, K, Wang, Q, Chen, LW, et al.. Alterations in the iron homeostasis network of hepatocytes caused by hepatitis B virus. Clin Lab 2019;65. https://doi.org/10.7754/clin.lab.2018.180914.Search in Google Scholar

64. Wang, XH, Cheng, PP, Jiang, F, Jiao, XY. The effect of hepatitis B virus infection on hepcidin expression in hepatitis B patients. Ann Clin Lab Sci 2013;43:126–34.Search in Google Scholar

65. Penkova-Radicheva, MP, Gulubova, M, Ananiev, J, Ivanova, R, Mateva, L. Serum hepcidin levels in patients with chronic liver diseases. J Hepatol 2013;58:S546. https://doi.org/10.1016/j.jceh.2013.03.061.Search in Google Scholar

66. Fujita, N, Miyachi, H, Tanaka, H, Takeo, M, Nakagawa, N, Kobayashi, Y, et al.. Iron overload is associated with hepatic oxidative damage to DNA in nonalcoholic steatohepatitis. Cancer Epidemiol Biomarkers Prev 2009;18:424–32. https://doi.org/10.1158/1055-9965.epi-08-0725.Search in Google Scholar PubMed

67. Kew, MC. Hepatic iron overload and hepatocellular carcinoma. Liver Cancer 2014;3:31–40. https://doi.org/10.1159/000343856.Search in Google Scholar PubMed PubMed Central

68. Nishina, S, Hino, K, Korenaga, M, Vecchi, C, Pietrangelo, A, Mizukami, Y, et al.. Hepatitis C virus-induced reactive oxygen species raise hepatic iron level in mice by reducing hepcidin transcription. Gastroenterology 2008;134:226–38. https://doi.org/10.1053/j.gastro.2007.10.011.Search in Google Scholar PubMed

69. Miura, K, Taura, K, Kodama, Y, Schnabl, B, Brenner, DA. Hepatitis C virus-induced oxidative stress suppresses hepcidin expression through increased histone deacetylase activity. Hepatology 2008;48:1420–9. https://doi.org/10.1002/hep.22486.Search in Google Scholar PubMed

70. Hasan, Y, Brown, K. Viral eradication restores normal iron status in chronic hepatitis C patients with abnormal iron studies. Ann Hepatol 2020;19:422–6. https://doi.org/10.1016/j.aohep.2020.03.002.Search in Google Scholar PubMed

71. Bloomer, SA, Brown, KE. Hepcidin and iron metabolism in experimental liver injury. Am J Pathol 2021;191:1165–79. https://doi.org/10.1016/j.ajpath.2021.04.005.Search in Google Scholar PubMed

72. Harrison-Findik, DD, Schafer, D, Klein, E, Timchenko, NA, Kulaksiz, H, Clemens, D, et al.. Alcohol metabolism-mediated oxidative stress down-regulates hepcidin transcription and leads to increased duodenal iron transporter expression. J Biol Chem 2006;281:22974–82. https://doi.org/10.1074/jbc.m602098200.Search in Google Scholar

73. Bekri, S, Gual, P, Anty, R, Luciani, N, Dahman, M, Ramesh, B, et al.. Increased adipose tissue expression of hepcidin in severe obesity is independent from diabetes and NASH. Gastroenterology 2006;131:788–96. https://doi.org/10.1053/j.gastro.2006.07.007.Search in Google Scholar PubMed

74. Barisani, D, Pelucchi, S, Mariani, R, Galimberti, S, Trombini, P, Fumagalli, D, et al.. Hepcidin and iron-related gene expression in subjects with dysmetabolic hepatic iron overload. J Hepatol 2008;49:123–33. https://doi.org/10.1016/j.jhep.2008.03.011.Search in Google Scholar PubMed

75. Rametta, R, Dongiovanni, P, Pelusi, S, Francione, P, Iuculano, F, Borroni, V, et al.. Hepcidin resistance in dysmetabolic iron overload. Liver Int 2016;36:1540–8. https://doi.org/10.1111/liv.13124.Search in Google Scholar PubMed

76. Thompson, SG. Why sources of heterogeneity in meta-analysis should be investigated. BMJ 1994;309:1351–5. https://doi.org/10.1136/bmj.309.6965.1351.Search in Google Scholar PubMed PubMed Central

77. Sandercock, P. The authors say: ‘The data are not so robust because of heterogeneity’ – so, how should I deal with this systematic review? Meta-analysis and the clinician. Cerebrovasc Dis 2011;31:615–20. https://doi.org/10.1159/000326068.Search in Google Scholar PubMed

78. Girelli, D, Nemeth, E, Swinkels, DW. Hepcidin in the diagnosis of iron disorders. Blood 2016;127:2809–13. https://doi.org/10.1182/blood-2015-12-639112.Search in Google Scholar PubMed PubMed Central

79. Kroot, JJ, Kemna, EH, Bansal, SS, Busbridge, M, Campostrini, N, Girelli, D, et al.. Results of the first international round robin for the quantification of urinary and plasma hepcidin assays: need for standardization. Haematologica 2009;94:1748–52. https://doi.org/10.3324/haematol.2009.010322.Search in Google Scholar PubMed PubMed Central

80. Peslova, G, Petrak, J, Kuzelova, K, Hrdy, I, Halada, P, Kuchel, PW, et al.. Hepcidin, the hormone of iron metabolism, is bound specifically to alpha-2-macroglobulin in blood. Blood 2009;113:6225–36. https://doi.org/10.1182/blood-2009-01-201590.Search in Google Scholar PubMed


Supplementary Material

This article contains supplementary material (https://doi.org/10.1515/cclm-2023-0540).


Received: 2023-05-22
Accepted: 2023-07-25
Published Online: 2023-08-07
Published in Print: 2024-02-26

© 2023 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 9.5.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2023-0540/html
Scroll to top button