1
|
Khazaei M, Ardeshir RA. Protective effects of sulfated polysaccharides from Enteromorpha intestinalis on oxidative stress, liver iron overload and Ferroptosis in Zebra fish exposed to ethanol. Biomed Pharmacother 2024; 181:117715. [PMID: 39615168 DOI: 10.1016/j.biopha.2024.117715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
The study investigates the protective effects of sulfated polysaccharides extracted from Enteromorpha intestinalis (EIP) against oxidative stress, liver iron overload, and ferroptosis in zebrafish exposed to ethanol, a model for alcohol-related liver disease (ALD). The extracted polysaccharides were characterized for sulfate and sugar content, molecular weight, and functional groups. Adult male zebrafish were divided into three groups: control, ethanol-exposed (EE) (0.2 % ethanol (v/v) in the water), and ethanol-exposed with EIP supplementation (1 % EIP incorporated into the basal diet) (EE+EIP) for 30 days. The study measured liver oxidative stress indexes, serum enzymological indexes, liver and serum lipid profiles, liver iron ion content, and expression of ferroptosis-related genes. Histological analysis was conducted to assess lipid accumulation and iron deposition in liver tissues. The findings indicate that EIP supplementation significantly mitigates ethanol-induced liver damage. Specifically, EIP reduced malondialdehyde levels, increased antioxidant enzyme and non-enzymatic antioxidant activity, and decreased iron ion accumulation and the area of iron granules in the liver tissue. Additionally, EIP treatment lowered lipids levels and aminotransferase enzyme activity in the serum. In the ALD model, EIP inhibited ethanol-induced ferroptosis by modulating the expression of key genes: it decreased the expression of transferrin (tf), transferrin receptor (tfr), ferroportin (fpn), and ferritin heavy chain (fth), while increasing the expression of glutathione peroxidase 4 (gpx4) and solute carrier family 7 member 11 (slc7a11). EIP has protective effects against ethanol-induced liver injury in zebrafish, offering a foundation for further research into its hepatoprotective action and potential application in preventing and treating ALD.
Collapse
Affiliation(s)
- Marziyeh Khazaei
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| | - Rashid Alijani Ardeshir
- Marine Biotechnology Department, College of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran.
| |
Collapse
|
2
|
Ruivard M, Lobbes H. [Diagnosis and treatment of iron overload]. Rev Med Interne 2023; 44:656-661. [PMID: 37507250 DOI: 10.1016/j.revmed.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023]
Abstract
Etiological investigation of hyperferritinemia includes a full clinical examination, with the measurement of waist circumference, and simple biological tests including transferrin saturation. The classification between hyperferritinemia without iron overload (inflammation, excessive alcohol intake, cytolysis, L-ferritin mutation) or with iron overload is then relatively easy. Dysmetabolic iron overload syndrome is the most common iron overload disease and is defined by an unexplained serum ferritin level elevation associated with various metabolic syndrome criteria and mild hepatic iron content increase assessed by magnetic resonance imaging. Bloodlettings are often poorly tolerated without clear benefit. Type 1 genetic hemochromatosis (homozygous C282Y mutation on the HFE gene) leads to iron accumulation through an increase of dietary iron absorption due to hypohepcidinemia. More than 95% of hemochromatosis are type 1 hemochromatosis but the phenotypic expression is highly variable. Elastography is recommended to identify advanced hepatic fibrosis when serum ferritin exceeds 1000μg/L. Life expectancy is normal when bloodlettings are started early. Ferroportin gene mutation is an autosomal dominant disease with generally moderate iron overload. Chelators are used in iron overload associated with anaemia (myelodysplastic syndromes or transfusion-dependent thalassemia). Chelation is initiated when hepatic iron content exceeds 120μmol/g. Deferasirox is often used as first-line therapy, but deferiprone may be of interest despite haematological toxicity (neutropenia). Deferoxamine (parenteral route) is the treatment of choice for severe iron overload or emergency conditions.
Collapse
Affiliation(s)
- M Ruivard
- Service médecine Interne, CHU de Clermont-Ferrand, CHU d'Estaing, 63003 Clermont-Ferrand, France; UMR 6602 UCA/CNRS/SIGMA, thérapies guidées par l'image (TGI), université Clermont Auvergne, 63000 Clermont-Ferrand, France.
| | - H Lobbes
- Service médecine Interne, CHU de Clermont-Ferrand, CHU d'Estaing, 63003 Clermont-Ferrand, France; UMR 6602 UCA/CNRS/SIGMA, thérapies guidées par l'image (TGI), université Clermont Auvergne, 63000 Clermont-Ferrand, France.
| |
Collapse
|
3
|
Ali N, Ferrao K, Mehta KJ. Liver Iron Loading in Alcohol-Associated Liver Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1427-1439. [PMID: 36306827 DOI: 10.1016/j.ajpath.2022.08.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/15/2022] [Accepted: 08/31/2022] [Indexed: 02/04/2023]
Abstract
Alcohol-associated liver disease (ALD) is a common chronic liver disease with increasing incidence worldwide. Alcoholic liver steatosis/steatohepatitis can progress to liver fibrosis/cirrhosis, which can cause predisposition to hepatocellular carcinoma. ALD diagnosis and management are confounded by several challenges. Iron loading is a feature of ALD which can exacerbate alcohol-induced liver injury and promote ALD pathologic progression. Knowledge of the mechanisms that mediate liver iron loading can help identify cellular/molecular targets and thereby aid in designing adjunct diagnostic, prognostic, and therapeutic approaches for ALD. Herein, the cellular mechanisms underlying alcohol-induced liver iron loading are reviewed and how excess iron in patients with ALD can promote liver fibrosis and aggravate disease pathology is discussed. Alcohol-induced increase in hepatic transferrin receptor-1 expression and up-regulation of high iron protein in Kupffer cells (proposed) facilitate iron deposition and retention in the liver. Iron is loaded in both parenchymal and nonparenchymal liver cells. Iron-loaded liver can promote ferroptosis and thereby contribute to ALD pathology. Iron and alcohol can independently elevate oxidative stress. Therefore, a combination of excess iron and alcohol amplifies oxidative stress and accelerates liver injury. Excess iron-stimulated hepatocytes directly or indirectly (through Kupffer cell activation) activate the hepatic stellate cells via secretion of proinflammatory and profibrotic factors. Persistently activated hepatic stellate cells promote liver fibrosis, and thereby facilitate ALD progression.
Collapse
Affiliation(s)
- Najma Ali
- GKT School of Medical Education, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Kevin Ferrao
- GKT School of Medical Education, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Kosha J Mehta
- Centre for Education, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom.
| |
Collapse
|
4
|
Helfrich KK, Saini N, Kwan STC, Rivera OC, Mooney SM, Smith SM. Fetal anemia and elevated hepcidin in a mouse model of fetal alcohol spectrum disorder. Pediatr Res 2023; 94:503-511. [PMID: 36702950 PMCID: PMC11878275 DOI: 10.1038/s41390-023-02469-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 12/19/2022] [Accepted: 01/01/2023] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Prenatal alcohol exposure (PAE) impairs offspring growth and cognition, and this is worsened by concurrent iron deficiency. Alcohol disrupts fetal iron metabolism and produces functional iron deficiency, even when maternal iron status is adequate. We used a mouse model of moderate PAE to investigate the mechanisms underlying this dysregulated iron status. METHODS C57BL/6J female mice received 3 g/kg alcohol daily from embryonic day (E) 8.5-17.5 and were assessed at E17.5. RESULTS Alcohol reduced fetal hemoglobin, hematocrit, and red blood cell counts, despite elevated erythropoietin production. Alcohol suppressed maternal hepcidin expression and the upstream iron-sensing BMP/SMAD pathway, consistent with its effects in the nonpregnant state. In contrast, alcohol elevated fetal hepcidin, although this was not accompanied by an upregulation of the BMP/SMAD or proinflammatory IL-6/STAT3 pathways. Fetal expression of hepatic genes contributing to hemoglobin synthesis and iron metabolism were unaffected by alcohol, whereas those affecting ribosome biogenesis were suppressed, suggesting a novel candidate effector for this fetal anemia. CONCLUSION These data confirm and extend prior observations that PAE disrupts maternal and fetal iron metabolism and impairs the fetus's ability to regulate iron status. We propose this dysregulation increases gestational iron needs and represents a conserved response to PAE. IMPACT Prenatal alcohol exposure causes a functional iron deficiency in a model that also impairs cognition in later life. Prenatal alcohol exposure causes fetal anemia. This fetal anemia is accompanied by elevated hepcidin and erythropoietin. Findings are consistent with prior observations that prenatal alcohol exposure increases maternal-fetal iron requirements during pregnancy.
Collapse
Affiliation(s)
- Kaylee K Helfrich
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, 28081, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, 28081, USA
| | - Nipun Saini
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, 28081, USA
| | - Sze Ting Cecilia Kwan
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, 28081, USA
| | - Olivia C Rivera
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, 28081, USA
| | - Sandra M Mooney
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, 28081, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, 28081, USA
| | - Susan M Smith
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, 28081, USA.
- Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, 28081, USA.
| |
Collapse
|
5
|
Yehia A, Sousa RAL, Abulseoud OA. Sex difference in the association between blood alcohol concentration and serum ferritin. Front Psychiatry 2023; 14:1230406. [PMID: 37547205 PMCID: PMC10401063 DOI: 10.3389/fpsyt.2023.1230406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/07/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction The sex difference in alcohol use disorder (AUD) is ingrained in distinctive neurobiological responses between men and women, which necessitates further investigation for a more tailored management. Methods Minding the findings of iron dysregulation in AUD and the sex difference in iron homeostasis in multiple physiological and pathological settings, we examined the sex difference in the association between serum ferritin and blood alcohol concentration (BAC) in intoxicated males (n = 125) and females (n = 59). We included patients with both serum ferritin tested of any value and a BAC above the level of detection during the same hospital admission period. We investigated sex difference in the relationship between BAC, serum ferritin and liver enzymes in intoxicated critically ill and noncritically ill patients. Results We found a negative association between serum ferritin and BAC in critically ill, intoxicated females [R2 = 0.44, F(1,14) = 11.02, p = 0.005], with much attenuated serum ferritin in females compared to their male counterparts (194.5 ± 280.4 vs. 806.3 ± 3405.7 ng/L, p = 0.002). We found a positive association between serum ferritin and liver enzymes [alanine transaminase (ALT) and aspartate transferase (AST)] in critically ill intoxicated females [ALT: R2 = 0.48, F(1,10) = 9.1, p = 0.013; AST: R2 = 0.68, F(1,10) = 21.2, p = 0.001] and in noncritically ill intoxicated males [ALT: R2 = 0.1, F(1,83) = 9.4, p = 0.003; AST: R2 = 0.1, F(1,78) = 10.5, p = 0.002]. The effect of BAC on serum ferritin was not mediated by ALT [indirect effect: (B = 0.13, p = 0.1)]. We also found a significant effect of sex, anemia, intensive care unit (ICU) admission and mortality on serum ferritin. Discussion Our results suggest that high BAC in intoxicated female patients is associated with attenuated serum ferritin levels, questioning the role of low serum ferritin in female vulnerability to alcohol.
Collapse
Affiliation(s)
- Asmaa Yehia
- Department of Neuroscience, Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Phoenix, AZ, United States
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ricardo A L Sousa
- Department of Psychiatry and Psychology, Mayo Clinic Arizona, Phoenix, AZ, United States
| | - Osama A Abulseoud
- Department of Neuroscience, Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Phoenix, AZ, United States
- Department of Psychiatry and Psychology, Mayo Clinic Arizona, Phoenix, AZ, United States
| |
Collapse
|
6
|
Wang X, Zhou Y, Min J, Wang F. Zooming in and out of ferroptosis in human disease. Front Med 2023; 17:173-206. [PMID: 37121959 DOI: 10.1007/s11684-023-0992-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/12/2023] [Indexed: 05/02/2023]
Abstract
Ferroptosis is defined as an iron-dependent regulated form of cell death driven by lipid peroxidation. In the past decade, it has been implicated in the pathogenesis of various diseases that together involve almost every organ of the body, including various cancers, neurodegenerative diseases, cardiovascular diseases, lung diseases, liver diseases, kidney diseases, endocrine metabolic diseases, iron-overload-related diseases, orthopedic diseases and autoimmune diseases. Understanding the underlying molecular mechanisms of ferroptosis and its regulatory pathways could provide additional strategies for the management of these disease conditions. Indeed, there are an expanding number of studies suggesting that ferroptosis serves as a bona-fide target for the prevention and treatment of these diseases in relevant pre-clinical models. In this review, we summarize the progress in the research into ferroptosis and its regulatory mechanisms in human disease, while providing evidence in support of ferroptosis as a target for the treatment of these diseases. We also discuss our perspectives on the future directions in the targeting of ferroptosis in human disease.
Collapse
Affiliation(s)
- Xue Wang
- The Second Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ye Zhou
- Department of Endocrinology and Metabolism, Ningbo First Hospital, Ningbo, 315000, China
| | - Junxia Min
- The Second Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Fudi Wang
- The Second Affiliated Hospital, The First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
7
|
Zheng H, Yang F, Deng K, Wei J, Liu Z, Zheng YC, Xu H. Relationship between iron overload caused by abnormal hepcidin expression and liver disease: A review. Medicine (Baltimore) 2023; 102:e33225. [PMID: 36930080 PMCID: PMC10019217 DOI: 10.1097/md.0000000000033225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 02/16/2023] [Indexed: 03/18/2023] Open
Abstract
Iron is essential to organisms, the liver plays a vital role in its storage. Under pathological conditions, iron uptake by the intestine or hepatocytes increases, allowing excess iron to accumulate in liver cells. When the expression of hepcidin is abnormal, iron homeostasis in humans cannot be regulated, and resulting in iron overload. Hepcidin also regulates the release of iron from siderophores, thereby regulating the concentration of iron in plasma. Important factors related to hepcidin and systemic iron homeostasis include plasma iron concentration, body iron storage, infection, inflammation, and erythropoietin. This review summarizes the mechanism and regulation of iron overload caused by hepcidin, as well as related liver diseases caused by iron overload and treatment.
Collapse
Affiliation(s)
- Haoran Zheng
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Fan Yang
- Division of Liver Surgery, Department of Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Kaige Deng
- Division of Liver Surgery, Department of Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Jiaxin Wei
- Department of Emergency, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhenting Liu
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Yong-Chang Zheng
- Division of Liver Surgery, Department of Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Haifeng Xu
- Division of Liver Surgery, Department of Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
8
|
Kouroumalis E, Tsomidis I, Voumvouraki A. Iron as a therapeutic target in chronic liver disease. World J Gastroenterol 2023; 29:616-655. [PMID: 36742167 PMCID: PMC9896614 DOI: 10.3748/wjg.v29.i4.616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/03/2022] [Accepted: 12/31/2022] [Indexed: 01/20/2023] Open
Abstract
It was clearly realized more than 50 years ago that iron deposition in the liver may be a critical factor in the development and progression of liver disease. The recent clarification of ferroptosis as a specific form of regulated hepatocyte death different from apoptosis and the description of ferritinophagy as a specific variation of autophagy prompted detailed investigations on the association of iron and the liver. In this review, we will present a brief discussion of iron absorption and handling by the liver with emphasis on the role of liver macrophages and the significance of the iron regulators hepcidin, transferrin, and ferritin in iron homeostasis. The regulation of ferroptosis by endogenous and exogenous mod-ulators will be examined. Furthermore, the involvement of iron and ferroptosis in various liver diseases including alcoholic and non-alcoholic liver disease, chronic hepatitis B and C, liver fibrosis, and hepatocellular carcinoma (HCC) will be analyzed. Finally, experimental and clinical results following interventions to reduce iron deposition and the promising manipulation of ferroptosis will be presented. Most liver diseases will be benefited by ferroptosis inhibition using exogenous inhibitors with the notable exception of HCC, where induction of ferroptosis is the desired effect. Current evidence mostly stems from in vitro and in vivo experimental studies and the need for well-designed future clinical trials is warranted.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Liver Research Laboratory, University of Crete Medical School, Heraklion 71003, Greece
| | - Ioannis Tsomidis
- First Department of Internal Medicine, AHEPA University Hospital, Thessaloniki 54621, Greece
| | - Argyro Voumvouraki
- First Department of Internal Medicine, AHEPA University Hospital, Thessaloniki 54621, Greece
| |
Collapse
|
9
|
Hino K, Yanatori I, Hara Y, Nishina S. Iron and liver cancer: an inseparable connection. FEBS J 2022; 289:7810-7829. [PMID: 34543507 DOI: 10.1111/febs.16208] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/17/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023]
Abstract
Iron is an essential element for all organisms. Iron-containing proteins play critical roles in cellular functions. The biological importance of iron is largely attributable to its chemical properties as a transitional metal. However, an excess of 'free' reactive iron damages the macromolecular components of cells and cellular DNA through the production of harmful free radicals. On the contrary, most of the body's excess iron is stored in the liver. Not only hereditary haemochromatosis but also some liver diseases with mild-to-moderate hepatic iron accumulation, such as chronic hepatitis C, alcoholic liver disease and nonalcoholic steatohepatitis, are associated with a high risk for liver cancer development. These findings have attracted attention to the causative and promotive roles of iron in the development of liver cancer. In the last decade, accumulating evidence regarding molecules regulating iron metabolism or iron-related cell death programmes such as ferroptosis has shed light on the relationship between hepatic iron accumulation and hepatocarcinogenesis. In this review, we briefly present the current molecular understanding of iron regulation in the liver. Next, we describe the mechanisms underlying dysregulated iron metabolism depending on the aetiology of liver diseases. Finally, we discuss the causative and promotive roles of iron in cancer development.
Collapse
Affiliation(s)
- Keisuke Hino
- Department of Hepatology and Pancreatology, Kawasaki Medical School, Kurashiki, Japan
| | - Izumi Yanatori
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Japan
| | - Yuichi Hara
- Department of Hepatology and Pancreatology, Kawasaki Medical School, Kurashiki, Japan
| | - Sohji Nishina
- Department of Hepatology and Pancreatology, Kawasaki Medical School, Kurashiki, Japan
| |
Collapse
|
10
|
Xue M, Tian Y, Sui Y, Zhao H, Gao H, Liang H, Qiu X, Sun Z, Zhang Y, Qin Y. Protective effect of fucoidan against iron overload and ferroptosis-induced liver injury in rats exposed to alcohol. Biomed Pharmacother 2022; 153:113402. [DOI: 10.1016/j.biopha.2022.113402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/05/2022] [Accepted: 07/08/2022] [Indexed: 02/09/2023] Open
|
11
|
Chen H, Zhao W, Yan X, Huang T, Yang A. Overexpression of Hepcidin Alleviates Steatohepatitis and Fibrosis in a Diet-induced Nonalcoholic Steatohepatitis. J Clin Transl Hepatol 2022; 10:577-588. [PMID: 36062292 PMCID: PMC9396326 DOI: 10.14218/jcth.2021.00289] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS Iron overload can contribute to the progression of nonalcoholic fatty liver disease (NAFLD) to nonalcoholic steatohepatitis (NASH). Hepcidin (Hamp), which is primarily synthesized in hepatocytes, is a key regulator of iron metabolism. However, the role of Hamp in NASH remains unclear. Therefore, we aimed to elucidate the role of Hamp in the pathophysiology of NASH. METHODS Male mice were fed a choline-deficient L-amino acid-defined (CDAA) diet for 16 weeks to establish the mouse NASH model. A choline-supplemented amino acid-defined (CSAA) diet was used as the control diet. Recombinant adeno-associated virus genome 2 serotype 8 vector expressing Hamp (rAAV2/8-Hamp) or its negative control (rAAV2/8-NC) was administered intravenously at week 8 of either the CDAA or CSAA diet. RESULTS rAAV2/8-Hamp treatment markedly decreased liver weight and improved hepatic steatosis in the CDAA-fed mice, accompanied by changes in lipogenesis-related genes and adiponectin expression. Compared with the control group, rAAV2/8-Hamp therapy attenuated liver damage, with mice exhibiting reduced histological NAFLD inflammation and fibrosis, as well as lower levels of liver enzymes. Moreover, α-smooth muscle actin-positive activated hepatic stellate cells (HSCs) and CD68-postive macrophages increased in number in the CDAA-fed mice, which was reversed by rAAV2/8-Hamp treatment. Consistent with the in vivo findings, overexpression of Hamp increased adiponectin expression in hepatocytes and Hamp treatment inhibited HSC activation. CONCLUSIONS Overexpression of Hamp using rAAV2/8-Hamp robustly attenuated liver steatohepatitis, inflammation, and fibrosis in an animal model of NASH, suggesting a potential therapeutic role for Hamp.
Collapse
Affiliation(s)
- Hui Chen
- Digestive Department, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Correspondence to: Hui Chen, Digestive Department, Beijing Chaoyang Hospital, Capital Medical University, No. 5 Jingyuan Road, Shijingshan District, Beijing 100043, China. Tel: +86-10-51718484, Fax: +86-10-83165944, E-mail: . Aiting Yang, Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China. ORCID: https://orcid.org/0000-0002-5671-696X. Tel: +86-10-63139311, Fax: +86-10-83165944, E-mail:
| | - Wenshan Zhao
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Xuzhen Yan
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Tao Huang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Aiting Yang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center of Digestive Diseases, Beijing, China
- Beijing Clinical Medicine Institute, Beijing, China
- Correspondence to: Hui Chen, Digestive Department, Beijing Chaoyang Hospital, Capital Medical University, No. 5 Jingyuan Road, Shijingshan District, Beijing 100043, China. Tel: +86-10-51718484, Fax: +86-10-83165944, E-mail: . Aiting Yang, Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China. ORCID: https://orcid.org/0000-0002-5671-696X. Tel: +86-10-63139311, Fax: +86-10-83165944, E-mail:
| |
Collapse
|
12
|
Zhou X, Fu Y, Liu W, Mu Y, Zhang H, Chen J, Liu P. Ferroptosis in Chronic Liver Diseases: Opportunities and Challenges. Front Mol Biosci 2022; 9:928321. [PMID: 35720113 PMCID: PMC9205467 DOI: 10.3389/fmolb.2022.928321] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/16/2022] [Indexed: 01/01/2023] Open
Abstract
Ferroptosis, an iron-dependent non-apoptotic cell death characterized by lipid peroxidation, is a cell death pathway discovered in recent years. Ferroptosis plays an important role in tumors, ischemia-reperfusion injury, neurological diseases, blood diseases, etc. Recent studies have shown the importance of ferroptosis in chronic liver disease. This article summarizes the pathological mechanisms of ferroptosis involved in System Xc-, iron metabolism, lipid metabolism, and some GPX4-independent pathways, and the latest research on ferroptosis in chronic liver diseases such as alcoholic liver disease, non-alcoholic fatty liver disease, liver fibrosis, hepatocellular carcinoma. In addition, the current bottleneck issues that restrict the research on ferroptosis are proposed to provide ideas and strategies for exploring new therapeutic targets for chronic liver diseases.
Collapse
Affiliation(s)
- Xiaoxi Zhou
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Yadong Fu
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- Institute of Interdisciplinary Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Wei Liu
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Yongping Mu
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Hua Zhang
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Jiamei Chen
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Ping Liu
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- Institute of Interdisciplinary Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
13
|
Helfrich KK, Saini N, Kwan STC, Rivera OC, Hodges R, Smith SM. Gestational Iron Supplementation Improves Fetal Outcomes in a Rat Model of Prenatal Alcohol Exposure. Nutrients 2022; 14:nu14081653. [PMID: 35458215 PMCID: PMC9025692 DOI: 10.3390/nu14081653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/16/2022] [Accepted: 04/13/2022] [Indexed: 12/19/2022] Open
Abstract
Prenatal alcohol exposure causes neurodevelopmental disability and is associated with a functional iron deficiency in the fetus and neonate, even when the mother consumes an apparently iron-adequate diet. Here, we test whether gestational administration of the clinically relevant iron supplement Fer-In-Sol mitigates alcohol’s adverse impacts upon the fetus. Pregnant Long-Evans rats consumed an iron-adequate diet and received 5 g/kg alcohol by gavage for 7 days in late pregnancy. Concurrently, some mothers received 6 mg/kg oral iron. We measured maternal and fetal weights, hematology, tissue iron content, and oxidative damage on gestational day 20.5. Alcohol caused fetal anemia, decreased fetal body and brain weight, increased hepatic iron content, and modestly elevated hepatic malondialdehyde (p’s < 0.05). Supplemental iron normalized this brain weight reduction in alcohol-exposed males (p = 0.154) but not female littermates (p = 0.031). Iron also reversed the alcohol-induced fetal anemia and normalized both red blood cell numbers and hematocrit (p’s < 0.05). Iron had minimal adverse effects on the mother or fetus. These data show that gestational iron supplementation improves select fetal outcomes in prenatal alcohol exposure (PAE) including brain weight and hematology, suggesting that this may be a clinically feasible approach to improve prenatal iron status and fetal outcomes in alcohol-exposed pregnancies.
Collapse
Affiliation(s)
- Kaylee K. Helfrich
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; (K.K.H.); (S.T.C.K.); (O.C.R.); (R.H.)
- Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| | - Nipun Saini
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; (K.K.H.); (S.T.C.K.); (O.C.R.); (R.H.)
- Correspondence: (N.S.); (S.M.S.)
| | - Sze Ting Cecilia Kwan
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; (K.K.H.); (S.T.C.K.); (O.C.R.); (R.H.)
| | - Olivia C. Rivera
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; (K.K.H.); (S.T.C.K.); (O.C.R.); (R.H.)
| | - Rachel Hodges
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; (K.K.H.); (S.T.C.K.); (O.C.R.); (R.H.)
| | - Susan M. Smith
- UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA; (K.K.H.); (S.T.C.K.); (O.C.R.); (R.H.)
- Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
- Correspondence: (N.S.); (S.M.S.)
| |
Collapse
|
14
|
Li H, Hu L, Wang L, Wang Y, Shao M, Chen Y, Wu W, Wang L. Iron Activates cGAS-STING Signaling and Promotes Hepatic Inflammation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:2211-2220. [PMID: 35133148 DOI: 10.1021/acs.jafc.1c06681] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Iron deposition and chronic inflammation are associated with chronic liver diseases, such as alcoholic liver disease, nonalcoholic fatty liver disease, and chronic hepatitis B and C. However, the relationship between iron deposition and chronic inflammation in these diseases is still unclear. In the current study, we aimed to investigate the effect of iron on chronic inflammation in HepG2 cells and mice liver. We demonstrated that iron treatment enhanced the expression of cGAS, STING, and their downstream targets, including TBK1, IRF-3, and NF-κB in HepG2 cells and mice liver. We also found that treatment of HepG2 cells and mice with ferric ammonium citrate increased the expression of inflammatory cytokines, such as IFN-β. Finally, we found that genes involved in iron metabolism and the STING signaling pathway were up-regulated in liver cancer tissues, and the survival time of patients with high expression of these genes in tumor tissues was significantly shortened. These results suggest that iron overload may promote the progress of the chronic liver disease by activating cGAS-STING-mediated chronic inflammation, which provides a new idea for the development of drugs for the treatment of the chronic liver disease.
Collapse
Affiliation(s)
- Hailang Li
- Department of Pharmacy, Xiamen Medical College, Xiamen 361023, China
| | - Ling Hu
- Department of Biotechnology, Quanzhou Normal University, Quanzhou 362000, China
| | - Liwen Wang
- Department of Biotechnology, Quanzhou Normal University, Quanzhou 362000, China
| | - Yixuan Wang
- Department of Biotechnology, Quanzhou Normal University, Quanzhou 362000, China
| | - Meiqi Shao
- Department of Biotechnology, Quanzhou Normal University, Quanzhou 362000, China
| | - Yupei Chen
- Department of Pharmacy, Xiamen Medical College, Xiamen 361023, China
| | - Wenlin Wu
- Department of Biotechnology, Quanzhou Normal University, Quanzhou 362000, China
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou Normal University, Quanzhou 362000, China
| | - Lei Wang
- School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China
| |
Collapse
|
15
|
Chen Q, Gao M, Yang H, Mei L, Zhong R, Han P, Liu P, Zhao L, Wang J, Li J. Serum ferritin levels are associated with advanced liver fibrosis in treatment-naive autoimmune hepatitis. BMC Gastroenterol 2022; 22:23. [PMID: 35034629 PMCID: PMC8762965 DOI: 10.1186/s12876-022-02098-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/28/2021] [Indexed: 02/06/2023] Open
Abstract
Background and aim The association between iron-metabolism-related variables and liver fibrosis in chronic hepatitis C and nonalcoholic fatty liver disease is now well known. However, the relationship has not been extensively studied in autoimmune hepatitis (AIH). We aimed to investigate the association between variables associated with iron metabolism and advanced liver fibrosis among untreated patients with AIH. Methods Ninety-seven untreated AIH patients were enrolled in this cross-sectional study. All participants underwent iron metabolism index detection and liver biopsy. Multiple logistic regression analysis was used to explore the association of iron-metabolism-related variables with advanced liver fibrosis. Results Among the 97 AIH patients, 38 (39.2%) had advanced liver fibrosis, and 59 (60.8%) did not. In multivariate logistic regression analysis, immunoglobulin G (odds ratio [OR], 1.123; 95% confidence interval [CI] 1.023–1.232, P = 0.014), platelet count (OR 0.988; 95% CI 0.979–0.997, P = 0.013), prothrombin time (OR 1.758; 95% CI 1.143–2.704, P = 0.010) and ferritin (OR 1.002; 95% CI 1.001–1.004, P = 0.012) were independent risk factors for predicting advanced liver fibrosis in AIH patients. Conclusion Higher serum ferritin was independently associated with advanced liver fibrosis among patients with treatment-naive AIH.
Collapse
Affiliation(s)
- Qingling Chen
- Department of Hepatology, Second People's Clinical College of Tianjin Medical University, Tianjin, China.,Department of Hepatology, Tianjin Second People's Hospital, No. 7, Sudi South Road, Nankai District, Tianjin, 300192, China
| | - Min Gao
- Department of Hepatology, Tianjin Second People's Hospital, No. 7, Sudi South Road, Nankai District, Tianjin, 300192, China
| | - Hang Yang
- Department of Hepatology, Second People's Clinical College of Tianjin Medical University, Tianjin, China.,Department of Hepatology, Tianjin Second People's Hospital, No. 7, Sudi South Road, Nankai District, Tianjin, 300192, China
| | - Ling Mei
- Department of Hepatology, Second People's Clinical College of Tianjin Medical University, Tianjin, China.,Department of Hepatology, Tianjin Second People's Hospital, No. 7, Sudi South Road, Nankai District, Tianjin, 300192, China
| | - Rui Zhong
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ping Han
- Department of Hepatology, Tianjin Second People's Hospital, No. 7, Sudi South Road, Nankai District, Tianjin, 300192, China
| | - Peiyan Liu
- Department of Hepatology, Second People's Clinical College of Tianjin Medical University, Tianjin, China.,Department of Hepatology, Tianjin Second People's Hospital, No. 7, Sudi South Road, Nankai District, Tianjin, 300192, China
| | - Lili Zhao
- Department of Hepatology, Tianjin Second People's Hospital, No. 7, Sudi South Road, Nankai District, Tianjin, 300192, China
| | - Jing Wang
- Department of Hepatology, Tianjin Second People's Hospital, No. 7, Sudi South Road, Nankai District, Tianjin, 300192, China
| | - Jia Li
- Department of Hepatology, Tianjin Second People's Hospital, No. 7, Sudi South Road, Nankai District, Tianjin, 300192, China.
| |
Collapse
|
16
|
Ferrao K, Ali N, Mehta KJ. Iron and iron-related proteins in alcohol consumers: cellular and clinical aspects. J Mol Med (Berl) 2022; 100:1673-1689. [PMID: 36214835 PMCID: PMC9691479 DOI: 10.1007/s00109-022-02254-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 01/05/2023]
Abstract
Alcohol-associated liver disease (ALD) is one of the most common chronic liver diseases. Its pathological spectrum includes the overlapping stages of hepatic steatosis/steatohepatitis that can progress to liver fibrosis and cirrhosis; both are risk factors for hepatocellular carcinoma. Moreover, ALD diagnosis and management pose several challenges. The early pathological stages are reversible by alcohol abstinence, but these early stages are often asymptomatic, and currently, there is no specific laboratory biomarker or diagnostic test that can confirm ALD etiology. Alcohol consumers frequently show dysregulation of iron and iron-related proteins. Examination of iron-related parameters in this group may aid in early disease diagnosis and better prognosis and management. For this, a coherent overview of the status of iron and iron-related proteins in alcohol consumers is essential. Therefore, here, we collated and reviewed the alcohol-induced alterations in iron and iron-related proteins. Reported observations include unaltered, increased, or decreased levels of hemoglobin and serum iron, increments in intestinal iron absorption (facilitated via upregulations of duodenal divalent metal transporter-1 and ferroportin), serum ferritin and carbohydrate-deficient transferrin, decrements in serum hepcidin, decreased or unaltered levels of transferrin, increased or unaltered levels of transferrin saturation, and unaltered levels of soluble transferrin receptor. Laboratory values of iron and iron-related proteins in alcohol consumers are provided for reference. The causes and mechanisms underlying these alcohol-induced alterations in iron parameters and anemia in ALD are explained. Notably, alcohol consumption by hemochromatosis (iron overload) patients worsens disease severity due to the synergistic effects of excess iron and alcohol.
Collapse
Affiliation(s)
- Kevin Ferrao
- GKT School of Medical Education, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Najma Ali
- GKT School of Medical Education, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Kosha J Mehta
- Centre for Education, Faculty of Life Sciences and Medicine, King's College London, London, UK.
| |
Collapse
|
17
|
Association of Habitual Dietary Intake with Liver Iron-A Population-Based Imaging Study. Nutrients 2021; 14:nu14010132. [PMID: 35011009 PMCID: PMC8746950 DOI: 10.3390/nu14010132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 01/04/2023] Open
Abstract
Iron-related disorders of the liver can result in serious health conditions, such as liver cirrhosis. Evidence on the role of modifiable lifestyle factors like nutrition in liver iron storage is lacking. Thus, we aimed to assess the association of habitual diet with liver iron content (LIC). We investigated 303 participants from the population-based KORA-MRI study who underwent whole-body magnetic resonance imaging (MRI). Dietary habits were evaluated using repeated 24 h food lists and a food frequency questionnaire. Sex-stratified multiple linear regression models were applied to quantify the association between nutrition variables of interest and LIC, adjusting for liver fat content (LFC), energy intake, and age. Mean age of participants was 56.4 ± 9.0 years and 44.2% were female. Mean LIC was 1.23 ± 0.12 mg/g dry weight, with higher values in men than in women (1.26 ± 0.13 and 1.20 ± 0.10 mg/g, p < 0.001). Alcohol intake was positively associated with LIC (men: β = 1.94; women: β = 4.98, p-values < 0.03). Significant negative associations with LIC were found for fiber (β = −5.61, p < 0.001) and potassium (β = −0.058, p = 0.034) for female participants only. Furthermore, LIC was highly correlated with liver fat content in both sexes. Our findings suggests that there are sex-specific associations of habitual dietary intake and LIC. Alcohol, fiber, and potassium may play a considerable role in liver iron metabolism.
Collapse
|
18
|
Martín-González C, Pelazas-González R, Fernández-Rodríguez C, Alemán-Valls R, Martínez-Riera A, Ortega-Toledo P, García-Rodríguez A, Rodríguez-Gaspar M, González-Reimers E. Ferritin and liver fibrosis among patients with chronic hepatitis C virus infection. J Trace Elem Med Biol 2020; 61:126542. [PMID: 32417635 DOI: 10.1016/j.jtemb.2020.126542] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 03/29/2020] [Accepted: 04/27/2020] [Indexed: 12/25/2022]
Abstract
INTRODUCTION In chronic hepatitis C virus (HCV) infection there is increased iron absorption leading to iron overload, a fact that may promote ferritin synthesis. Theoretically, increased ferritin should promote ongoing liver fibrosis but disparate results have been described. OBJECTIVE We analyze the behavior of iron metabolism- related variables, comparing them with fibrosis and inflammatory activity in liver biopsy in HCV infected patients. PATIENTS AND METHODS We analyzed among 90 HCV patients subjected to liver biopsy prior to antiviral treatment the relationships of serum levels of iron, ferritin, transferrin, transferrin saturation index (TSI) and total iron binding capacity (TIBC) with liver fibrosis and histological severity, assessed by Metavir-f, Metavir-a and Knodell indices, as well as with liver function, and also compared the aforementioned iron metabolism- related variables with 34 controls. RESULTS Patients showed higher values of sideremia (T = 2.04; p = 0.044) and transferrin (T = 2.29; p = 0.004) compared with controls; but not ferritin, that was significantly higher among the 33 patients who also consumed alcohol (Z = 2.05; p = 0.041). Most patients showed a well preserved liver function (86 cases, Child A). Patients with Child B or C showed higher ferritin levels (Z = 2.68; p = 0.007) and TSI (Z = 2.41; p = 0.016), but lower transferrin and TIBC (Z = 3.25; p = 0.001) than Child A patients. Transferrin and TIBC were directly related to albumin (ρ = 0.24; p = 0.026), whereas bilirubin showed direct relationships with iron (ρ = 0.25; p = 0.016), TSI (ρ = 0.39; p < 0.001) and ferritin (ρ = 0.36; p < 0.001). Both ferritin (ρ = -0.22; p = 0.04) and TSI (ρ = -0.25; p = 0.016) were related to platelet count. No relationships were observed between iron variables and Knodell index, but serum iron, serum transferrin, and TSI were directly related to Metavir-f score (ρ = 0.28; p = 0.009, ρ = 0.22; p = 0.044, and ρ = 0.22; p = 0.044, in this order). CONCLUSION Alterations of iron related variables are relatively subtle in our series of 90 well compensated HCV patients. Serum ferritin was not related to liver fibrosis and increases only when alcoholism co-exists with HCV infection.
Collapse
Affiliation(s)
- Candelaria Martín-González
- Servicio de Medicina Interna. Hospital Universitario de Canarias. Universidad de La Laguna. Tenerife, Canary Islands, Spain.
| | - Ricardo Pelazas-González
- Servicio de Medicina Interna. Hospital Universitario de Canarias. Universidad de La Laguna. Tenerife, Canary Islands, Spain
| | - Camino Fernández-Rodríguez
- Servicio de Medicina Interna. Hospital Universitario de Canarias. Universidad de La Laguna. Tenerife, Canary Islands, Spain
| | - Remedios Alemán-Valls
- Servicio de Medicina Interna. Hospital Universitario de Canarias. Universidad de La Laguna. Tenerife, Canary Islands, Spain
| | - Antonio Martínez-Riera
- Servicio de Medicina Interna. Hospital Universitario de Canarias. Universidad de La Laguna. Tenerife, Canary Islands, Spain
| | - Paula Ortega-Toledo
- Servicio de Medicina Interna. Hospital Universitario de Canarias. Universidad de La Laguna. Tenerife, Canary Islands, Spain
| | - Alen García-Rodríguez
- Servicio de Medicina Interna. Hospital Universitario de Canarias. Universidad de La Laguna. Tenerife, Canary Islands, Spain
| | - Melchor Rodríguez-Gaspar
- Servicio de Medicina Interna. Hospital Universitario de Canarias. Universidad de La Laguna. Tenerife, Canary Islands, Spain.
| | - Emilio González-Reimers
- Servicio de Medicina Interna. Hospital Universitario de Canarias. Universidad de La Laguna. Tenerife, Canary Islands, Spain.
| |
Collapse
|
19
|
Macías-Rodríguez RU, Inzaugarat ME, Ruiz-Margáin A, Nelson LJ, Trautwein C, Cubero FJ. Reclassifying Hepatic Cell Death during Liver Damage: Ferroptosis-A Novel Form of Non-Apoptotic Cell Death? Int J Mol Sci 2020; 21:1651. [PMID: 32121273 PMCID: PMC7084577 DOI: 10.3390/ijms21051651] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/10/2020] [Accepted: 02/14/2020] [Indexed: 12/11/2022] Open
Abstract
Ferroptosis has emerged as a new type of cell death in different pathological conditions, including neurological and kidney diseases and, especially, in different types of cancer. The hallmark of this regulated cell death is the presence of iron-driven lipid peroxidation; the activation of key genes related to this process such as glutathione peroxidase-4 (gpx4), acyl-CoA synthetase long-chain family member-4 (acsl4), carbonyl reductase [NADPH] 3 (cbr3), and prostaglandin peroxidase synthase-2 (ptgs2); and morphological changes including shrunken and electron-dense mitochondria. Iron overload in the liver has long been recognized as both a major trigger of liver damage in different diseases, and it is also associated with liver fibrosis. New evidence suggests that ferroptosis might be a novel type of non-apoptotic cell death in several liver diseases including non-alcoholic steatohepatitis (NASH), alcoholic liver disease (ALD), drug-induced liver injury (DILI), viral hepatitis, and hemochromatosis. The interaction between iron-related lipid peroxidation, cellular stress signals, and antioxidant systems plays a pivotal role in the development of this novel type of cell death. In addition, integrated responses from lipidic mediators together with free iron from iron-containing enzymes are essential to understanding this process. The presence of ferroptosis and the exact mechanisms leading to this non-apoptotic type of cell death in the liver remain scarcely elucidated. Recognizing ferroptosis as a novel type of cell death in the liver could lead to the understanding of the complex interaction between different types of cell death, their role in progression of liver fibrosis, the development of new biomarkers, as well as the use of modulators of ferroptosis, allowing improved theranostic approaches in the clinic.
Collapse
Affiliation(s)
- Ricardo U. Macías-Rodríguez
- Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico;
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (M.E.I.); (C.T.)
- MICTLÁN-Network (Mechanisms of Liver Injury, Cell Death and Translational Nutrition in Liver Diseases Research Network), Mexico City 14080, Mexico
- Liver Fibrosis and Nutrition Lab (LFN Lab), Mexico City 14080, Mexico
| | - María Eugenia Inzaugarat
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (M.E.I.); (C.T.)
- MICTLÁN-Network (Mechanisms of Liver Injury, Cell Death and Translational Nutrition in Liver Diseases Research Network), Mexico City 14080, Mexico
| | - Astrid Ruiz-Margáin
- Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico;
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (M.E.I.); (C.T.)
- MICTLÁN-Network (Mechanisms of Liver Injury, Cell Death and Translational Nutrition in Liver Diseases Research Network), Mexico City 14080, Mexico
- Liver Fibrosis and Nutrition Lab (LFN Lab), Mexico City 14080, Mexico
| | - Leonard J. Nelson
- Institute for Bioengineering (IBioE), School of Engineering, Faraday Building, The University of Edinburgh, Edinburgh EH9 3 JL, UK;
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (M.E.I.); (C.T.)
| | - Francisco Javier Cubero
- MICTLÁN-Network (Mechanisms of Liver Injury, Cell Death and Translational Nutrition in Liver Diseases Research Network), Mexico City 14080, Mexico
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, 28040 Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), 28041 Madrid, Spain
| |
Collapse
|
20
|
Saini N, Helfrich KK, Kwan STC, Huebner SM, Abazi J, Flentke GR, Blohowiak SE, Kling PJ, Smith SM. Alcohol's Dysregulation of Maternal-Fetal IL-6 and p-STAT3 Is a Function of Maternal Iron Status. Alcohol Clin Exp Res 2019; 43:2332-2343. [PMID: 31524964 DOI: 10.1111/acer.14200] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/06/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Prenatal alcohol exposure (PAE) causes long-term growth and neurodevelopmental deficits that are worsened by maternal iron deficiency (ID). In our preclinical rat model, PAE causes fetal anemia, brain ID, and elevated hepatic iron via increased maternal and fetal hepcidin synthesis. These changes are normalized by a prenatal iron-fortified (IF) diet. Here, we hypothesize that iron status and PAE dysregulate the major upstream pathways that govern hepcidin production-EPO/BMP6/SMAD and IL-6/JAK2/STAT3. METHODS Pregnant, Long Evans rat dams consumed ID (2 to 6 ppm iron), iron-sufficient (IS, 100 ppm iron), or IF (500 ppm iron) diets and received alcohol (5 g/kg) or isocaloric maltodextrin daily from gestational days (GD) 13.5 to 19.5. Protein and gene expression were quantified in the 6 experimental groups at GD 20.5. RESULTS PAE did not affect Epo or Bmp6 expression, but reduced p-SMAD1/5/8/SMAD1/5/8 protein ratios in both IS and ID maternal and fetal liver (all p's < 0.01). In contrast, PAE stimulated maternal hepatic expression of Il-6 (p = 0.03) and elevated p-STAT3/STAT3 protein ratios in both IS and ID maternal and fetal liver (all p's < 0.02). PAE modestly elevated maternal Il-1β, Tnf-α, and Ifn-γ. Fetal cytokine responses to PAE were muted compared with dams, and PAE did not affect hepatic Il-6 (p = 0.78) in IS and ID fetuses. Dietary iron fortification sharply attenuated Il-6 expression in response to PAE, with IF driving a 150-fold decrease (p < 0.001) in maternal liver and a 10-fold decrease (p < 0.01) in fetal liver. The IF diet also normalized p-STAT3/STAT3 ratios in both maternal and fetal liver. CONCLUSIONS These findings suggest that alcohol-driven stimulation of the IL-6/JAK2/STAT3 pathway mediates the elevated hepcidin observed in the PAE dam and fetus. Normalization of these signals by IF suggests that dysregulated hepcidin is driven by alcohol's disruption of the IL-6/JAK2/STAT3 pathway. Prenatal dietary IF represents a potential therapeutic approach for PAE that warrants further investigation.
Collapse
Affiliation(s)
- Nipun Saini
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina
| | - Kaylee K Helfrich
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina
| | - Sze Ting Cecilia Kwan
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina
| | - Shane M Huebner
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Juna Abazi
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - George R Flentke
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina
| | - Sharon E Blohowiak
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Pamela J Kling
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Susan M Smith
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina
| |
Collapse
|
21
|
Wu X, Wang Y, Jia R, Fang F, Liu Y, Cui W. Computational and biological investigation of the soybean lecithin-gallic acid complex for ameliorating alcoholic liver disease in mice with iron overload. Food Funct 2019; 10:5203-5214. [PMID: 31380553 DOI: 10.1039/c9fo01022j] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Alcoholic liver disease (ALD) is associated with significant morbidity and mortality globally. In this study, the soybean lecithin-gallic acid complex was synthesized, and its physicochemical properties were evaluated, which confirmed the complex formation. Compared with the free state of the drug, gallic acid exhibited significantly different physicochemical properties after it was complexed with soybean lecithin. To clarify the binding mode between two monomers, computational investigation was performed. From the computational data, we deduced the structure of the compound and predicted that it has a high affinity for human phosphatidylcholine transfer protein and exhibits strong pharmacological activities in vivo. The complex not only effectively ameliorated liver fibrosis, lipid peroxidation, and oxidative stress, but also reduced liver iron overload in a mouse ALD model induced by alcohol (p < 0.05). Additionally, it regulated iron metabolism by inhibiting TfR1 expression (p < 0.05) and promoting hepcidin expression (p < 0.05). These results suggest that the soybean lecithin-gallic acid complex ameliorates hepatic damage and iron overload induced by alcohol and exert hepatoprotective effects.
Collapse
Affiliation(s)
- Xiangqun Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, P. R. China.
| | | | | | | | | | | |
Collapse
|
22
|
Czaja AJ. Letter: high ferritin to haemoglobin ratio is related to early mortality in patients with alcoholic hepatitis - author's reply. Aliment Pharmacol Ther 2019; 49:1457-1458. [PMID: 31074897 DOI: 10.1111/apt.15275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| |
Collapse
|
23
|
Czaja AJ. Review article: iron disturbances in chronic liver diseases other than haemochromatosis - pathogenic, prognostic, and therapeutic implications. Aliment Pharmacol Ther 2019; 49:681-701. [PMID: 30761559 DOI: 10.1111/apt.15173] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/08/2019] [Accepted: 01/16/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Disturbances in iron regulation have been described in diverse chronic liver diseases other than hereditary haemochromatosis, and iron toxicity may worsen liver injury and outcome. AIMS To describe manifestations and consequences of iron dysregulation in chronic liver diseases apart from hereditary haemochromatosis and to encourage investigations that clarify pathogenic mechanisms, define risk thresholds for iron toxicity, and direct management METHODS: English abstracts were identified in PubMed by multiple search terms. Full length articles were selected for review, and secondary and tertiary bibliographies 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 characterised most chronic liver diseases with the exception of non-alcoholic fatty liver disease, and the finding has been associated mainly with suppression of transcriptional activity of the hepcidin gene. Iron overload has been associated with oxidative 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.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| |
Collapse
|
24
|
Miyanishi K, Tanaka S, Sakamoto H, Kato J. The role of iron in hepatic inflammation and hepatocellular carcinoma. Free Radic Biol Med 2019; 133:200-205. [PMID: 30017991 DOI: 10.1016/j.freeradbiomed.2018.07.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 02/06/2023]
Abstract
Iron is an essential for organisms and the liver plays a major role in its storage. In pathologic conditions, where iron absorption from the intestine or iron uptake into the hepatocytes is increased, excess iron accumulates in the hepatocytes, leading to hepatocyte injury through the production of free radicals. Iron exerts its toxicity by catalyzing the generation of reactive oxygen species (ROS). ROS causes cell injury by inducing damage to the lysosomal, cytoplasmic, nuclear and mitochondrial membranes, apoptosis through activation of the caspase cascade, and hyperoxidation of fatty chains. In this manuscript, we reviewed the articles regarding role of iron in hepatic inflammation and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Koji Miyanishi
- Department of Medical Oncology, Sapporo Medical University, School of Medicine, South-1, West-16, Chuo-ku, Sapporo 060-8543, Japan.
| | - Shingo Tanaka
- Department of Medical Oncology, Sapporo Medical University, School of Medicine, South-1, West-16, Chuo-ku, Sapporo 060-8543, Japan; Department of Infection Control, and Laboratory Medicine, Sapporo Medical University, School of Medicine, Japan
| | - Hiroki Sakamoto
- Department of Medical Oncology, Sapporo Medical University, School of Medicine, South-1, West-16, Chuo-ku, Sapporo 060-8543, Japan
| | - Junji Kato
- Department of Medical Oncology, Sapporo Medical University, School of Medicine, South-1, West-16, Chuo-ku, Sapporo 060-8543, Japan
| |
Collapse
|
25
|
Mehta KJ, Farnaud SJ, Sharp PA. Iron and liver fibrosis: Mechanistic and clinical aspects. World J Gastroenterol 2019; 25:521-538. [PMID: 30774269 PMCID: PMC6371002 DOI: 10.3748/wjg.v25.i5.521] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/02/2019] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is characterised by excessive deposition of extracellular matrix that interrupts normal liver functionality. It is a pathological stage in several untreated chronic liver diseases such as the iron overload syndrome hereditary haemochromatosis, viral hepatitis, alcoholic liver disease, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis and diabetes. Interestingly, regardless of the aetiology, iron-loading is frequently observed in chronic liver diseases. Excess iron can feed the Fenton reaction to generate unquenchable amounts of free radicals that cause grave cellular and tissue damage and thereby contribute to fibrosis. Moreover, excess iron can induce fibrosis-promoting signals in the parenchymal and non-parenchymal cells, which accelerate disease progression and exacerbate liver pathology. Fibrosis regression is achievable following treatment, but if untreated or unsuccessful, it can progress to the irreversible cirrhotic stage leading to organ failure and hepatocellular carcinoma, where resection or transplantation remain the only curative options. Therefore, understanding the role of iron in liver fibrosis is extremely essential as it can help in formulating iron-related diagnostic, prognostic and treatment strategies. These can be implemented in isolation or in combination with the current approaches to prepone detection, and halt or decelerate fibrosis progression before it reaches the irreparable stage. Thus, this review narrates the role of iron in liver fibrosis. It examines the underlying mechanisms by which excess iron can facilitate fibrotic responses. It describes the role of iron in various clinical pathologies and lastly, highlights the significance and potential of iron-related proteins in the diagnosis and therapeutics of liver fibrosis.
Collapse
Affiliation(s)
- Kosha J Mehta
- School of Population Health and Environmental Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, United Kingdom
- Division of Human Sciences, School of Applied Sciences, London South Bank University, London SE1 0AA, United Kingdom
| | - Sebastien Je Farnaud
- Faculty Research Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry CV1 2DS, United Kingdom
| | - Paul A Sharp
- Department of Nutritional Sciences, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London SE1 9NH, United Kingdom
| |
Collapse
|
26
|
Brunacci F, Rocha VS, De Carli E, Espósito BP, Ruano R, Colli C. Increased serum iron in preeclamptic women is likely due to low hepcidin levels. Nutr Res 2018; 53:32-39. [DOI: 10.1016/j.nutres.2018.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 01/30/2018] [Accepted: 03/15/2018] [Indexed: 01/06/2023]
|
27
|
Vela D. Low hepcidin in liver fibrosis and cirrhosis; a tale of progressive disorder and a case for a new biochemical marker. Mol Med 2018; 24:5. [PMID: 30134796 PMCID: PMC6016890 DOI: 10.1186/s10020-018-0008-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/13/2018] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is a precursor of liver cirrhosis, which is associated with increased mortality. Though liver biopsy remains the gold standard for the diagnosis of fibrosis, noninvasive biochemical methods are cost-effective, practical and are not linked with major risks of complications. In this respect, serum hepcidin, has emerged as a new marker of fibrosis and cirrhosis. In this review the discussion uncovers molecular links between hepcidin disturbance and liver fibrosis/cirrhosis. The discussion also expands on clinical studies that suggest that hepcidin can potentially be used as a biochemical parameter of fibrosis/cirrhosis and target of therapeutic strategies to treat liver diseases. The debatable issues such as the complicated nature of hepcidin disturbance in non-alcoholic liver disease, serum levels of hepcidin in acute hepatitis C virus infection, cause of hepcidin disturbance in autoimmune hepatitis and hepatic insulin resistance are discussed, with potential solutions unveiled in order to be studied by future research.
Collapse
Affiliation(s)
- Driton Vela
- Department of Physiology, Faculty of Medicine, University of Prishtina, Martyr's Boulevard n.n, Prishtina, 10000, Kosovo.
| |
Collapse
|
28
|
Collins JF, Flores SR, Wang X, Anderson GJ. Mechanisms and Regulation of Intestinal Iron Transport. PHYSIOLOGY OF THE GASTROINTESTINAL TRACT 2018:1451-1483. [DOI: 10.1016/b978-0-12-809954-4.00060-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
29
|
Does Hypoxia Cause Carcinogenic Iron Accumulation in Alcoholic Liver Disease (ALD)? Cancers (Basel) 2017; 9:cancers9110145. [PMID: 29068390 PMCID: PMC5704163 DOI: 10.3390/cancers9110145] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 10/19/2017] [Accepted: 10/20/2017] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease (ALD) is a leading health risk worldwide. Hepatic iron overload is frequently observed in ALD patients and it is an important and independent factor for disease progression, survival, and the development of primary liver cancer (HCC). At a systemic level, iron homeostasis is controlled by the liver-secreted hormone hepcidin. Hepcidin regulation is complex and still not completely understood. It is modulated by many pathophysiological conditions associated with ALD, such as inflammation, anemia, oxidative stress/H2O2, or hypoxia. Namely, the data on hypoxia-signaling of hepcidin are conflicting, which seems to be mainly due to interpretational limitations of in vivo data and methodological challenges. Hence, it is often overlooked that hepcidin-secreting hepatocytes are physiologically exposed to 2–7% oxygen, and that key oxygen species such as H2O2 act as signaling messengers in such a hypoxic environment. Indeed, with the recently introduced glucose oxidase/catalase (GOX/CAT) system it has been possible to independently study hypoxia and H2O2 signaling. First preliminary data indicate that hypoxia enhances H2O2-mediated induction of hepcidin, pointing towards oxidases such as NADPH oxidase 4 (NOX4). We here review and discuss novel concepts of hypoxia signaling that could help to better understand hepcidin-associated iron overload in ALD.
Collapse
|
30
|
Mehta KJ, Farnaud S, Patel VB. HFE mRNA expression is responsive to intracellular and extracellular iron loading: short communication. Mol Biol Rep 2017; 44:399-403. [PMID: 28840425 PMCID: PMC5640751 DOI: 10.1007/s11033-017-4123-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 08/19/2017] [Indexed: 11/28/2022]
Abstract
In liver hepatocytes, the HFE gene regulates cellular and systemic iron homeostasis by modulating cellular iron-uptake and producing the iron-hormone hepcidin in response to systemic iron elevation. However, the mechanism of iron-sensing in hepatocytes remain enigmatic. Therefore, to study the effect of iron on HFE and hepcidin (HAMP) expressions under distinct extracellular and intracellular iron-loading, we examined the effect of holotransferrin treatment (1, 2, 5 and 8 g/L for 6 h) on intracellular iron levels, and mRNA expressions of HFE and HAMP in wild-type HepG2 and previously characterized iron-loaded recombinant-TfR1 HepG2 cells. Gene expression was analyzed by real-time PCR and intracellular iron was measured by ferrozine assay. Data showed that in the wild-type cells, where intracellular iron content remained unchanged, HFE expression remained unaltered at low holotransferrin treatments but was upregulated upon 5 g/L (p < 0.04) and 8 g/L (p = 0.05) treatments. HAMP expression showed alternating elevations and increased upon 1 g/L (p < 0.05) and 5 g/L (p < 0.05). However, in the recombinant cells that showed higher intracellular iron levels than wild-type cells, HFE and HAMP expressions were elevated only at low 1 g/L treatment (p < 0.03) and were repressed at 2 g/L treatment (p < 0.03). Under holotransferrin-untreated conditions, the iron-loaded recombinant cells showed higher expressions of HFE (p < 0.03) and HAMP (p = 0.05) than wild-type cells. HFE mRNA was independently elevated by extracellular and intracellular iron-excess. Thus, it may be involved in sensing both, extracellular and intracellular iron. Repression of HAMP expression under simultaneous intracellular and extracellular iron-loading resembles non-hereditary iron-excess pathologies.
Collapse
Affiliation(s)
- Kosha J Mehta
- Department of Biomedical Sciences, University of Westminster, 115 New Cavendish Street, London, W1W 6UW, UK
| | - Sebastien Farnaud
- School of Life Sciences, Coventry University, 138 James Starley Building, Coventry, CV1 5FB, UK
| | - Vinood B Patel
- Department of Biomedical Sciences, University of Westminster, 115 New Cavendish Street, London, W1W 6UW, UK.
| |
Collapse
|
31
|
Agarwal R, Narayan J, Bhattacharyya A, Saraswat M, Tomar AK. Gene expression profiling, pathway analysis and subtype classification reveal molecular heterogeneity in hepatocellular carcinoma and suggest subtype specific therapeutic targets. Cancer Genet 2017; 216-217:37-51. [PMID: 29025594 DOI: 10.1016/j.cancergen.2017.06.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/12/2017] [Accepted: 06/30/2017] [Indexed: 02/07/2023]
Abstract
A very low 5-year survival rate among hepatocellular carcinoma (HCC) patients is mainly due to lack of early stage diagnosis, distant metastasis and high risk of postoperative recurrence. Hence ascertaining novel biomarkers for early diagnosis and patient specific therapeutics is crucial and urgent. Here, we have performed a comprehensive analysis of the expression data of 423 HCC patients (373 tumors and 50 controls) downloaded from The Cancer Genome Atlas (TCGA) followed by pathway enrichment by gene ontology annotations, subtype classification and overall survival analysis. The differential gene expression analysis using non-parametric Wilcoxon test revealed a total of 479 up-regulated and 91 down-regulated genes in HCC compared to controls. The list of top differentially expressed genes mainly consists of tumor/cancer associated genes, such as AFP, THBS4, LCN2, GPC3, NUF2, etc. The genes over-expressed in HCC were mainly associated with cell cycle pathways. In total, 59 kinases associated genes were found over-expressed in HCC, including TTK, MELK, BUB1, NEK2, BUB1B, AURKB, PLK1, CDK1, PKMYT1, PBK, etc. Overall four distinct HCC subtypes were predicted using consensus clustering method. Each subtype was unique in terms of gene expression, pathway enrichment and median survival. Conclusively, this study has exposed a number of interesting genes which can be exploited in future as potential markers of HCC, diagnostic as well as prognostic and subtype classification may guide for improved and specific therapy.
Collapse
Affiliation(s)
- Rahul Agarwal
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, 110029 India
| | - Jitendra Narayan
- Unité de recherche en biologie environnementale et évolutive (URBE), University of Namur, Belgium
| | | | - Mayank Saraswat
- Transplantation Laboratory, Haartmaninkatu 3, University of Helsinki, Helsinki, Finland
| | - Anil Kumar Tomar
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India.
| |
Collapse
|
32
|
Mehta K, Busbridge M, Renshaw D, Evans RW, Farnaud S, Patel VB. Characterization of hepcidin response to holotransferrin in novel recombinant TfR1 HepG2 cells. Blood Cells Mol Dis 2016; 61:37-45. [PMID: 27667164 DOI: 10.1016/j.bcmd.2016.06.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 06/28/2016] [Accepted: 06/28/2016] [Indexed: 12/14/2022]
Abstract
Hepcidin is the key regulator of systemic iron homeostasis. The iron-sensing mechanisms and the role of intracellular iron in modulating hepatic hepcidin secretion are unclear. Therefore, we created a novel cell line, recombinant-TfR1 HepG2, expressing iron-response-element-independent TFRC mRNA to promote cellular iron-overload and examined the effect of excess holotransferrin (5g/L) on cell-surface TfR1, iron content, hepcidin secretion and mRNA expressions of TFRC, HAMP, SLC40A1, HFE and TFR2. Results showed that the recombinant cells exceeded levels of cell-surface TfR1 in wild-type cells under basal (2.8-fold; p<0.03) and holotransferrin-supplemented conditions for 24h and 48h (4.4- and 7.5-fold, respectively; p<0.01). Also, these cells showed higher intracellular iron content than wild-type cells under basal (3-fold; p<0.03) and holotransferrin-supplemented conditions (6.6-fold at 4h; p<0.01). However, hepcidin secretion was not higher than wild-type cells. Moreover, holotransferrin treatment to recombinant cells did not elevate HAMP responses compared to untreated or wild-type cells. In conclusion, increased intracellular iron content in recombinant cells did not increase hepcidin responses compared to wild-type cells, resembling hemochromatosis. Furthermore, TFR2 expression altered within 4h of treatment, while HFE expression altered later at 24h and 48h, suggesting that TFR2 may function prior to HFE in HAMP regulation.
Collapse
Affiliation(s)
- Kosha Mehta
- Department of Biomedical Sciences, University of Westminster, London, UK
| | - Mark Busbridge
- Department of Clinical Biochemistry, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Derek Renshaw
- Faculty of Health & Life Sciences, Coventry University, Coventry, UK
| | - Robert W Evans
- School of Engineering and Design, Brunel University, London, UK
| | | | - Vinood B Patel
- Department of Biomedical Sciences, University of Westminster, London, UK.
| |
Collapse
|
33
|
Neuman MG, Malnick S, Maor Y, Nanau RM, Melzer E, Ferenci P, Seitz HK, Mueller S, Mell H, Samuel D, Cohen LB, Kharbanda KK, Osna NA, Ganesan M, Thompson KJ, McKillop IH, Bautista A, Bataller R, French SW. Alcoholic liver disease: Clinical and translational research. Exp Mol Pathol 2015; 99:596-610. [PMID: 26342547 DOI: 10.1016/j.yexmp.2015.09.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 09/01/2015] [Indexed: 02/05/2023]
Abstract
The present review spans a broad spectrum of topics dealing with alcoholic liver disease (ALD), including clinical research, translational research, pathogenesis and therapies. A special accent is placed on alcohol misuse, as alcohol is a legally commercialized and taxable product. Drinking alcohol, particularly from a young age, is a major health problem. Alcoholism is known to contribute to morbidity and mortality. A systematic literature search was performed in order to obtain updated data (2008-2015). The review is focused on genetic polymorphisms of alcohol metabolizing enzymes and the role of cytochrome p450 2E1 and iron in ALD. Alcohol-mediated hepatocarcinogenesis is also discussed in the presence or absence of co-morbidities such as viral hepatitis C as well as therapeutic the role of innate immunity in ALD-HCV. Moreover, emphasis was placed on alcohol and drug interactions, as well as liver transplantation for end-stage ALD. Finally, the time came to eradicate alcohol-induced liver and intestinal damage by using betaine.
Collapse
Affiliation(s)
- Manuela G Neuman
- In Vitro Drug Safety and Biotechnology, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Stephen Malnick
- Division of Gastroenterology, Kaplan Health Sciences Centre, Department of Medicine, Faculty of Medicine, Hebrew University, Rehovot, Israel
| | - Yaakov Maor
- Division of Gastroenterology, Kaplan Health Sciences Centre, Department of Medicine, Faculty of Medicine, Hebrew University, Rehovot, Israel
| | - Radu M Nanau
- In Vitro Drug Safety and Biotechnology, Toronto, Ontario, Canada
| | - Ehud Melzer
- Division of Gastroenterology, Kaplan Health Sciences Centre, Department of Medicine, Faculty of Medicine, Hebrew University, Rehovot, Israel
| | | | - Helmut K Seitz
- University of Heidelberg, Heidelberg, Germany; Department of Medicine, Gastroenterology and Hepatology, Centre for Alcohol Research, Salem Medical Centre, Heidelberg, Germany
| | - Sebastian Mueller
- University of Heidelberg, Heidelberg, Germany; Department of Medicine, Gastroenterology and Hepatology, Centre for Alcohol Research, Salem Medical Centre, Heidelberg, Germany
| | - Haim Mell
- Israel Antidrug and Alcohol Authority, Jerusalem, Israel
| | - Didier Samuel
- Liver Transplant Unit, Research Inserm-Paris XI Unit 785, Centre Hepatobiliaire, Hopital Paul Brousse, Villejuif, Paris, France
| | - Lawrence B Cohen
- Division of Gastroenterology, Sunnybrook Health Sciences Centre and Department of Internal Medicine, University of Toronto, Toronto, Canada
| | - Kusum K Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Internal Medicine, Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Natalia A Osna
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Internal Medicine, Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Murali Ganesan
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Internal Medicine, Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kyle J Thompson
- Department of Surgery, Carolinas Medical Center, Charlotte, NC 28203, USA
| | - Iain H McKillop
- Department of Surgery, Carolinas Medical Center, Charlotte, NC 28203, USA
| | - Abraham Bautista
- Office of Extramural Activities, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA
| | - Ramon Bataller
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | |
Collapse
|
34
|
Udali S, Guarini P, Ruzzenente A, Ferrarini A, Guglielmi A, Lotto V, Tononi P, Pattini P, Moruzzi S, Campagnaro T, Conci S, Olivieri O, Corrocher R, Delledonne M, Choi SW, Friso S. DNA methylation and gene expression profiles show novel regulatory pathways in hepatocellular carcinoma. Clin Epigenetics 2015; 7:43. [PMID: 25945129 PMCID: PMC4419480 DOI: 10.1186/s13148-015-0077-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 03/26/2015] [Indexed: 12/30/2022] Open
Abstract
Background Alcohol is a well-known risk factor for hepatocellular carcinoma (HCC), but the mechanisms underlying the alcohol-related hepatocarcinogenesis are still poorly understood. Alcohol alters the provision of methyl groups within the hepatic one-carbon metabolism, possibly inducing aberrant DNA methylation. Whether specific pathways are epigenetically regulated in alcohol-associated HCC is, however, unknown. The aim of the present study was to investigate the genome-wide promoter DNA methylation and gene expression profiles in non-viral, alcohol-associated HCC. From eight HCC patients undergoing curative surgery, array-based DNA methylation and gene expression data of all annotated genes were analyzed by comparing HCC tissue and homologous cancer-free liver tissue. Results After merging the DNA methylation with gene expression data, we identified 159 hypermethylated-repressed, 30 hypomethylated-induced, 49 hypermethylated-induced, and 56 hypomethylated-repressed genes. Notably, promoter DNA methylation emerged as a novel regulatory mechanism for the transcriptional repression of genes controlling the retinol metabolism (ADH1A, ADH1B, ADH6, CYP3A43, CYP4A22, RDH16), iron homeostasis (HAMP), one-carbon metabolism (SHMT1), and genes with a putative, newly identified function as tumor suppressors (FAM107A, IGFALS, MT1G, MT1H, RNF180). Conclusions A genome-wide DNA methylation approach merged with array-based gene expression profiles allowed identifying a number of novel, epigenetically regulated candidate tumor-suppressor genes in alcohol-associated hepatocarcinogenesis. Retinol metabolism genes and SHMT1 are also epigenetically regulated through promoter DNA methylation in alcohol-associated HCC. Due to the reversibility of epigenetic mechanisms by environmental/nutritional factors, these findings may open up to novel interventional strategies for hepatocarcinogenesis prevention in HCC related to alcohol, a modifiable dietary component. Electronic supplementary material The online version of this article (doi:10.1186/s13148-015-0077-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Silvia Udali
- Department of Medicine, University of Verona School of Medicine, Policlinico 'G.B. Rossi', P.le L.A. Scuro, 10, 37134 Verona, Italy
| | - Patrizia Guarini
- Department of Medicine, University of Verona School of Medicine, Policlinico 'G.B. Rossi', P.le L.A. Scuro, 10, 37134 Verona, Italy
| | - Andrea Ruzzenente
- Department of Surgery, University of Verona School of Medicine, Policlinico 'G.B. Rossi', P.le L.A. Scuro, 10, 37134 Verona, Italy
| | - Alberto Ferrarini
- Department of Biotechnology, Genetics and Heredity Section, University of Verona School of Agroindustrial Biotechnology, Ca' Vignal 1, Strada Le Grazie 15, 37134 Verona, Italy
| | - Alfredo Guglielmi
- Department of Surgery, University of Verona School of Medicine, Policlinico 'G.B. Rossi', P.le L.A. Scuro, 10, 37134 Verona, Italy
| | - Valentina Lotto
- Department of Medicine, University of Verona School of Medicine, Policlinico 'G.B. Rossi', P.le L.A. Scuro, 10, 37134 Verona, Italy
| | - Paola Tononi
- Department of Biotechnology, Genetics and Heredity Section, University of Verona School of Agroindustrial Biotechnology, Ca' Vignal 1, Strada Le Grazie 15, 37134 Verona, Italy
| | - Patrizia Pattini
- Department of Medicine, University of Verona School of Medicine, Policlinico 'G.B. Rossi', P.le L.A. Scuro, 10, 37134 Verona, Italy
| | - Sara Moruzzi
- Department of Medicine, University of Verona School of Medicine, Policlinico 'G.B. Rossi', P.le L.A. Scuro, 10, 37134 Verona, Italy
| | - Tommaso Campagnaro
- Department of Surgery, University of Verona School of Medicine, Policlinico 'G.B. Rossi', P.le L.A. Scuro, 10, 37134 Verona, Italy
| | - Simone Conci
- Department of Surgery, University of Verona School of Medicine, Policlinico 'G.B. Rossi', P.le L.A. Scuro, 10, 37134 Verona, Italy
| | - Oliviero Olivieri
- Department of Medicine, University of Verona School of Medicine, Policlinico 'G.B. Rossi', P.le L.A. Scuro, 10, 37134 Verona, Italy
| | - Roberto Corrocher
- Department of Medicine, University of Verona School of Medicine, Policlinico 'G.B. Rossi', P.le L.A. Scuro, 10, 37134 Verona, Italy
| | - Massimo Delledonne
- Department of Biotechnology, Genetics and Heredity Section, University of Verona School of Agroindustrial Biotechnology, Ca' Vignal 1, Strada Le Grazie 15, 37134 Verona, Italy
| | - Sang-Woon Choi
- Friedman School of Nutrition Science and Policy Tufts University, 150 Harrison Ave, Boston, MA 02111 USA ; Chaum Life Center, CHA University, 4-1, Cheongdam-dong, Gangnam-gu, 135-948 Seoul, Korea
| | - Simonetta Friso
- Department of Medicine, University of Verona School of Medicine, Policlinico 'G.B. Rossi', P.le L.A. Scuro, 10, 37134 Verona, Italy
| |
Collapse
|
35
|
Maras JS, Maiwall R, Harsha HC, Das S, Hussain MS, Kumar C, Bihari C, Rastogi A, Kumar M, Trehanpati N, Sharma S, Pandey A, Sarin SK. Dysregulated iron homeostasis is strongly associated with multiorgan failure and early mortality in acute-on-chronic liver failure. Hepatology 2015; 61:1306-20. [PMID: 25475192 DOI: 10.1002/hep.27636] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 10/22/2014] [Accepted: 11/20/2014] [Indexed: 12/11/2022]
Abstract
UNLABELLED Acute-on-chronic liver failure (ACLF) is an ailment with high incidence of multiorgan failure (MOF) and consequent mortality. Dysregulated iron homeostasis and macrophage dysfunction are linked to increased incidence of MOF. We investigated whether a panel of circulating iron-regulating proteins are associated with development of MOF and can predict 15- or 30-day mortality in ACLF patients. One hundred twenty patients with ACLF, 20 patients with compensated cirrhosis, and 20 healthy controls were studied. Relative protein expression profiling was performed in the derivative cohort and confirmed in the validation cohort. A panel of iron regulators and indices were determined. Multiparametric flow cytometry for quantitation of labile iron pool (LIP) was performed. Validation studies confirmed lower serum transferrin (Tf) and ceruloplasmin levels in ACLF and ACLF-MOF, compared to patients with cirrhosis and controls (P < 0.01). Serum iron and ferritin levels were markedly elevated (P < 0.001; P < 0.05) and hepcidin levels were lower (P < 0.001) in ACLF patients with MOF than those without and other groups (P < 0.001). Percentage Tf saturation (%SAT) was higher in ACLF-MOF (39.2%; P < 0.001) and correlated with poor outcome (hazard ratio: 6.970; P < 0.01). Intracellular LIP indices were significantly elevated in the subsets of circulating macrophages in ACLF-MOF, compared to other groups (P < 0.01). Whereas expression of iron-regulatory genes was markedly down-regulated, genes related to endoplasmic reticulum stress, apoptosis, and inflammation were up-regulated in ACLF patients, compared to patients with cirrhosis. Severe dysregulation of autophagy mechanisms was also observed in the former. CONCLUSIONS Iron metabolism and transport are severely deranged in ACLF patients and more so in those with MOF. %SAT, circulating hepcidin, and LIP in macrophages correlate with disease severity and %SAT could be used for early prognostication in ACLF patients.
Collapse
Affiliation(s)
- Jaswinder Singh Maras
- Departments of Research, Institute of Liver & Biliary Sciences (ILBS), New Delhi, India
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Background: Primary iron overload in African Americans has been reported predominantly from autopsy studies. Methods: We characterized hepatic iron phenotypes in 83 African Americans who underwent liver biopsy during the interval 1990 to 1995. We tabulated pathology report form data, iron grades in hepatocytes (0–4) and Kupffer cells (0–3) and abnormal liver histology. Increased iron was defined as hepatocyte or Kupffer iron grades ≥2, respectively. Heavy iron was defined as hepatocyte iron grade 3 or 4. Primary iron overload was defined as the presence of grade 3 or 4 hepatocellular iron in the absence of evidence of chronic alcohol effect, viral hepatitis, steatosis, unexplained inflammation, chronic erythrocyte transfusion or chronic ingestion of iron supplements. Results: There were 37 men and 46 women (mean age: 53 ± 15 [SD] years). We observed heavy ethanol consumption, 12.0%; viral hepatitis, 26.5%; steatosis without heavy ethanol consumption, 43.4%; inflammation, 45.6%; fibrosis, 26.2% and bridging fibrosis/cirrhosis, 29.4%. Logistic regression on bridging fibrosis/cirrhosis revealed positive associations with heavy ethanol consumption (P = 0.0410) and viral hepatitis (P = 0.0044). The 22 patients (26.5%) with increased iron had greater mean age, proportion of men and heavy ethanol consumption. Five patients had heavy iron staining, among whom were 3 women (mean age: 54 years) with primary iron overload. Two of the 3 women had cirrhosis and diabetes mellitus. Conclusions: Among 83 adult African Americans who underwent liver biopsy, 3.6% had hepatic iron phenotypes consistent with primary iron overload.
Collapse
|
37
|
|
38
|
Bi J, Ge S. Potential roles of BMP9 in liver fibrosis. Int J Mol Sci 2014; 15:20656-67. [PMID: 25393508 PMCID: PMC4264188 DOI: 10.3390/ijms151120656] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/13/2014] [Accepted: 11/04/2014] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis is a common phenomenon that is associated with several pathologies and is characterized by excessive extracellular matrix deposition that leads to progressive liver dysfunction. Bone morphogenetic protein 9 (BMP9) is the most recently discovered member of the BMP family. BMP9 bound with high affinity to activin receptor-like kinase 1 (ALK1) and endoglin in non-parenchymal liver cells. In addition, BMP9 activated Smad1/Smad5/Smad8 and induced the expression of the target genes inhibitor of differentiation 1 (Id1), hepcidin, Snail and the co-receptor endoglin in liver cells. Although the role of BMP9 in liver fibrosis is currently poorly understood, the presence of BMP9-activated proteins and its target genes have been reported to be associated with liver fibrosis development. This review summarizes the indirect connection between BMP9 and liver fibrosis, with a focus on the BMP9 signaling pathway members ALK1, endoglin, Id1, hepcidin and Snail. The observations on the role of BMP9 in regulating liver fibrosis may help in understanding the pathology mechanisms of liver disease. Furthermore, BMP9 could be served as a potent biomarker and the target of potential therapeutic drugs to treat hepatocytes fibrosis.
Collapse
Affiliation(s)
- Jianjun Bi
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
39
|
Williams JA, Manley S, Ding WX. New advances in molecular mechanisms and emerging therapeutic targets in alcoholic liver diseases. World J Gastroenterol 2014; 20:12908-12933. [PMID: 25278688 PMCID: PMC4177473 DOI: 10.3748/wjg.v20.i36.12908] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/07/2014] [Accepted: 04/16/2014] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease is a major health problem in the United States and worldwide. Chronic alcohol consumption can cause steatosis, inflammation, fibrosis, cirrhosis and even liver cancer. Significant progress has been made to understand key events and molecular players for the onset and progression of alcoholic liver disease from both experimental and clinical alcohol studies. No successful treatments are currently available for treating alcoholic liver disease; therefore, development of novel pathophysiological-targeted therapies is urgently needed. This review summarizes the recent progress on animal models used to study alcoholic liver disease and the detrimental factors that contribute to alcoholic liver disease pathogenesis including miRNAs, S-adenosylmethionine, Zinc deficiency, cytosolic lipin-1β, IRF3-mediated apoptosis, RIP3-mediated necrosis and hepcidin. In addition, we summarize emerging adaptive protective effects induced by alcohol to attenuate alcohol-induced liver pathogenesis including FoxO3, IL-22, autophagy and nuclear lipin-1α.
Collapse
|
40
|
Siddique A, Nelson JE, Aouizerat B, Yeh MM, Kowdley KV. Iron deficiency in patients with nonalcoholic Fatty liver disease is associated with obesity, female gender, and low serum hepcidin. Clin Gastroenterol Hepatol 2014; 12:1170-8. [PMID: 24269922 PMCID: PMC4028425 DOI: 10.1016/j.cgh.2013.11.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 11/11/2013] [Accepted: 11/13/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Iron deficiency is often observed in obese individuals. The iron regulatory hormone hepcidin is regulated by iron and cytokines interleukin (IL) 6 and IL1β. We examine the relationship between obesity, circulating levels of hepcidin, and IL6 and IL1β, and other risk factors in patients with nonalcoholic fatty liver disease (NAFLD) with iron deficiency. METHODS We collected data on 675 adult subjects (>18 years old) enrolled in the Nonalcoholic Steatohepatitis Clinical Research Network. Subjects with transferrin saturation <20% were categorized as iron deficient, whereas those with transferrin saturation ≥20% were classified as iron normal. We assessed clinical, demographic, anthropometric, laboratory, dietary, and histologic data from patients, and serum levels of hepcidin and cytokines IL6 and IL1β. Univariate and multivariate analysis were used to identify risk factors for iron deficiency. RESULTS One-third of patients (231 of 675; 34%) were iron deficient. Obesity, diabetes, and metabolic syndrome were more common in subjects with iron deficiency (P < .01), compared with those that were iron normal. Serum levels of hepcidin were significantly lower in subjects with iron deficiency (61 ± 45 vs 81 ± 51 ng/mL; P < .0001). Iron deficiency was significantly associated with female gender, obesity, increased body mass index and waist circumference, presence of diabetes, lower alcohol consumption, black or American Indian/Alaska Native race (P ≤ .018), and increased levels of IL6 and IL1β (6.6 vs 4.8 for iron normal, P ≤ .0001; and 0.45 vs 0.32 for iron normal, P ≤ .005). CONCLUSIONS Iron deficiency is prevalent in patients with NAFLD and associated with female gender, increased body mass index, and nonwhite race. Serum levels of hepcidin were lower in iron-deficient subjects, reflecting an appropriate physiologic response to decreased circulating levels of iron, rather than a primary cause of iron deficiency in the setting of obesity and NAFLD.
Collapse
Affiliation(s)
- Asma Siddique
- Liver Center of Excellence, Digestive Disease Institute, Virginia Mason Medical Center, Seattle, Washington
| | - James E Nelson
- Liver Center of Excellence, Digestive Disease Institute, Virginia Mason Medical Center, Seattle, Washington; Benaroya Research Institute, Virginia Mason Medical Center, Seattle, Washington
| | - Bradley Aouizerat
- Department of Physiological Nursing and Institute for Human Genetics, University of California at San Francisco, San Francisco, California
| | - Matthew M Yeh
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Kris V Kowdley
- Liver Center of Excellence, Digestive Disease Institute, Virginia Mason Medical Center, Seattle, Washington; Benaroya Research Institute, Virginia Mason Medical Center, Seattle, Washington.
| |
Collapse
|
41
|
A male patient with ferroportin disease B and a female patient with iron overload similar to ferroportin disease B. Clin J Gastroenterol 2014; 7:260-4. [DOI: 10.1007/s12328-014-0487-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 03/09/2014] [Indexed: 10/25/2022]
|
42
|
Effects of Chinese Medicine Tong xinluo on Diabetic Nephropathy via Inhibiting TGF- β 1-Induced Epithelial-to-Mesenchymal Transition. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:123497. [PMID: 24864150 PMCID: PMC4016864 DOI: 10.1155/2014/123497] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 03/18/2014] [Accepted: 03/22/2014] [Indexed: 12/31/2022]
Abstract
Diabetic nephropathy (DN) is a major cause of chronic kidney failure and characterized by interstitial and glomeruli fibrosis. Epithelial-to-mesenchymal transition (EMT) plays an important role in the pathogenesis of DN. Tong xinluo (TXL), a Chinese herbal compound, has been used in China with established therapeutic efficacy in patients with DN. To investigate the molecular mechanism of TXL improving DN, KK-Ay mice were selected as models for the evaluation of pathogenesis and treatment in DN. In vitro, TGF-β1 was used to induce EMT. Western blot (WB), immunofluorescence staining, and real-time polymerase chain reaction (RT-PCR) were applied to detect the changes of EMT markers in vivo and in vitro, respectively. Results showed the expressions of TGF-β1 and its downstream proteins smad3/p-smad3 were greatly reduced in TXL group; meantime, TXL restored the expression of smad7. As a result, the expressions of collagen IV (Col IV) and fibronectin (FN) were significantly decreased in TXL group. In vivo, 24 h-UAER (24-hour urine albumin excretion ratio) and BUN (blood urea nitrogen) were decreased and Ccr (creatinine clearance ratio) was increased in TXL group compared with DN group. In summary, the present study demonstrates that TXL successfully inhibits TGF-β1-induced epithelial-to-mesenchymal transition in DN, which may account for the therapeutic efficacy in TXL-mediated renoprotection.
Collapse
|
43
|
Mueller K, Sunami Y, Stuetzle M, Guldiken N, Kucukoglu O, Mueller S, Kulaksiz H, Schwarz P, Strnad P. CHOP-mediated hepcidin suppression modulates hepatic iron load. J Pathol 2013; 231:532-42. [PMID: 23749468 DOI: 10.1002/path.4221] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 05/16/2013] [Accepted: 06/05/2013] [Indexed: 12/22/2022]
Abstract
The liver is the central regulator of iron metabolism and accordingly, chronic liver diseases often lead to systemic iron overload due to diminished expression of the iron-regulatory hormone hepcidin. To study the largely unknown regulation of iron metabolism in the context of hepatic disease, we used two established models of chronic liver injury, ie repeated carbon tetrachloride (CCl(4)) or thioacetamide (TAA) injections. To determine the impact of CCAAT/enhancer-binding protein (C/EBP)-homologous protein (CHOP) on hepcidin production, the effect of a single TAA injection was determined in wild-type and CHOP knockout mice. Furthermore, CHOP and hepcidin expression was assessed in control subjects and patients with alcoholic liver disease. Both chronic injury models developed a distinct iron overload in macrophages. TAA-, but not CCl(4) - injected mice displayed additional iron accumulation in hepatocytes, resulting in a significant hepatic and systemic iron overload which was due to suppressed hepcidin levels. C/EBPα signalling, a known hepcidin inducer, was markedly inhibited in TAA mice, due to lower C/EBPα levels and overexpression of CHOP, a C/EBPα inhibitor. A single TAA injection resulted in a long-lasting (> 6 days) suppression of hepcidin levels and CHOP knockouts (compared to wild-types) displayed significantly attenuated hepcidin down-regulation in response to acute TAA administration. CHOP mRNA levels increased 5-fold in alcoholic liver disease patients versus controls (p < 0.005) and negatively correlated with hepcidin expression. Our results establish CHOP as an important regulator of hepatic hepcidin expression in chronic liver disease. The differences in iron metabolism between the two widely used fibrosis models likely reflect the differential regulation of hepcidin expression in human liver disease.
Collapse
Affiliation(s)
- Katrin Mueller
- Department of Internal Medicine I, University Hospital Ulm, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Costa Matos L, Batista P, Monteiro N, Ribeiro J, Cipriano MA, Henriques P, Girão F, Carvalho A. Iron stores assessment in alcoholic liver disease. Scand J Gastroenterol 2013; 48:712-8. [PMID: 23534461 DOI: 10.3109/00365521.2013.781217] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND The relation between alcoholic liver disease (ALD) and iron overload is well known. Liver biopsy is the gold standard for assessing iron stores. MRI is also validated for liver iron concentration (LIC) assessment. We aimed to assess the effect of active drinking in liver iron stores and the practicability of measuring LIC by MRI in ALD patients. MATERIALS AND METHODS We measured LIC by MRI in 58 ALD patients. We divided patients into two groups - with and without active alcoholism - and we compared several variables between them. We evaluated MRI-LIC, liver iron stores grade, ferritin and necroinflammatory activity grade for significant correlations. RESULTS Significant necroinflammation (40.0% vs. 4.3%), LIC (40.1 vs. 24.3 µmol/g), and ferritin (1259.7 vs. 568.7 pmol/L) were significantly higher in drinkers. LIC values had a strong association with iron stores grade (r s = 0.706). Ferritin correlated with LIC (r s = 0.615), iron stores grade (r s = 0.546), and necroinflammation (r s = 0.313). The odds ratio for elevated serum ferritin when actively drinking was 7.32. CONCLUSION Active alcoholism is associated with increased ALD activity. It is also the key factor in iron overload. Scheuers' semiquantitative score with Perls' staining gives a fairly accurate picture of liver iron overload. Serum ferritin also shows a good correlation with LIC values and biopsy iron stores grade. As most patients present only with mild iron overload, serum ferritin measurement and semiquantitative evaluation of iron stores are adequate, considering MRI high cost. However, if MRI is required to evaluate liver structure, LIC assessment could be performed without added cost.
Collapse
Affiliation(s)
- Luís Costa Matos
- Faculty of Medicine of the University of Coimbra, 3004-504 Coimbra, Portugal.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Wang QM, Du JL, Duan ZJ, Guo SB, Sun XY, Liu Z. Inhibiting heme oxygenase-1 attenuates rat liver fibrosis by removing iron accumulation. World J Gastroenterol 2013; 19:2921-2934. [PMID: 23704825 PMCID: PMC3660817 DOI: 10.3748/wjg.v19.i19.2921] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 03/08/2013] [Accepted: 03/29/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of the heme oxygenase (HO)-1/carbon monoxide system on iron deposition and portal pressure in rats with hepatic fibrosis induced by bile duct ligation (BDL).
METHODS: Male Sprague-Dawley rats were divided randomly into a Sham group, BDL group, Fe group, deferoxamine (DFX) group, zinc protoporphyrin (ZnPP) group and cobalt protoporphyrin (CoPP) group. The levels of HO-1 were detected using different methods. The serum carboxyhemoglobin (COHb), iron, and portal vein pressure (PVP) were also quantified. The plasma and mRNA levels of hepcidin were measured. Hepatic fibrosis and its main pathway were assessed using Van Gieson’s stain, hydroxyproline, transforming growth factor-β1 (TGF-β1), nuclear factor-E2-related factor 2 (Nrf2), matrix metalloproteinase-2 (MMP-2) and tissue inhibitor of metalloproteinase-1 (TIMP-1).
RESULTS: Serum COHb and protein and mRNA expression levels of HO-1 and Nrf2 were increased in the BDL group compared with the Sham group and were much higher in the CoPP group. The ZnPP group showed lower expression of HO-1 and Nrf2 and lower COHb. The levels of iron and PVP were enhanced in the BDL group but were lower in the ZnPP and DFX groups and were higher in the CoPP and Fe groups. Hepcidin levels were higher, whereas superoxide dismutase levels were increased and malonaldehyde levels were decreased in the ZnPP and DFX groups. The ZnPP group also showed inhibited TGF-β1 expression and regulated TIMP-1/MMP-2 expression, as well as obviously attenuated liver fibrosis.
CONCLUSION: Reducing hepatic iron deposition and CO levels by inhibiting HO-1 activity though the Nrf2/Keap pathway could be helpful in improving hepatic fibrosis and regulating PVP.
Collapse
|
46
|
COSTA-MATOS L, BATISTA P, MONTEIRO N, HENRIQUES P, GIRÃO F, CARVALHO A. HFE MUTATIONS AND IRON OVERLOAD IN PATIENTS WITH ALCOHOLIC LIVER DISEASE. ARQUIVOS DE GASTROENTEROLOGIA 2013; 50:35-41. [DOI: 10.1590/s0004-28032013000100008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 11/30/2012] [Indexed: 12/31/2022]
Abstract
ContextAlcoholic liver disease (ALD) is generally associated with iron overload, which may contribute to its pathogenesis, through increased oxidative stress and cellular damage. There are conflicting reports in literature about hemochromatosis (HFE) gene mutations and the severity of liver disease in alcoholic patients.ObjectivesTo compare the prevalence of mutations in the hemochromatosis (HFE) gene between patients with ALD and healthy controls; to assess the relation of HFE mutations with liver iron stores and liver disease severity.MethodsLiver biopsy specimens were obtained from 63 ALD patients (during routine treatment) and 52 healthy controls (during elective cholecystectomy). All individuals underwent routine liver function tests and HFE genotyping (to detect wild-type sequences and C282Y, H63D, S65C, E168Q, E168X, V59M, H63H, P160delC, Q127H, Q283P, V53M and W164X mutations). Associations between HFE mutations and risk of excessive liver iron stores, abnormal serum ferritin, liver fibrosis, or necroinflammatory activity were assessed by multivariate logistic regression analysis.ResultsALD patients had significantly higher serum ferritin and transferrin saturation than controls (both P<0.05), but the distribution of HFE mutations was similar between the two groups. For ALD patients, the odds ratio for having at least one HFE mutation and excessive liver iron stores was 17.23 (95% confidence interval (CI): 2.09-142.34, P = 0.008). However, the presence of at least one HFE mutation was not associated with an increased risk of liver fibrosis or necroinflammatory activity. Active alcohol ingestion showed the strongest association to increased serum ferritin (OR = 8.87, 95% CI: 2.11-34.78, P = 0.003).ConclusionsALD patients do not present with a differential profile of HFE mutations from healthy controls. In ALD patients, however, the presence of at least one HFE mutation increases the risk of having excessive liver iron stores but has no detectable effects on liver disease activity or severity.
Collapse
Affiliation(s)
- Luís COSTA-MATOS
- University of Coimbra, Portugal; Tondela-Viseu Hospital Centre E.P.E., Portugal
| | | | | | | | | | | |
Collapse
|