1
|
Lin Z, Ying C, Si X, Xue N, Liu Y, Zheng R, Chen Y, Pu J, Zhang B. NOX4 exacerbates Parkinson's disease pathology by promoting neuronal ferroptosis and neuroinflammation. Neural Regen Res 2025; 20:2038-2052. [PMID: 38993139 PMCID: PMC11691449 DOI: 10.4103/nrr.nrr-d-23-01265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 11/18/2023] [Accepted: 03/18/2024] [Indexed: 07/13/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202507000-00026/figure1/v/2024-09-09T124005Z/r/image-tiff Parkinson's disease is primarily caused by the loss of dopaminergic neurons in the substantia nigra compacta. Ferroptosis, a novel form of regulated cell death characterized by iron accumulation and lipid peroxidation, plays a vital role in the death of dopaminergic neurons. However, the molecular mechanisms underlying ferroptosis in dopaminergic neurons have not yet been completely elucidated. NADPH oxidase 4 is related to oxidative stress, however, whether it regulates dopaminergic neuronal ferroptosis remains unknown. The aim of this study was to determine whether NADPH oxidase 4 is involved in dopaminergic neuronal ferroptosis, and if so, by what mechanism. We found that the transcriptional regulator activating transcription factor 3 increased NADPH oxidase 4 expression in dopaminergic neurons and astrocytes in an 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine-induced Parkinson's disease model. NADPH oxidase 4 inhibition improved the behavioral impairments observed in the Parkinson's disease model animals and reduced the death of dopaminergic neurons. Moreover, NADPH oxidase 4 inhibition reduced lipid peroxidation and iron accumulation in the substantia nigra of the Parkinson's disease model animals. Mechanistically, we found that NADPH oxidase 4 interacted with activated protein kinase C α to prevent ferroptosis of dopaminergic neurons. Furthermore, by lowering the astrocytic lipocalin-2 expression, NADPH oxidase 4 inhibition reduced 1-methyl-4-phenyl-1,2,3,6 tetrahydropyridine-induced neuroinflammation. These findings demonstrate that NADPH oxidase 4 promotes ferroptosis of dopaminergic neurons and neuroinflammation, which contribute to dopaminergic neuron death, suggesting that NADPH oxidase 4 is a possible therapeutic target for Parkinson's disease.
Collapse
Affiliation(s)
- Zhihao Lin
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Changzhou Ying
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Xiaoli Si
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Naijia Xue
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yi Liu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ran Zheng
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Ying Chen
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jiali Pu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
2
|
Zheng C, Li S, Lyu H, Chen C, Mueller J, Dropmann A, Hammad S, Dooley S, He S, Mueller S. Direct Ingestion of Oxidized Red Blood Cells (Efferocytosis) by Hepatocytes. Hepat Med 2024; 16:65-77. [PMID: 39247515 PMCID: PMC11380495 DOI: 10.2147/hmer.s469990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024] Open
Abstract
Purpose Both hepatic iron accumulation and hemolysis have been identified as independent prognostic factor in alcohol-related liver disease (ALD); however, the mechanisms still remain poorly understood. We here demonstrate that hepatocytes are able to directly ingest aged and ethanol-primed red blood cells (RBCs), a process termed efferocytosis. Methods Efferocytosis of RBCs was directly studied in vitro and observed by live microscopy for real-time visualization. RBCs pretreated with either CuSO4 or ethanol following co-incubation with Huh7 cells and murine primary hepatocytes. Heme oxygenase-1 (HO-1) and other targets were measured by q-PCR. Results As shown by live microscopy, oxidized RBCs, but not intact RBCs, are rapidly ingested by both Huh7 cells and murine primary hepatocytes within 10 minutes. In some cases, more than 10 RBCs were seen within hepatocytes, surrounding the nucleus. RBC efferocytosis also rapidly induces HO1, its upstream regulator Nuclear factor erythroid 2-related factor 2 (Nrf2) and ferritin, indicating efficient heme degradation. Preliminary data further suggest that hepatocyte efferocytosis of oxidized RBCs is, at least in part, mediated by scavenging receptors such as ASGPR1. Of note, pretreatment of RBCs with ethanol but also heme and bilirubin also initiated efferocytosis. In a cohort of heavy human drinkers, a significant correlation of hepatic ASGPR1 with the heme degradation pathway was observed. Conclusion We here demonstrate that hepatocytes can directly ingest and degrade oxidized RBCs through efferocytosis, a process that can be also triggered by ethanol, heme and bilirubin. Our findings are highly suggestive for a novel mechanism of hepatic iron overload in ALD patients.
Collapse
Affiliation(s)
- Chaowen Zheng
- Center for Alcohol Research, University of Heidelberg, Heidelberg, Germany
- Division of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Siyuan Li
- Center for Alcohol Research, University of Heidelberg, Heidelberg, Germany
| | - Huanran Lyu
- Center for Alcohol Research, University of Heidelberg, Heidelberg, Germany
| | - Cheng Chen
- Center for Alcohol Research, University of Heidelberg, Heidelberg, Germany
| | - Johannes Mueller
- Center for Alcohol Research, University of Heidelberg, Heidelberg, Germany
| | - Anne Dropmann
- Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Seddik Hammad
- Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Department of Forensic Medicine and Veterinary Toxicology, Faculty of Veterinary Medicine, South Valley University, Quena, Egypt
| | - Steven Dooley
- Molecular Hepatology Section, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Songqing He
- Division of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Sebastian Mueller
- Center for Alcohol Research, University of Heidelberg, Heidelberg, Germany
- Viscera AG Bauchmedizin, Bern, Switzerland
| |
Collapse
|
3
|
Wang S, Ren H, Fan C, Lin Q, Liu M, Tian J. Ochratoxin A Induces Renal Cell Ferroptosis by Disrupting Iron Homeostasis and Increasing ROS. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:1734-1744. [PMID: 38133486 DOI: 10.1021/acs.jafc.3c04495] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Mycotoxin ochratoxin A (OTA) is a critical food safety concern due to its nephron-toxic effects and is detected in a wide range of food and feedstuffs. OTA nephrotoxicity is related to oxidative stress and damage. However, the mediator(s) of the excessive oxidative stress is unclear. The current study used human kidney cell lines to investigate whether and how intracellular iron contributed to OTA-induced ROS accumulation and how OTA-induced iron-dependent ferroptotic cell death. Our results showed that OTA treatment affected the cell viability and induced the typical characteristics of cell ferroptosis. Furthermore, gene and protein expression results indicated that OTA disrupted iron homeostasis by upregulating the expression levels of iron importer TFR1 and FTH, while downregulating the expression level of iron exporter FPN and dramatically increasing its negative regulator Hepcidin. The changes were consistent with the induction of intracellular iron accumulation and elevated levels of oxidative stress and lipid peroxidation. Additionally, co-treatment with OTA and an iron chelator significantly improved cell viability, reduced cellular total iron and ROS, and reversed OTA-induced changes in iron metabolism gene expression levels. Interestingly, the addition of a ROS scavenger also reversed cell death and changes in mRNA and protein expression levels of iron metabolism genes but to a lesser degree than that of the iron-chelating agent. Our results revealed that OTA induced ferroptosis in renal cells by disrupting iron homeostasis and increasing ROS.
Collapse
Affiliation(s)
- Sen Wang
- International Cooperation Joint Laboratory of Jiangsu Province Colleges and Universities, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province 221116, China
| | - Hui Ren
- International Cooperation Joint Laboratory of Jiangsu Province Colleges and Universities, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province 221116, China
| | - Chen Fan
- International Cooperation Joint Laboratory of Jiangsu Province Colleges and Universities, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province 221116, China
| | - Qian Lin
- International Cooperation Joint Laboratory of Jiangsu Province Colleges and Universities, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province 221116, China
| | - Man Liu
- International Cooperation Joint Laboratory of Jiangsu Province Colleges and Universities, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province 221116, China
| | - Jun Tian
- International Cooperation Joint Laboratory of Jiangsu Province Colleges and Universities, School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu Province 221116, China
| |
Collapse
|
4
|
Nienaber A, Uyoga MA, Dolman-Macleod RC, Malan L. Iron Status and Supplementation during Tuberculosis. Microorganisms 2023; 11:microorganisms11030785. [PMID: 36985358 PMCID: PMC10055784 DOI: 10.3390/microorganisms11030785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Tuberculosis (TB) is characterised by chronic non-resolving inflammation. The effects of the host immune and inflammatory response to reduce iron acquisition by the bacteria, together with other contributing factors, predispose TB patients to anaemia of infection and iron deficiency anaemia (IDA). The presence of anaemia in TB patients has been linked to poor clinical outcomes. However, due to the reliance of the bacteria on iron, the management of anaemia in TB is complicated, and anaemia of infection is likely to resolve with correct TB drug treatment. On the other hand, IDA may require iron supplementation. This review aims to describe iron metabolism in TB and how this contributes to the development of iron deficiency and anaemia. Additionally, we summarise the evidence on the association between iron status and clinical outcomes as well as the available preclinical and clinical trials on iron supplementation in TB.
Collapse
Affiliation(s)
- Arista Nienaber
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa
| | - Mary A Uyoga
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa
| | - Robin C Dolman-Macleod
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa
| | - Linda Malan
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa
| |
Collapse
|
5
|
Abstract
Osteosarcoma (OS) is the most common primary solid malignant tumour of bone, with rapid progression and a very poor prognosis. Iron is an essential nutrient that makes it an important player in cellular activities due to its inherent ability to exchange electrons, and its metabolic abnormalities are associated with a variety of diseases. The body tightly regulates iron content at the systemic and cellular levels through various mechanisms to prevent iron deficiency and overload from damaging the body. OS cells regulate various mechanisms to increase the intracellular iron concentration to accelerate proliferation, and some studies have revealed the hidden link between iron metabolism and the occurrence and development of OS. This article briefly describes the process of normal iron metabolism, and focuses on the research progress of abnormal iron metabolism in OS from the systemic and cellular levels.
Collapse
Affiliation(s)
- Xiaowei Ma
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, People's Republic of China
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, 12 Health Road, Hebei Province, Shijiazhuang, 050011, People's Republic of China
| | - Jiazheng Zhao
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, 12 Health Road, Hebei Province, Shijiazhuang, 050011, People's Republic of China
| | - Helin Feng
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Nanli, Panjiayuan, Chaoyang District, Beijing, 100021, People's Republic of China.
| |
Collapse
|
6
|
Suárez-Ortegón MF, Arbeláez A, Moreno-Navarrete JM, Ortega-Ávila JG, Mosquera M, Fernández-Real JM. Soluble Transferrin Receptor, Antioxidant Status and Cardiometabolic Risk in Apparently Healthy Individuals. Antioxidants (Basel) 2022; 12:antiox12010019. [PMID: 36670881 PMCID: PMC9854855 DOI: 10.3390/antiox12010019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/06/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Body iron excess appears to be related to insulin resistance and cardiometabolic risk and increased oxidative stress might be involved in this relationship. Very few studies have described the association between soluble transferrin receptor (sTfR) levels and cardiometabolic risk in the general population or antioxidant status. There were 239 subjects (20−65 years old) included in this cross-sectional study. Linear regressions adjusting for BMI, menopausal status, insulin resistance (HOMA-IR), physical inactivity, alcohol intake and subclinical/chronic inflammation were used to describe the association between sTfR, total antioxidant capacity (TAC), and measures of cardio-metabolic risk. sTfR levels were positively associated with TAC in men (βeta [95% confidence interval ]: 0.31 [0.14 to 0.48]) and women (βeta = 0.24 [0.07 to 0.40]) in non-adjusted and adjusted models (p < 0.05). In men, sTfR levels were inversely associated with waist circumference (βeta [95% confidence interval]: −1.12 [−2.30 to −0.22]) and fasting glucose (−2.7 (−4.82 to −0.57), and positively with LDL cholesterol (12.41 (6.08 to 18.57) before and after adjustments for confounding variables. LDL cholesterol had a significant and positive association with TAC in non-adjusted and adjusted models in men (p < 0.05). sTfR levels are significantly associated with antioxidant status and a few specific cardio-metabolic risk variables, independently of covariates that included serum ferritin and hepcidin. This might imply that iron biomarkers in regard to cardiometabolic risk reflect physiological contexts other than iron metabolism.
Collapse
Affiliation(s)
- Milton Fabian Suárez-Ortegón
- Departamento de Alimentación y Nutrición, Facultad de Ciencias de La Salud, Pontificia Universidad Javeriana Seccional Cali, Cali 760030, Colombia
- Nutrition Group, Universidad del Valle, Cali 760030, Colombia
- Correspondence: (M.F.S.-O.); (J.M.F.-R.); Tel.: +57-321-82-00 (M.F.S.-O.); +34-972-940-200 (J.M.F.-R.)
| | - Alejandra Arbeláez
- Nutrition Group, Universidad del Valle, Cali 760030, Colombia
- Physiological Sciences Department, Universidad del Valle, Cali 760030, Colombia
| | - José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d’Investigació Biomèdica de Girona (IdIBGi), 28029 Madrid, Spain
| | - José Guillermo Ortega-Ávila
- Nutrition Group, Universidad del Valle, Cali 760030, Colombia
- Departamento de Ciencias Básicas, Facultad de Ciencias de La Salud, Pontificia Universidad Javeriana Seccional Cali, Cali 760030, Colombia
| | - Mildrey Mosquera
- Nutrition Group, Universidad del Valle, Cali 760030, Colombia
- Physiological Sciences Department, Universidad del Valle, Cali 760030, Colombia
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d’Investigació Biomèdica de Girona (IdIBGi), 28029 Madrid, Spain
- CIBEROBN (CB06/03/010), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Correspondence: (M.F.S.-O.); (J.M.F.-R.); Tel.: +57-321-82-00 (M.F.S.-O.); +34-972-940-200 (J.M.F.-R.)
| |
Collapse
|
7
|
Abstract
The liver is the major target organ of continued alcohol consumption at risk and resulting alcoholic liver disease (ALD) is the most common liver disease worldwide. The underlying molecular mechanisms are still poorly understood despite decades of scientific effort limiting our abilities to identify those individuals who are at risk to develop the disease, to develop appropriate screening strategies and, in addition, to develop targeted therapeutic approaches. ALD is predestined for the newly evolving translational medicine, as conventional clinical and health care structures seem to be constrained to fully appreciate this disease. This concept paper aims at summarizing the 15 years translational experience at the Center of Alcohol Research in Heidelberg, namely based on the long-term prospective and detailed characterization of heavy drinkers with mortality data. In addition, novel experimental findings will be presented. A special focus will be the long-known hepatic iron accumulation, the somewhat overlooked role of the hematopoietic system and novel insights into iron sensing and the role of hepcidin. Our preliminary work indicates that enhanced red blood cell (RBC) turnover is critical for survival in ALD patients. RBC turnover is not primarily due to vitamin deficiency but rather to ethanol toxicity directly targeted to erythrocytes but also to the bone marrow stem cell compartment. These novel insights also help to explain long-known aspects of ALD such as mean corpuscular volume of erythrocytes (MCV) and elevated aspartate transaminase (GOT/AST) levels. This work also aims at identifying future projects, naming unresolved observations, and presenting novel hypothetical concepts still requiring future validation.
Collapse
|
8
|
Testa MTDJ, Cella PS, Marinello PC, Frajacomo FTT, Padilha CDS, Perandini PC, Moura FA, Duarte JA, Cecchini R, Guarnier FA, Deminice R. Resistance Training Attenuates Activation of STAT3 and Muscle Atrophy in Tumor-Bearing Mice. Front Oncol 2022; 12:880787. [PMID: 35847939 PMCID: PMC9283857 DOI: 10.3389/fonc.2022.880787] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose Although the role of signal transducers and activators of transcription (STAT3) in cachexia due to the association of circulating IL-6 and muscle wasting has been extensively demonstrated, the effect of resistance training on STAT3 in mediating muscle atrophy in tumor-bearing mice is unknown. The aim of this study is to investigate the effects of resistance exercise training on inflammatory cytokines and oxidative-mediated STAT3 activation and muscle loss prevention in tumor-bearing mice. Methods Male Swiss mice were inoculated with Ehrlich tumor cells and exposed or not exposed to resistance exercise protocol of ladder climbing. Skeletal muscle STAT3 protein content was measured, compared between groups, and tested for possible association with plasma interleukins and local oxidative stress markers. Components of the ubiquitin-proteasome and autophagy pathways were assessed by real-time PCR or immunoblotting. Results Resistance training prevented STAT3 excessive activation in skeletal muscle mediated by the overabundance of plasma IL-6 and muscle oxidative stress. These mechanisms contributed to preventing the increased key genes and proteins of ubiquitin-proteasome and autophagy pathways in tumor-bearing mice, such as Atrogin-1, LC3B-II, and Beclin-1. Beyond preventing muscle atrophy, RT also prevented strength loss and impaired locomotor capacity, hallmarks of sarcopenia. Conclusion Our results suggest that STAT3 inhibition is central in resistance exercise protective effects against cancer-induced muscle atrophy and strength loss.
Collapse
Affiliation(s)
| | - Paola Sanches Cella
- Department of Physical Education, State University of Londrina, Londrina, Brazil
| | - Poliana Camila Marinello
- Department of Physical Education, State University of Londrina, Londrina, Brazil
- Department of General Pathology, State University of Londrina, Londrina, Brazil
| | | | - Camila de Souza Padilha
- Department of Physical Education, State University of Londrina, Londrina, Brazil
- Department of Physical Education, State University of São Paulo (UNESP), Presidente Prudente, Brazil
| | | | - Felipe Arruda Moura
- Department of Physical Education, State University of Londrina, Londrina, Brazil
| | | | - Rubens Cecchini
- Department of General Pathology, State University of Londrina, Londrina, Brazil
| | | | - Rafael Deminice
- Department of Physical Education, State University of Londrina, Londrina, Brazil
- *Correspondence: Rafael Deminice, ; orcid.org/0000-0002-9246-1079
| |
Collapse
|
9
|
Padovani D, Galardon E. Molecular Basis for the Interaction of Catalase with d-Penicillamine: Rationalization of Some of Its Deleterious Effects. Chem Res Toxicol 2022; 35:412-421. [PMID: 35191669 DOI: 10.1021/acs.chemrestox.1c00313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
d-Penicillamine (d-Pen) is a sulfur compound used in the management of rheumatoid arthritis, Wilson's disease (WD), and alcohol dependence. Many side effects are associated with its use, particularly after long-term treatment. However, the molecular basis for such side effects is poorly understood. Based on the well-known oxidase activity of hemoproteins and the participation of catalase in cellular H2O2 redox signaling, we posit that d-Pen could inactivate catalase, thus disturbing H2O2 levels. Herein, we report on the molecular basis that could partly explain the side effects associated with this drug compound, and we demonstrate that it induces the formation of compound II, a temporarily inactive state of the enzyme, through two distinct mechanisms. Initially, d-Pen reacts with native catalase and/or iron metal ions, used to mimic non-heme iron overload observed in long-term treated WD patients, to generate thiyl radicals. These radicals partake in a futile redox cycle, thus producing superoxide radical anions O2•- and hydrogen peroxide H2O2. Then, either H2O2 unexpectedly reacts with reduced CAT-Fe(II) to produce compound II or both aforementioned reactive oxygen species intervene in compound II generation through compound I formation and then reduction. These findings support the evidence that d-Pen could perturb H2O2 redox homeostasis through transient but recurring catalase inactivation, which may in part rationalize some deleterious effects observed with this therapeutic agent, as discussed.
Collapse
Affiliation(s)
- Dominique Padovani
- UMR 8601, LCBPT, CNRS-Université de Paris, 45 rue des Sts Pères, 75006 Paris, France
| | - Erwan Galardon
- UMR 8601, LCBPT, CNRS-Université de Paris, 45 rue des Sts Pères, 75006 Paris, France
| |
Collapse
|
10
|
Changes in Hepcidin Levels in an Animal Model of Anemia of Chronic Inflammation: Mechanistic Insights Related to Iron Supplementation and Hepcidin Regulation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:4357756. [PMID: 34873429 PMCID: PMC8643261 DOI: 10.1155/2021/4357756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/21/2021] [Indexed: 12/14/2022]
Abstract
We examined changes in hepcidin (closely associated with anemia of chronic inflammation (ACI)) and upstream regulatory pathways after intravenous (IV) iron supplementation in an ACI animal model. ACI was induced in male Sprague-Dawley rats by intraperitoneally administering complete Freund's adjuvant (CFA). Two weeks after starting CFA treatment, ACI rats received IV iron (CFA-iron) or vehicle (CFA-saline). Three days after IV iron treatment, iron profiles, hepcidin levels, and expression of proteins involved in the signaling pathways upstream of hepcidin transcription in the liver were measured. In CFA-treated rats, anemia with a concomitant increase in the levels of serum inflammatory cytokines and reactive oxygen species occurred. In CFA-iron rats, hemoglobin (Hb) concentration was still lower than that in control rats. In CFA-saline rats, hepatic hepcidin and ferritin levels increased compared with those in control rats and were further increased in CFA-iron rats. In CFA-saline rats, NADPH oxidase- (NOX-) 2, NOX-4, and superoxide dismutase levels in the liver were upregulated compared with those in control rats and their levels were further increased in CFA-iron rats. In CFA-saline rats, activities of the IL-6/STAT and BMP/SMAD pathways were enhanced in the liver compared with those in control rats and their levels were further increased in CFA-iron rats, whereas IL-6 expression remained unaffected after IV iron administration. In HepG2 cells, iron caused phosphorylation of STAT-3 and SMAD1/5 and knockdown of STAT-3 and SMAD1/5 using siRNAs reduced iron-induced hepcidin upregulation to levels similar to those in corresponding control cells. Renal erythropoietin expression and serum erythroferrone concentration were lower in CFA-iron rats than those in control rats. In ACI rats, IV iron supplementation did not recover Hb within three days despite an increase in hepatic ferritin levels, which might be attributable to an additional increase in hepcidin levels that was already upregulated under ACI conditions. Both STAT-3 phosphorylation and SMAD1/5 phosphorylation were associated with hepcidin upregulation after IV iron treatment, and this seems to be linked to iron-induced oxidative stress.
Collapse
|
11
|
Wang S, Chen C, Yu L, Mueller J, Rausch V, Mueller S. Bone morphogenetic protein 6-mediated crosstalk between endothelial cells and hepatocytes recapitulates the iron-sensing pathway in vitro. J Biol Chem 2021; 297:101378. [PMID: 34740612 PMCID: PMC8637636 DOI: 10.1016/j.jbc.2021.101378] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/21/2022] Open
Abstract
Liver sinusoidal endothelial cell–derived bone morphogenetic protein 6 (BMP6) and the BMP6–small mothers against decapentaplegic homolog (SMAD) signaling pathway are essential for the expression of hepcidin, the secretion of which is considered the systemic master switch of iron homeostasis. However, there are continued controversies related to the strong and direct suppressive effect of iron on hepatocellular hepcidin in vitro in contrast to in vivo conditions. Here, we directly studied the crosstalk between endothelial cells (ECs) and hepatocytes using in vitro coculture models that mimic hepcidin signaling in vivo. Huh7 cells were directly cocultured with ECs, and EC conditioned media (CM) were also used to culture Huh7 cells and primary mouse hepatocytes. To explore the reactions of ECs to surrounding iron, they were grown in the presence of ferric ammonium citrate and heme, two iron-containing molecules. We found that both direct coculture with ECs and EC-CM significantly increased hepcidin expression in Huh7 cells. The upstream SMAD pathway, including phosphorylated SMAD1/5/8, SMAD1, and inhibitor of DNA binding 1, was induced by EC-CM, promoting hepcidin expression. Efficient blockage of this EC-mediated hepcidin upregulation by an inhibitor of the BMP6 receptor ALK receptor tyrosine kinase 2/3 or BMP6 siRNA identified BMP6 as a major hepcidin regulator in this coculture system, which highly fits the model of hepcidin regulation by iron in vivo. In addition, EC-derived BMP6 and hepcidin were highly sensitive to levels of not only ferric iron but also heme as low as 500 nM. We here establish a hepatocyte–endothelial coculture system to fully recapitulate iron regulation by hepcidin using EC-derived BMP6.
Collapse
Affiliation(s)
- Shijin Wang
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Cheng Chen
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Linna Yu
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Johannes Mueller
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Vanessa Rausch
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Sebastian Mueller
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
12
|
Yu LN, Wang SJ, Chen C, Rausch V, Elshaarawy O, Mueller S. Direct modulation of hepatocyte hepcidin signaling by iron. World J Hepatol 2021; 13:1378-1393. [PMID: 34786173 PMCID: PMC8568584 DOI: 10.4254/wjh.v13.i10.1378] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/04/2021] [Accepted: 08/27/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Liver-secreted hepcidin is the systemic master switch of iron homeostasis and decreased levels of hepcidin are considered to cause iron overload not only in hereditary hemochromatosis but also in hemolytic anemia and chronic liver diseases. The regulation of hepcidin is complex and its response to iron is still not completely understood.
AIM To study the direct effect of iron on various established hepcidin signaling pathways in hepatoma cells or primary hepatocytes.
METHODS Hepcidin mRNA expression was studied by quantitative real-time (qRT)-PCR in the presence of various forms of iron including ferric ammonium citrate (FAC) in hepatoma cells (Huh7), murine primary hepatocytes and an established co-culture model of phorbol myristate acetate-differentiated THP-1 monocytes and Huh7 cells. To analyze hepcidin signaling, the response to bone morphogenetic protein 6 (BMP6), interleukin (IL)-6, IL-1β, hypoxia and lipopolysaccharide (LPS) were studied. Hepcidin and small mothers against decapentaplegic 6 (SMAD6) mRNA levels were assessed by qRT-PCR and the expression of phosphorylated signal transducer and activator of transcription 3 (phospho-STAT3), STAT3, phospho-SMAD1/5/8 and SMAD1 proteins were analyzed by western blot.
RESULTS All iron III forms including FAC efficiently blocked hepcidin mRNA expression at non-toxic dosages in Huh7 cells or primary hepatocytes in a time and dose-dependent manner (P < 0.001; P < 0.05). Hepcidin blockage could be efficiently blunted by iron chelators salicylaldehyde isonicotinoyl hydrazone (SIH) and Desferal (P < 0.001). FAC also inhibited BMP6, hypoxia, IL-1β and IL-6-mediated hepcidin induction (P < 0.001; P < 0.001; P < 0.05; P < 0.001), and FAC also inhibited LPS-mediated hepatic hepcidin induction in co-culture model (P < 0.001). Moreover, FAC reduced SMAD6 mRNA and p-SMAD1/5/8 protein expression at basal or upon stimulation by BMP6 (P < 0.05; P < 0.01), and FAC also reduced SMAD6 and p-SMAD1/5/8 expression under hypoxia (P < 0.01; P < 0.05). However, FAC has no significant effect on p-STAT3 protein expression at basal or upon stimulation by various stimuli. Notably, in the presence of the BMP/SMAD signaling pathway inhibitor LDN193189 Hydrochloride (LDN), FAC was unable to further decrease hepcidin, SMAD6 and p-SMAD1/5/8 expression compared with LDN alone.
CONCLUSION Iron directly blocks hepatocellular hepcidin signaling through the BMP/SMAD pathway but independent of STAT3. This mechanism may contribute to continued iron overload in many pathophysiological conditions ultimately causing a vicious cycle of continued hepcidin suppression.
Collapse
Affiliation(s)
- Lin-Na Yu
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg 69121, Germany
| | - Shi-Jin Wang
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg 69121, Germany
| | - Cheng Chen
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg 69121, Germany
| | - Vanessa Rausch
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg 69121, Germany
| | - Omar Elshaarawy
- Department of Hepatology, Gastroenterology and Liver Transplantation, National Liver Institute, Menoufia University, Shebine Elkom 35121, El Salvador
- Department of Gastroenterology, Royal Liverpool University Hospital, Liverpool L7 8XP, United Kingdom
| | - Sebastian Mueller
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg 69121, Germany
| |
Collapse
|
13
|
Chen C, Wang S, Yu L, Mueller J, Fortunato F, Rausch V, Mueller S. H 2O 2-mediated autophagy during ethanol metabolism. Redox Biol 2021; 46:102081. [PMID: 34343907 PMCID: PMC8350071 DOI: 10.1016/j.redox.2021.102081] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/15/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Alcoholic liver disease (ALD) is the most common liver disease worldwide and its underlying molecular mechanisms are still poorly understood. Moreover, conflicting data have been reported on potentially protective autophagy, the exact role of ethanol-metabolizing enzymes and ROS. METHODS Expression of LC3B, CYP2E1, and NOX4 was studied in a mouse model of acute ethanol exposure by immunoblotting and immunohistochemistry. Autophagy was further studied in primary mouse hepatocytes and huh7 cells in response to ethanol and its major intermediator acetaldehyde. Experiments were carried out in cells overexpressing CYP2E1 and knock down of NOX4 using siRNA. The response to external H2O2 was studied by using the GOX/CAT system. Autophagic flux was monitored using the mRFP-GFP-LC3 plasmid, while rapamycin and chloroquine served as positive and negative controls. RESULTS Acute ethanol exposure of mice over 24 h significantly induced autophagy as measured by LC3B expression but also induced the ROS-generating CYP2E1 and NOX4 enzymes. Notably, ethanol but not its downstream metabolite acetaldehyde induced autophagy in primary mouse hepatocytes. In contrast, autophagy could only be induced in huh7 cells in the presence of overexpressed CYP2E1. In addition, overexpression of NOX4 also significantly increased autophagy, which could be blocked by siRNA mediated knock down. The antioxidant N-acetylcysteine (NAC) also efficiently blocked CYP2E1-and NOX4-mediated induction of autophagy. Finally, specific and non-toxic production of H2O2 by the GOX/CAT system as evidenced by elevated peroxiredoxin (Prx-2) also induced LC3B which was efficiently blocked by NAC. H2O2 strongly increased the autophagic flux as measured by mRFP-GFP-LC3 plasmid. CONCLUSION We here provide evidence that short-term ethanol exposure induces autophagy in hepatocytes both in vivo and in vitro through the generation of ROS. These data suggest that suppression of autophagy by ethanol is most likely due to longer alcohol exposure during chronic alcohol consumption with the accumulation of e.g. misfolded proteins.
Collapse
Affiliation(s)
- Cheng Chen
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Shijin Wang
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Linna Yu
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Johannes Mueller
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Franco Fortunato
- Department of Surgery, University of Heidelberg, Heidelberg, Germany
| | - Vanessa Rausch
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Sebastian Mueller
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
14
|
Probst S, Fels J, Scharner B, Wolff NA, Roussa E, van Swelm RPL, Lee WK, Thévenod F. Role of hepcidin in oxidative stress and cell death of cultured mouse renal collecting duct cells: protection against iron and sensitization to cadmium. Arch Toxicol 2021; 95:2719-2735. [PMID: 34181029 PMCID: PMC8298330 DOI: 10.1007/s00204-021-03106-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/17/2021] [Indexed: 11/05/2022]
Abstract
The liver hormone hepcidin regulates systemic iron homeostasis. Hepcidin is also expressed by the kidney, but exclusively in distal nephron segments. Several studies suggest hepcidin protects against kidney damage involving Fe2+ overload. The nephrotoxic non-essential metal ion Cd2+ can displace Fe2+ from cellular biomolecules, causing oxidative stress and cell death. The role of hepcidin in Fe2+ and Cd2+ toxicity was assessed in mouse renal cortical [mCCD(cl.1)] and inner medullary [mIMCD3] collecting duct cell lines. Cells were exposed to equipotent Cd2+ (0.5-5 μmol/l) and/or Fe2+ (50-100 μmol/l) for 4-24 h. Hepcidin (Hamp1) was transiently silenced by RNAi or overexpressed by plasmid transfection. Hepcidin or catalase expression were evaluated by RT-PCR, qPCR, immunoblotting or immunofluorescence microscopy, and cell fate by MTT, apoptosis and necrosis assays. Reactive oxygen species (ROS) were detected using CellROX™ Green and catalase activity by fluorometry. Hepcidin upregulation protected against Fe2+-induced mIMCD3 cell death by increasing catalase activity and reducing ROS, but exacerbated Cd2+-induced catalase dysfunction, increasing ROS and cell death. Opposite effects were observed with Hamp1 siRNA. Similar to Hamp1 silencing, increased intracellular Fe2+ prevented Cd2+ damage, ROS formation and catalase disruption whereas chelation of intracellular Fe2+ with desferrioxamine augmented Cd2+ damage, corresponding to hepcidin upregulation. Comparable effects were observed in mCCD(cl.1) cells, indicating equivalent functions of renal hepcidin in different collecting duct segments. In conclusion, hepcidin likely binds Fe2+, but not Cd2+. Because Fe2+ and Cd2+ compete for functional binding sites in proteins, hepcidin affects their free metal ion pools and differentially impacts downstream processes and cell fate.
Collapse
Affiliation(s)
- Stephanie Probst
- Faculty of Health, Institute of Physiology, Pathophysiology and Toxicology and ZBAF (Centre for Biomedical Education and Research), School of Medicine, Witten/Herdecke University, Stockumer Str 12 (Thyssenhaus), 58453, Witten, Germany
| | - Johannes Fels
- Faculty of Health, Institute of Physiology, Pathophysiology and Toxicology and ZBAF (Centre for Biomedical Education and Research), School of Medicine, Witten/Herdecke University, Stockumer Str 12 (Thyssenhaus), 58453, Witten, Germany
| | - Bettina Scharner
- Faculty of Health, Institute of Physiology, Pathophysiology and Toxicology and ZBAF (Centre for Biomedical Education and Research), School of Medicine, Witten/Herdecke University, Stockumer Str 12 (Thyssenhaus), 58453, Witten, Germany
| | - Natascha A Wolff
- Faculty of Health, Institute of Physiology, Pathophysiology and Toxicology and ZBAF (Centre for Biomedical Education and Research), School of Medicine, Witten/Herdecke University, Stockumer Str 12 (Thyssenhaus), 58453, Witten, Germany
| | - Eleni Roussa
- Department of Molecular Embryology, Faculty of Medicine, Institute of Anatomy and Cell Biology, University of Freiburg, Albertstr. 17, 79104, Freiburg, Germany
| | - Rachel P L van Swelm
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
| | - Wing-Kee Lee
- Faculty of Health, Institute of Physiology, Pathophysiology and Toxicology and ZBAF (Centre for Biomedical Education and Research), School of Medicine, Witten/Herdecke University, Stockumer Str 12 (Thyssenhaus), 58453, Witten, Germany
- AG Physiology and Pathophysiology of Cells and Membranes, Medical School OWL, Bielefeld University, Morgenbreede 1, 33615, Bielefeld, Germany
| | - Frank Thévenod
- Faculty of Health, Institute of Physiology, Pathophysiology and Toxicology and ZBAF (Centre for Biomedical Education and Research), School of Medicine, Witten/Herdecke University, Stockumer Str 12 (Thyssenhaus), 58453, Witten, Germany.
| |
Collapse
|
15
|
van der Post S, Birchenough GMH, Held JM. NOX1-dependent redox signaling potentiates colonic stem cell proliferation to adapt to the intestinal microbiota by linking EGFR and TLR activation. Cell Rep 2021; 35:108949. [PMID: 33826887 PMCID: PMC10327654 DOI: 10.1016/j.celrep.2021.108949] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 01/25/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
The colon epithelium is a primary point of interaction with the microbiome and is regenerated by a few rapidly cycling colonic stem cells (CSCs). CSC self-renewal and proliferation are regulated by growth factors and the presence of bacteria. However, the molecular link connecting the diverse inputs that maintain CSC homeostasis remains largely unknown. We report that CSC proliferation is mediated by redox-dependent activation of epidermal growth factor receptor (EGFR) signaling via NADPH oxidase 1 (NOX1). NOX1 expression is CSC specific and is restricted to proliferative CSCs. In the absence of NOX1, CSCs fail to generate ROS and have a reduced proliferation rate. NOX1 expression is regulated by Toll-like receptor activation in response to the microbiota and serves to link CSC proliferation with the presence of bacterial components in the crypt. The TLR-NOX1-EGFR axis is therefore a critical redox signaling node in CSCs facilitating the quiescent-proliferation transition and responds to the microbiome to maintain colon homeostasis.
Collapse
Affiliation(s)
- Sjoerd van der Post
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA; Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - George M H Birchenough
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Jason M Held
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA; Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA; Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
16
|
Cancer Related Anemia: An Integrated Multitarget Approach and Lifestyle Interventions. Nutrients 2021; 13:nu13020482. [PMID: 33535496 PMCID: PMC7912724 DOI: 10.3390/nu13020482] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer is often accompanied by worsening of the patient's iron profile, and the resulting anemia could be a factor that negatively impacts antineoplastic treatment efficacy and patient survival. The first line of therapy is usually based on oral or intravenous iron supplementation; however, many patients remain anemic and do not respond. The key might lie in the pathogenesis of the anemia itself. Cancer-related anemia (CRA) is characterized by a decreased circulating serum iron concentration and transferrin saturation despite ample iron stores, pointing to a more complex problem related to iron homeostatic regulation and additional factors such as chronic inflammatory status. This review explores our current understanding of iron homeostasis in cancer, shedding light on the modulatory role of hepcidin in intestinal iron absorption, iron recycling, mobilization from liver deposits, and inducible regulators by infections and inflammation. The underlying relationship between CRA and systemic low-grade inflammation will be discussed, and an integrated multitarget approach based on nutrition and exercise to improve iron utilization by reducing low-grade inflammation, modulating the immune response, and supporting antioxidant mechanisms will also be proposed. Indeed, a Mediterranean-based diet, nutritional supplements and exercise are suggested as potential individualized strategies and as a complementary approach to conventional CRA therapy.
Collapse
|
17
|
Nairz M, Weiss G. Iron in infection and immunity. Mol Aspects Med 2020; 75:100864. [PMID: 32461004 DOI: 10.1016/j.mam.2020.100864] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/25/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022]
Abstract
Iron is an essential micronutrient for virtually all living cells. In infectious diseases, both invading pathogens and mammalian cells including those of the immune system require iron to sustain their function, metabolism and proliferation. On the one hand, microbial iron uptake is linked to the virulence of most human pathogens. On the other hand, the sequestration of iron from bacteria and other microorganisms is an efficient strategy of host defense in line with the principles of 'nutritional immunity'. In an acute infection, host-driven iron withdrawal inhibits the growth of pathogens. Chronic immune activation due to persistent infection, autoimmune disease or malignancy however, sequesters iron not only from infectious agents, autoreactive lymphocytes and neoplastic cells but also from erythroid progenitors. This is one of the key mechanisms which collectively result in the anemia of chronic inflammation. In this review, we highlight the most important interconnections between iron metabolism and immunity, focusing on host defense against relevant infections and on the clinical consequences of anemia of inflammation.
Collapse
Affiliation(s)
- Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Austria.
| |
Collapse
|
18
|
Nienaber A, Baumgartner J, Dolman RC, Ozturk M, Zandberg L, Hayford FEA, Brombacher F, Blaauw R, Parihar SP, Smuts CM, Malan L. Omega-3 Fatty Acid and Iron Supplementation Alone, but Not in Combination, Lower Inflammation and Anemia of Infection in Mycobacterium tuberculosis-Infected Mice. Nutrients 2020; 12:E2897. [PMID: 32971969 PMCID: PMC7551947 DOI: 10.3390/nu12092897] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
Progressive inflammation and anemia are common in tuberculosis (TB) and linked to poor clinical outcomes. Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have inflammation-resolving properties, whereas iron supplementation in TB may have limited efficacy and enhance bacterial growth. We investigated effects of iron and EPA/DHA supplementation, alone and in combination, on inflammation, anemia, iron status markers and clinical outcomes in Mycobacterium tuberculosis-infected C3HeB/FeJ mice. One week post-infection, mice received the AIN-93 diet without (control) or with supplemental iron (Fe), EPA/DHA, or Fe+EPA/DHA for 3 weeks. Mice supplemented with Fe or EPA/DHA had lower soluble transferrin receptor, ferritin and hepcidin than controls, but these effects were attenuated in Fe+EPA/DHA mice. EPA/DHA increased inflammation-resolving lipid mediators and lowered lung IL-1α, IFN-γ, plasma IL-1β, and TNF-α. Fe lowered lung IL-1α, IL-1β, plasma IL-1β, TNF-α, and IL-6. However, the cytokine-lowering effects in the lungs were attenuated with Fe+EPA/DHA. Mice supplemented with EPA/DHA had lower lung bacterial loads than controls, but this effect was attenuated in Fe+EPA/DHA mice. Thus, individually, post-infection EPA/DHA and iron supplementation lowered systemic and lung inflammation and mitigated anemia of infection in TB, but not when combined. EPA/DHA also enhanced bactericidal effects and could support inflammation resolution and management of anemia.
Collapse
Affiliation(s)
- Arista Nienaber
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa; (J.B.); (R.C.D.); (L.Z.); (F.E.A.H.); (C.M.S.); (L.M.)
| | - Jeannine Baumgartner
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa; (J.B.); (R.C.D.); (L.Z.); (F.E.A.H.); (C.M.S.); (L.M.)
- Laboratory of Human Nutrition, ETH, 8092 Zurich, Switzerland
| | - Robin C. Dolman
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa; (J.B.); (R.C.D.); (L.Z.); (F.E.A.H.); (C.M.S.); (L.M.)
| | - Mumin Ozturk
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, University of Cape Town, Cape Town 7925, South Africa; (M.O.); (F.B.); (S.P.P.)
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, University of Cape Town, Cape Town 7925, South Africa
| | - Lizelle Zandberg
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa; (J.B.); (R.C.D.); (L.Z.); (F.E.A.H.); (C.M.S.); (L.M.)
| | - Frank E. A. Hayford
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa; (J.B.); (R.C.D.); (L.Z.); (F.E.A.H.); (C.M.S.); (L.M.)
- Department of Nutrition and Dietetics, School of biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra Box KB143, Ghana
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, University of Cape Town, Cape Town 7925, South Africa; (M.O.); (F.B.); (S.P.P.)
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, University of Cape Town, Cape Town 7925, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa
| | - Renee Blaauw
- Division of Human Nutrition, Stellenbosch University, Tygerberg, Cape Town 7505, South Africa;
| | - Suraj P. Parihar
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town-Component, University of Cape Town, Cape Town 7925, South Africa; (M.O.); (F.B.); (S.P.P.)
- Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, University of Cape Town, Cape Town 7925, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town 7925, South Africa
- Division of Medical Microbiology, Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Cornelius M. Smuts
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa; (J.B.); (R.C.D.); (L.Z.); (F.E.A.H.); (C.M.S.); (L.M.)
| | - Linda Malan
- Centre of Excellence for Nutrition, North-West University, Potchefstroom 2531, South Africa; (J.B.); (R.C.D.); (L.Z.); (F.E.A.H.); (C.M.S.); (L.M.)
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Erythropoiesis is a complex multistep process going from committed erythroid progenitors to mature red cells. Although recent advances allow the characterization of some components of erythropoiesis, much still remains to be investigated particularly on stress erythropoiesis. This review summarizes recent progresses made to understand the impact of oxidative stress on normal and pathologic erythropoiesis. RECENT FINDINGS During erythroid maturation, reactive oxygen species might function as second messenger through either transient oxidation of cysteine residues on signaling targets or modulation of intracellular signaling pathways. Thus, in erythropoiesis, efficient cytoprotective systems are required to limit possible reactive oxygen species-related toxic effects especially in stress erythropoiesis characterized by severe oxidation such as β-thalassemia. In addition, prolonged or severe oxidative stress impairs autophagy, which might contribute to the block of erythroid maturation in stress erythropoiesis. Understanding the functional role of cytoprotective systems such as peroxiredoxin-2 or classical molecular chaperones such as the heat shock proteins will contribute to develop innovative therapeutic strategies for ineffective erythropoiesis. SUMMARY We provide an update on cytoprotective mechanisms against oxidation in normal and stress erythropoiesis. We discuss the role of oxidative sensors involved in modulation of intracellular signaling during erythroid maturation process in normal and stress erythropoiesis.
Collapse
|
20
|
Mohammed F, Gorla M, Bisoyi V, Tammineni P, Sepuri NBV. Rotenone‐induced reactive oxygen species signal the recruitment of STAT3 to mitochondria. FEBS Lett 2020; 594:1403-1412. [DOI: 10.1002/1873-3468.13741] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 01/07/2020] [Indexed: 12/18/2022]
Affiliation(s)
| | - Madhavi Gorla
- Department of Biochemistry University of Hyderabad India
| | - Vandana Bisoyi
- Department of Biochemistry University of Hyderabad India
| | | | | |
Collapse
|
21
|
Griess B, Mir S, Datta K, Teoh-Fitzgerald M. Scavenging reactive oxygen species selectively inhibits M2 macrophage polarization and their pro-tumorigenic function in part, via Stat3 suppression. Free Radic Biol Med 2020; 147:48-60. [PMID: 31863907 PMCID: PMC10035558 DOI: 10.1016/j.freeradbiomed.2019.12.018] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/21/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022]
Abstract
Tumor associated macrophages (TAM) enhance the aggressiveness of breast cancer via promoting cancer cell growth, metastasis, and suppression of the patient's immune system. These TAMs are polarized in breast cancer with features more closely resembling the pro-tumorigenic and immunosuppressive M2 type rather than the anti-tumor and pro-inflammatory M1 type. The goal of our study was to examine primary human monocyte-derived M1 and M2 macrophages for key redox differences and determine sensitivities of these macrophages to the redox-active drug, MnTE-2-PyP5+. This compound reduced levels of M2 markers and inhibited their ability to promote cancer cell growth and suppress T cell activation. The surface levels of the T cell suppressing molecule, PD-L2, were reduced by MnTE-2-PyP5+ in a dose-dependent manner. This study also examined key differences in ROS generation and scavenging between M1 and M2 macrophages. Our results indicate that M2 macrophages have lower levels of reactive oxygen species (ROS) and lower production of extracellular hydrogen peroxide compared to the M1 macrophages. These differences are due in part to reduced expression levels of pro-oxidants, Nox2, Nox5, and the non-enzymatic members of the Nox complex, p22phox and p47phox, as well as higher levels of antioxidant enzymes, Cu/ZnSOD, Gpx1, and catalase. More importantly, we found that despite having lower ROS levels, M2 macrophages require ROS for proper polarization, as addition of hydrogen peroxide increased M2 markers. These TAM-like macrophages are also more sensitive to the ROS modulator and a pan-Nox inhibitor. Both MnTE-2-PyP5+ and DPI inhibited expression levels of M2 marker genes. We have further shown that this inhibition was partly mediated through a decrease in Stat3 activation during IL4-induced M2 polarization. Overall, this study reveals key redox differences between M1 and M2 primary human macrophages and that redox-active drugs can be used to inhibit the pro-tumor and immunosuppressive phenotype of TAM-like M2 macrophages. This study also provides rationale for combining MnTE-2-PyP5+ with immunotherapies.
Collapse
Affiliation(s)
- Brandon Griess
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Shakeel Mir
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Melissa Teoh-Fitzgerald
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
22
|
Hamano H, Ikeda Y, Watanabe H, Horinouchi Y, Izawa-Ishizawa Y, Imanishi M, Zamami Y, Takechi K, Miyamoto L, Ishizawa K, Tsuchiya K, Tamaki T. The uremic toxin indoxyl sulfate interferes with iron metabolism by regulating hepcidin in chronic kidney disease. Nephrol Dial Transplant 2019; 33:586-597. [PMID: 28992067 DOI: 10.1093/ndt/gfx252] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 06/24/2017] [Indexed: 12/11/2022] Open
Abstract
Background Hepcidin secreted by hepatocytes is a key regulator of iron metabolism throughout the body. Hepcidin concentrations are increased in chronic kidney disease (CKD), contributing to abnormalities in iron metabolism. Levels of indoxyl sulfate (IS), a uremic toxin, are also elevated in CKD. However, the effect of IS accumulation on iron metabolism remains unclear. Methods We used HepG2 cells to determine the mechanism by which IS regulates hepcidin concentrations. We also used a mouse model of adenine-induced CKD. The CKD mice were divided into two groups: one was treated using AST-120 and the other received no treatment. We examined control mice, CKD mice, CKD mice treated using AST-120 and mice treated with IS via drinking water. Results In the in vitro experiments using HepG2 cells, IS increased hepcidin expression in a dose-dependent manner. Silencing of the aryl hydrocarbon receptor (AhR) inhibited IS-induced hepcidin expression. Furthermore, IS induced oxidative stress and antioxidant drugs diminished IS-induced hepcidin expression. Adenine-induced CKD mice demonstrated an increase in hepcidin concentrations; this increase was reduced by AST-120, an oral adsorbent of the uremic toxin. CKD mice showed renal anemia, decreased plasma iron concentration, increased plasma ferritin and increased iron content in the spleen. Ferroportin was decreased in the duodenum and increased in the spleen. These changes were ameliorated by AST-120 treatment. Mice treated by direct IS administration showed hepatic hepcidin upregulation. Conclusions IS affects iron metabolism in CKD by participating in hepcidin regulation via pathways that depend on AhR and oxidative stress.
Collapse
Affiliation(s)
- Hirofumi Hamano
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.,Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Yasumasa Ikeda
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hiroaki Watanabe
- Department of Clinical Pharmacology and Therapeutics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yuya Horinouchi
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yuki Izawa-Ishizawa
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Masaki Imanishi
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Yoshito Zamami
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan.,Department of Clinical Pharmacology and Therapeutics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Kenshi Takechi
- Clinical Trial Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | - Licht Miyamoto
- Department of Medical Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Keisuke Ishizawa
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan.,Department of Clinical Pharmacology and Therapeutics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Koichiro Tsuchiya
- Department of Medical Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Toshiaki Tamaki
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
23
|
Cui Q, Chen FY, Chen HY, Peng H, Wang KJ. Benzo[a]pyrene (BaP) exposure generates persistent reactive oxygen species (ROS) to inhibit the NF-κB pathway in medaka (Oryzias melastigma). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 251:502-509. [PMID: 31103010 DOI: 10.1016/j.envpol.2019.04.063] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/04/2019] [Accepted: 04/12/2019] [Indexed: 06/09/2023]
Abstract
Benzo[a]pyrene (BaP), a common environmental pollutant, can modulate the immune-associated signal pathway NF-κB, which is one of the critical signal pathways involved in various immune responses. BaP exposure usually generates reactive oxygen species (ROS), but whether ROS are predominantly involved in the modulation mechanism of the NF-κB pathway has not been clearly understood. In this study, an in vivo examination of Oryzias melastigma demonstrated that BaP exposure led to a down-regulation of the NF-κB pathway and increased levels of ROS. Conversely, in vitro results using the medaka liver cell line DIT-29 and a widely applied H2O2 method showed the opposite: up-regulation of the NF-κB pathway. However, the down-regulation of NF-κB upon BaP exposure in vitro was inhibited by the addition of a ROS inhibitor, indicating ROS are involved in the modulation of NF-κB. The discrepancy between in vivo and in vitro results of ROS impacts on NF-κB activation might be related to the concentration and persistence of ROS. Using a modified luminol detection system, BaP was found to generate sustained physiological concentrations of ROS for 24 h, while an H2O2 bolus generated ROS for less than 30 min. Furthermore, a steady-state sub-micromolar H2O2 system (H2O2ss) was developed in parallel as a positive control of ROS, by which H2O2 could be maintained for 24 h. Comparative evaluation using H2O2, H2O2ss and BaP exposures on the medaka cell line with pGL4.32 demonstrated that the persistent physiological concentrations of ROS generated upon BaP exposure or treatment with H2O2ss inhibited the NF-κB pathway, but direct H2O2 exposure had the opposite effect. Moreover, a western-blot assay and EMSA detection further confirmed the modulation of the NF-κB pathway in DIT-29. Taken together, this study shows that BaP exposure inhibits the NF-κB pathway by generating sustained physiological concentrations of ROS.
Collapse
Affiliation(s)
- Qian Cui
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Science, Xiamen University, Xiamen, Fujian, China
| | - Fang-Yi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Science, Xiamen University, Xiamen, Fujian, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, Fujian, China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Xiamen University, Xiamen, Fujian, China
| | - Hui-Yun Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Science, Xiamen University, Xiamen, Fujian, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, Fujian, China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Xiamen University, Xiamen, Fujian, China
| | - Hui Peng
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Science, Xiamen University, Xiamen, Fujian, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, Fujian, China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Xiamen University, Xiamen, Fujian, China
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Science, Xiamen University, Xiamen, Fujian, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, Fujian, China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
24
|
IL-1 beta-mediated macrophage-hepatocyte crosstalk upregulates hepcidin under physiological low oxygen levels. Redox Biol 2019; 24:101209. [PMID: 31108461 PMCID: PMC6526398 DOI: 10.1016/j.redox.2019.101209] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/25/2019] [Accepted: 04/27/2019] [Indexed: 12/19/2022] Open
Abstract
In mammals, the iron masterswitch hepcidin efficiently controls iron recycling by the macrophage-liver axis but the exact interplay between macrophages and hepatocytes remains poorly understood. We here study hepcidin response during macrophage differentiation as well as the macrophage-hepatocyte crosstalk and its subsequent effects on hepatocyte hepcidin using an in vitro co-culture model that mimics the physiological liver microenvironment. We show that macrophage differentiation strongly induces hepcidin by 60-fold both in THP1 macrophages and primary isolated monocyte-derived macrophages. Removal of H2O2 by catalase or inhibition of NOX2 efficiently blocked hepcidin induction. After differentiation, macrophage hepcidin accounted for 10% of total hepatocyte hepcidin and did not respond to low oxygen levels. In contrast, co-culture of differentiated macrophages with Huh7 cells significantly induced hepatocyte hepcidin, which was further potentiated under low oxygen levels. Hepatocyte hepcidin was also upregulated when Huh7 cells were solely exposed to macrophage-conditioned hypoxic medium. A cytokine screen identified macrophage secreted IL-1β as major inducer of hepcidin in hepatocytes. In confirmation, treatment of Huh7 cells with the IL-1 receptor antagonist (anakinra) completely blunted macrophage-mediated hepcidin transcription in hepatocytes. Finally, detailed analysis of potentially involved signaling pathways points toward STAT3 and CEBPδ-mediated hepcidin induction independent of IL-6. In conclusion, our study demonstrates a strong NOX2-mediated hepcidin induction during macrophage differentiation. These differentiated macrophages are able to efficiently induce hepatocyte hepcidin mainly through secretion of IL-1β. Our data highlight a hitherto unrecognized role of macrophage-hepatocyte crosstalk for a joint and oxygen-dependent hepcidin production through STAT3 and CEBPδ. Hepcidin is strongly induced during NOX2-mediated macrophage differentiation in a H2O2-dependent manner. In contrast to hepatocyte hepcidin, macrophage hepcidin transcription is not modulated by low O2 level. Macrophage released IL1-β strongly induces hepatocyte hepcidin via STAT3 signaling. IL1-β mediated hepatocyte hepcidin induction is independent of IL-6. Despite the mandatory requirement of STAT3, CEBPδ also involved in IL1-β induced hepatocyte hepcidin transcription.
Collapse
|
25
|
Morris G, Berk M, Maes M, Puri BK. Could Alzheimer's Disease Originate in the Periphery and If So How So? Mol Neurobiol 2019; 56:406-434. [PMID: 29705945 PMCID: PMC6372984 DOI: 10.1007/s12035-018-1092-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 04/17/2018] [Indexed: 12/11/2022]
Abstract
The classical amyloid cascade model for Alzheimer's disease (AD) has been challenged by several findings. Here, an alternative molecular neurobiological model is proposed. It is shown that the presence of the APOE ε4 allele, altered miRNA expression and epigenetic dysregulation in the promoter region and exon 1 of TREM2, as well as ANK1 hypermethylation and altered levels of histone post-translational methylation leading to increased transcription of TNFA, could variously explain increased levels of peripheral and central inflammation found in AD. In particular, as a result of increased activity of triggering receptor expressed on myeloid cells 2 (TREM-2), the presence of the apolipoprotein E4 (ApoE4) isoform, and changes in ANK1 expression, with subsequent changes in miR-486 leading to altered levels of protein kinase B (Akt), mechanistic (previously mammalian) target of rapamycin (mTOR) and signal transducer and activator of transcription 3 (STAT3), all of which play major roles in microglial activation, proliferation and survival, there is activation of microglia, leading to the subsequent (further) production of cytokines, chemokines, nitric oxide, prostaglandins, reactive oxygen species, inducible nitric oxide synthase and cyclooxygenase-2, and other mediators of inflammation and neurotoxicity. These changes are associated with the development of amyloid and tau pathology, mitochondrial dysfunction (including impaired activity of the electron transport chain, depleted basal mitochondrial potential and oxidative damage to key tricarboxylic acid enzymes), synaptic dysfunction, altered glycogen synthase kinase-3 (GSK-3) activity, mTOR activation, impairment of autophagy, compromised ubiquitin-proteasome system, iron dyshomeostasis, changes in APP translation, amyloid plaque formation, tau hyperphosphorylation and neurofibrillary tangle formation.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Level 1 North, Main Block, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
- Florey Institute for Neuroscience and Mental Health, Kenneth Myer Building, University of Melbourne, 30 Royal Parade, Parkville, Victoria, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, 35 Poplar Rd, Parkville, Victoria, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK.
| |
Collapse
|
26
|
Cui Q, Chen FY, Zhang M, Peng H, Wang KJ. Transcriptomic analysis revealing hepcidin expression in Oryzias melastigma regulated through the JAK-STAT signaling pathway upon exposure to BaP. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 206:134-141. [PMID: 30476743 DOI: 10.1016/j.aquatox.2018.11.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 06/09/2023]
Abstract
Our previous study revealed that an antimicrobial peptide hepcidin, can be significantly up-regulated either with LPS challenge or upon exposure to Benzo[a]pyrene (BaP) in red sea bream, but the molecular mechanism involved in whether the transcriptional expression of hepcidin induced by LPS or BaP is regulated through a similar signaling pathway is not yet known. To elucidate the underlying molecular mechanism, the marine model fish Oryzias melastigma was exposed to 1 μg/L BaP as well as challenged with 5 μg of LPS per fish. Samples at 3 h post-LPS challenge, and 2 d and 3 d post-BaP exposure were separately collected for transcriptome analysis. General analysis of the predicted immune-associated unigenes based on the transcriptomic data showed that the percentages of modulated immune-associated genes were 7% with LPS challenge, and 3% and 7% with BaP exposure at 2 and 3 days, respectively. Genes involved in functions like antimicrobial activity, neutrophil activation, and leukocyte chemotaxis were up-regulated with LPS challenge, whereas more than half of the immune associated genes including the KLF family were down-regulated upon BaP exposure, indicating a difference in the modulated immune genes between LPS challenge and BaP exposure. Specific comparative analyses of the immune-associated signal pathways NOD, TOLL, NF-κB and JAK-STAT with LPS challenge or upon exposure to BaP, indicated that most of the modulated genes in association with the NOD, TOLL and NF-κB pathways were induced with LPS challenge but only a few after exposure to BaP, suggesting that BaP exposure was generally not associated with any of the three signal pathways. Interestingly, further transcriptomic analysis revealed that 5 of the 8 modulated genes associated with the JAK-STAT pathway were down-regulated, while 2 inhibiting genes were up-regulated after BaP exposure for 2 days whereas LPS challenge resulted in only less than half modulated, suggesting the possibility of down-regulation caused by BaP exposure through JAK-STAT pathway. Further testing using an EPC cell culture demonstrated that expression of the hepcidin1 gene was less involved in the known signal pathways, such as c/EBP, BMP, and NF-κB, but instead mostly in association with the JAK-STAT pathway upon BaP exposure.
Collapse
Affiliation(s)
- Qian Cui
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Science, Xiamen University, Xiamen, Fujian, China
| | - Fang-Yi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Science, Xiamen University, Xiamen, Fujian, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, Fujian, China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Xiamen University, Xiamen, Fujian, China
| | - Min Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Science, Xiamen University, Xiamen, Fujian, China
| | - Hui Peng
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Science, Xiamen University, Xiamen, Fujian, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, Fujian, China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Xiamen University, Xiamen, Fujian, China
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Science, Xiamen University, Xiamen, Fujian, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, Xiamen University, Xiamen, Fujian, China; Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
27
|
Pfeifhofer-Obermair C, Tymoszuk P, Petzer V, Weiss G, Nairz M. Iron in the Tumor Microenvironment-Connecting the Dots. Front Oncol 2018; 8:549. [PMID: 30534534 PMCID: PMC6275298 DOI: 10.3389/fonc.2018.00549] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/06/2018] [Indexed: 12/18/2022] Open
Abstract
Iron metabolism and tumor biology are intimately linked. Iron facilitates the production of oxygen radicals, which may either result in iron-induced cell death, ferroptosis, or contribute to mutagenicity and malignant transformation. Once transformed, malignant cells require high amounts of iron for proliferation. In addition, iron has multiple regulatory effects on the immune system, thus affecting tumor surveillance by immune cells. For these reasons, inconsiderate iron supplementation in cancer patients has the potential of worsening disease course and outcome. On the other hand, chronic immune activation in the setting of malignancy alters systemic iron homeostasis and directs iron fluxes into myeloid cells. While this response aims at withdrawing iron from tumor cells, it may impair the effector functions of tumor-associated macrophages and will result in iron-restricted erythropoiesis and the development of anemia, subsequently. This review summarizes our current knowledge of the interconnections of iron homeostasis with cancer biology, discusses current clinical controversies in the treatment of anemia of cancer and focuses on the potential roles of iron in the solid tumor microenvironment, also speculating on yet unknown molecular mechanisms.
Collapse
Affiliation(s)
- Christa Pfeifhofer-Obermair
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Verena Petzer
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
28
|
Chaiswing L, St. Clair WH, St. Clair DK. Redox Paradox: A Novel Approach to Therapeutics-Resistant Cancer. Antioxid Redox Signal 2018; 29:1237-1272. [PMID: 29325444 PMCID: PMC6157438 DOI: 10.1089/ars.2017.7485] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 01/05/2018] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Cancer cells that are resistant to radiation and chemotherapy are a major problem limiting the success of cancer therapy. Aggressive cancer cells depend on elevated intracellular levels of reactive oxygen species (ROS) to proliferate, self-renew, and metastasize. As a result, these aggressive cancers maintain high basal levels of ROS compared with normal cells. The prominence of the redox state in cancer cells led us to consider whether increasing the redox state to the condition of oxidative stress could be used as a successful adjuvant therapy for aggressive cancers. Recent Advances: Past attempts using antioxidant compounds to inhibit ROS levels in cancers as redox-based therapy have met with very limited success. However, recent clinical trials using pro-oxidant compounds reveal noteworthy results, which could have a significant impact on the development of strategies for redox-based therapies. CRITICAL ISSUES The major objective of this review is to discuss the role of the redox state in aggressive cancers and how to utilize the shift in redox state to improve cancer therapy. We also discuss the paradox of redox state parameters; that is, hydrogen peroxide (H2O2) as the driver molecule for cancer progression as well as a target for cancer treatment. FUTURE DIRECTIONS Based on the biological significance of the redox state, we postulate that this system could potentially be used to create a new avenue for targeted therapy, including the potential to incorporate personalized redox therapy for cancer treatment.
Collapse
Affiliation(s)
- Luksana Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky-Lexington, Lexington, Kentucky
| | - William H. St. Clair
- Department of Radiation Medicine, University of Kentucky-Lexington, Lexington, Kentucky
| | - Daret K. St. Clair
- Department of Toxicology and Cancer Biology, University of Kentucky-Lexington, Lexington, Kentucky
| |
Collapse
|
29
|
Madeddu C, Gramignano G, Astara G, Demontis R, Sanna E, Atzeni V, Macciò A. Pathogenesis and Treatment Options of Cancer Related Anemia: Perspective for a Targeted Mechanism-Based Approach. Front Physiol 2018; 9:1294. [PMID: 30294279 PMCID: PMC6159745 DOI: 10.3389/fphys.2018.01294] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 08/28/2018] [Indexed: 01/28/2023] Open
Abstract
Cancer-related anemia (CRA) is a common sign occurring in more than 30% of cancer patients at diagnosis before the initiation of antineoplastic therapy. CRA has a relevant influence on survival, disease progression, treatment efficacy, and the patients' quality of life. It is more often detected in patients with advanced stage disease, where it represents a specific symptom of the neoplastic disease, as a consequence of chronic inflammation. In fact, CRA is characterized by biological and hematologic features that resemble those described in anemia associated to chronic inflammatory disease. Proinflammatory cytokine, mainly IL-6, which are released by both tumor and immune cells, play a pivotal action in CRA etiopathogenesis: they promote alterations in erythroid progenitor proliferation, erythropoietin (EPO) production, survival of circulating erythrocytes, iron balance, redox status, and energy metabolism, all of which can lead to anemia. The discovery of hepcidin allowed a greater knowledge of the relationships between immune cells, iron metabolism, and anemia in chronic inflammatory diseases. Additionally, chronic inflammation influences a compromised nutritional status, which in turn might induce or contribute to CRA. In the present review we examine the multifactorial pathogenesis of CRA discussing the main and novel mechanisms by which immune, nutritional, and metabolic components affect its onset and severity. Moreover, we analyze the status of the art and the perspective for the treatment of CRA. Notably, despite the high incidence and clinical relevance of CRA, controlled clinical studies testing the most appropriate treatment for CRA are scarce, and its management in clinical practice remains challenging. The present review may be useful to indicate the development of an effective approach based on a detailed assessment of all factors potentially involved in the pathogenesis of CRA. This mechanism-based approach is essential for clinicians to plan a safe, targeted, and successful therapy, thereby promoting a relevant amelioration of patients' quality of life.
Collapse
Affiliation(s)
- Clelia Madeddu
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | - Giorgio Astara
- Department of Medical Oncology, Azienda Ospedaliero Universitaria Cagliari, Cagliari, Italy
| | - Roberto Demontis
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Elisabetta Sanna
- Department of Gynecologic Oncology, Azienda Ospedaliera Brotzu, Cagliari, Italy
| | - Vinicio Atzeni
- Hospital Medical Management, Azienda Ospedaliera Brotzu, Cagliari, Italy
| | - Antonio Macciò
- Department of Gynecologic Oncology, Azienda Ospedaliera Brotzu, Cagliari, Italy
| |
Collapse
|
30
|
Nairz M, Dichtl S, Schroll A, Haschka D, Tymoszuk P, Theurl I, Weiss G. Iron and innate antimicrobial immunity-Depriving the pathogen, defending the host. J Trace Elem Med Biol 2018; 48:118-133. [PMID: 29773170 DOI: 10.1016/j.jtemb.2018.03.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/25/2018] [Accepted: 03/06/2018] [Indexed: 02/08/2023]
Abstract
The acute-phase response is triggered by the presence of infectious agents and danger signals which indicate hazards for the integrity of the mammalian body. One central feature of this response is the sequestration of iron into storage compartments including macrophages. This limits the availability of this essential nutrient for circulating pathogens, a host defence strategy known as 'nutritional immunity'. Iron metabolism and the immune response are intimately linked. In infections, the availability of iron affects both the efficacy of antimicrobial immune pathways and pathogen proliferation. However, host strategies to withhold iron from microbes vary according to the localization of pathogens: Infections with extracellular bacteria such as Staphylococcus aureus, Streptococcus, Klebsiella or Yersinia stimulate the expression of the iron-regulatory hormone hepcidin which targets the cellular iron-exporter ferroportin-1 causing its internalization and blockade of iron egress from absorptive enterocytes in the duodenum and iron-recycling macrophages. This mechanism disrupts both routes of iron delivery to the circulation, contributes to iron sequestration in the mononuclear phagocyte system and mediates the hypoferraemia of the acute phase response subsequently resulting in the development of anaemia of inflammation. When intracellular microbes are present, other strategies of microbial iron withdrawal are needed. For instance, in macrophages harbouring intracellular pathogens such as Chlamydia, Mycobacterium tuberculosis, Listeria monocytogenes or Salmonella Typhimurium, ferroportin-1-mediated iron export is turned on for the removal of iron from infected cells. This also leads to reduced iron availability for intra-macrophage pathogens which inhibits their growth and in parallel strengthens anti-microbial effector pathways of macrophages including the formation of inducible nitric oxide synthase and tumour necrosis factor. Iron plays a key role in infectious diseases both as modulator of the innate immune response and as nutrient for microbes. We need to gain a more comprehensive understanding of how the body can differentially respond to infection by extra- or intracellular pathogens. This knowledge may allow us to modulate mammalian iron homeostasis pharmaceutically and to target iron-acquisition systems of pathogens, thus enabling us to treat infections with novel strategies that act independent of established antimicrobials.
Collapse
Affiliation(s)
- Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria.
| | - Stefanie Dichtl
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Andrea Schroll
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - David Haschka
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Igor Theurl
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, Austria
| |
Collapse
|
31
|
van Swelm RPL, Vos M, Verhoeven F, Thévenod F, Swinkels DW. Endogenous hepcidin synthesis protects the distal nephron against hemin and hemoglobin mediated necroptosis. Cell Death Dis 2018; 9:550. [PMID: 29749404 PMCID: PMC5945780 DOI: 10.1038/s41419-018-0568-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/30/2018] [Accepted: 04/06/2018] [Indexed: 12/11/2022]
Abstract
Hemoglobinuria is associated with kidney injury in various hemolytic pathologies. Currently, there is no treatment available and its pathophysiology is not completely understood. Here we studied the potential detrimental effects of hemoglobin (Hb) exposure to the distal nephron (DN). Involvement of the DN in Hb kidney injury was suggested by the induction of renal hepcidin synthesis (p < 0.001) in mice repeatedly injected with intravenous Hb. Moreover, the hepcidin induction was associated with a decline in urinary kidney injury markers 24p3/NGAL and KIM1, suggesting a role for hepcidin in protection against Hb kidney injury. We demonstrated that uptake of Hb in the mouse cortical collecting duct cells (mCCDcl1) is mediated by multi-protein ligand receptor 24p3R, as indicated by a significant 90% reduction in Hb uptake (p < 0.001) after 24p3R silencing. Moreover, incubation of mCCDcl1 cells with Hb or hemin for 4 or 24 h resulted in hepcidin synthesis and increased mRNA expression of markers for oxidative, inflammatory and ER stress, but no cell death as indicated by apoptosis staining. A protective role for cellular hepcidin against Hb-induced injury was demonstrated by aggravation of oxidative, inflammatory and ER stress after 4 h Hb or hemin incubation in hepcidin silenced mCCDcl1 cells. Hepcidin silencing potentiated hemin-mediated cell death that could be diminished by co-incubation of Nec-1, suggesting that endogenous hepcidin prevents necroptosis. Combined, these results demonstrate that renal hepcidin synthesis protects the DN against hemin and hemoglobin-mediated injury.
Collapse
Affiliation(s)
- Rachel P L van Swelm
- Department of Laboratory Medicine, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.
| | - Madelon Vos
- Department of Laboratory Medicine, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Frank Verhoeven
- Department of Laboratory Medicine, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Frank Thévenod
- Institute of Physiology, Pathophysiology & Toxicology, Center for Biomedical Training and Research, University of Witten/Herdecke, Witten, Germany
| | - Dorine W Swinkels
- Department of Laboratory Medicine, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
32
|
Silva I, Rausch V, Peccerella T, Millonig G, Seitz HK, Mueller S. Hypoxia enhances H 2O 2-mediated upregulation of hepcidin: Evidence for NOX4-mediated iron regulation. Redox Biol 2018; 16:1-10. [PMID: 29459227 PMCID: PMC5832675 DOI: 10.1016/j.redox.2018.02.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 02/03/2018] [Accepted: 02/10/2018] [Indexed: 12/21/2022] Open
Abstract
The exact regulation of the liver-secreted peptide hepcidin, the key regulator of systemic iron homeostasis, is still poorly understood. It is potently induced by iron, inflammation, cytokines or H2O2 but conflicting results have been reported on hypoxia. In our current study, we first show that pronounced (1%) and mild (5%) hypoxia strongly induces hepcidin in human Huh7 hepatoma and primary liver cells predominantly at the transcriptional level via STAT3 using two hypoxia systems (hypoxia chamber and enzymatic hypoxia by the GOX/CAT system). SiRNA silencing of JAK1, STAT3 and NOX4 diminished the hypoxia-mediated effect while a role of HIF1α could be clearly ruled out by the response to hypoxia-mimetics and competition experiments with a plasmid harboring the oxygen-dependent degradation domain of HIF1α. Specifically, hypoxia drastically enhances the H2O2-mediated induction of hepcidin strongly pointing towards an oxidase as powerful upstream control of hepcidin. We finally provide evidences for an efficient regulation of hepcidin expression by NADPH-dependent oxidase 4 (NOX4) in liver cells. In summary, our data demonstrate that hypoxia strongly potentiates the peroxide-mediated induction of hepcidin via STAT3 signaling pathway. Moreover, oxidases such as NOX4 or artificially overexpressed urate oxidase (UOX) can induce hepcidin. It remains to be studied whether the peroxide-STAT3-hepcidin axis simply acts to continuously compensate for oxygen fluctuations or is directly involved in iron sensing per se. Hypoxia strongly induces hepcidin via STAT3 signaling. HIF1α is not involved in hepcidin regulation under hypoxia. Hypoxia enhances hydrogen peroxide-mediated hepcidin induction. Oxidases, such as NOX4 are powerful inducers of hepcidin.
Collapse
Affiliation(s)
- Inês Silva
- Center for Alcohol Research, University of Heidelberg and Salem Medical Center, Heidelberg, Germany
| | - Vanessa Rausch
- Center for Alcohol Research, University of Heidelberg and Salem Medical Center, Heidelberg, Germany
| | - Teresa Peccerella
- Center for Alcohol Research, University of Heidelberg and Salem Medical Center, Heidelberg, Germany
| | - Gunda Millonig
- Center for Alcohol Research, University of Heidelberg and Salem Medical Center, Heidelberg, Germany
| | - Helmut-Karl Seitz
- Center for Alcohol Research, University of Heidelberg and Salem Medical Center, Heidelberg, Germany
| | - Sebastian Mueller
- Center for Alcohol Research, University of Heidelberg and Salem Medical Center, Heidelberg, Germany.
| |
Collapse
|
33
|
Matte A, De Falco L, Federti E, Cozzi A, Iolascon A, Levi S, Mohandas N, Zamo A, Bruno M, Lebouef C, Janin A, Siciliano A, Ganz T, Federico G, Carlomagno F, Mueller S, Silva I, Carbone C, Melisi D, Kim DW, Choi SY, De Franceschi L. Peroxiredoxin-2: A Novel Regulator of Iron Homeostasis in Ineffective Erythropoiesis. Antioxid Redox Signal 2018; 28:1-14. [PMID: 28793778 DOI: 10.1089/ars.2017.7051] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
AIMS Iron overload (IO) is a life-threatening complication of chronic hemolytic disorders such as β-thalassemia. IO results in severe cellular oxidative damage, leading to organ failure. Peroxiredoxin-2 (Prx2), a typical 2-cysteine-(Cys)-peroxiredoxin, is an important component of the cytoprotective system, but its response to IO is still to be fully defined. RESULTS We studied the effects of IO on Prx2-knockout mice (Prx2-/-). The absence of Prx2 enhanced toxicity due to IO on erythropoiesis. We found that IO failed to induce the typical hepcidin (Hamp) upregulation in Prx2-/- mice due to its failure to activate the signal transducer and activator of transcription-3 (STAT3) with intact Jak2 signaling. In Prx2-/- mice, the loss of Hamp response was also observed after administration of a single dose of oral iron. When lipopolysaccharide (LPS) was used to explore IL6-STAT3 activation in Prx2-/- mice, STAT3 activation and Hamp upregulation were once again defective. Treatment with PEP-fusion-recombinant-Prx2 (PEP Prx2) significantly increased STAT3 activation with upregulation of Hamp expression in both IO- and LPS-exposed Prx2-/- mice. We also confirmed the beneficial effects of PEP Prx2 on Hamp expression through STAT3 activation in β-thalassemic mice. INNOVATION We propose that Prx2 plays a key role in responding to cytotoxicity of IO, directly targeting STAT3-transcriptional factor in a Jak2-independent fashion and regulating Hamp in response to canonical stimuli. CONCLUSION Collectively, our data highlight a novel role of Prx2 in iron homeostasis. Prx2 is a key cytoprotector against IO that is induced either by iron supplementation or due to chronic hemolysis as in β-thalassemia. Prx2 is required to support STAT3 transcriptional activity and regulation of Hamp expression. Antioxid. Redox Signal. 28, 1-14.
Collapse
Affiliation(s)
- Alessandro Matte
- 1 Department of Medicine, University of Verona-AOUI Verona , Verona, Italy
| | - Luigia De Falco
- 2 CEINGE and Department of Biochemistry, Federico II University , Naples, Italy
| | - Enrica Federti
- 1 Department of Medicine, University of Verona-AOUI Verona , Verona, Italy
| | - Anna Cozzi
- 3 Division of Neuroscience, San Raffaele Scientific Institute , Milano, Italy
| | - Achille Iolascon
- 2 CEINGE and Department of Biochemistry, Federico II University , Naples, Italy
| | - Sonia Levi
- 3 Division of Neuroscience, San Raffaele Scientific Institute , Milano, Italy .,4 Vita-Salute San Raffaele University , Milano, Italy
| | | | - Alberto Zamo
- 6 Department of Pathology and Diagnostic University of Verona-AOUI Verona , Verona, Italy
| | - Mariasole Bruno
- 2 CEINGE and Department of Biochemistry, Federico II University , Naples, Italy
| | | | - Anne Janin
- 7 Inserm, U1165, Paris, France .,8 Université Paris 7-Denis Diderot , Paris, France .,9 AP-HP , Hôpital Saint-Louis, Paris, France
| | - Angela Siciliano
- 1 Department of Medicine, University of Verona-AOUI Verona , Verona, Italy
| | - Tom Ganz
- 10 Department of Pathology and Laboratory of Medicine, UCLA School of Medicine , Los Angeles, California
| | - Giorgia Federico
- 11 Department of Molecular Medicine and Medical Biotechnologies Federico II University , Naples, Italy
| | - Francesca Carlomagno
- 11 Department of Molecular Medicine and Medical Biotechnologies Federico II University , Naples, Italy
| | - Sebastian Mueller
- 12 Medical Department, Salem Medical Center, University of Heidelberg , Heidelberg, Germany
| | - Ines Silva
- 12 Medical Department, Salem Medical Center, University of Heidelberg , Heidelberg, Germany
| | - Carmine Carbone
- 1 Department of Medicine, University of Verona-AOUI Verona , Verona, Italy
| | - Davide Melisi
- 1 Department of Medicine, University of Verona-AOUI Verona , Verona, Italy
| | - Dae Won Kim
- 13 Department of Biomedical Sciences, Institute of Bioscience and Biotechnology, Hallym University , Chunchon, Korea
| | - Soo Young Choi
- 13 Department of Biomedical Sciences, Institute of Bioscience and Biotechnology, Hallym University , Chunchon, Korea
| | | |
Collapse
|
34
|
Why should neuroscientists worry about iron? The emerging role of ferroptosis in the pathophysiology of neuroprogressive diseases. Behav Brain Res 2017; 341:154-175. [PMID: 29289598 DOI: 10.1016/j.bbr.2017.12.036] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 12/23/2017] [Accepted: 12/27/2017] [Indexed: 12/12/2022]
Abstract
Ferroptosis is a unique form of programmed death, characterised by cytosolic accumulation of iron, lipid hydroperoxides and their metabolites, and effected by the fatal peroxidation of polyunsaturated fatty acids in the plasma membrane. It is a major driver of cell death in neurodegenerative neurological diseases. Moreover, cascades underpinning ferroptosis could be active drivers of neuropathology in major psychiatric disorders. Oxidative and nitrosative stress can adversely affect mechanisms and proteins governing cellular iron homeostasis, such as the iron regulatory protein/iron response element system, and can ultimately be a source of abnormally high levels of iron and a source of lethal levels of lipid membrane peroxidation. Furthermore, neuroinflammation leads to the upregulation of divalent metal transporter1 on the surface of astrocytes, microglia and neurones, making them highly sensitive to iron overload in the presence of high levels of non-transferrin-bound iron, thereby affording such levels a dominant role in respect of the induction of iron-mediated neuropathology. Mechanisms governing systemic and cellular iron homeostasis, and the related roles of ferritin and mitochondria are detailed, as are mechanisms explaining the negative regulation of ferroptosis by glutathione, glutathione peroxidase 4, the cysteine/glutamate antiporter system, heat shock protein 27 and nuclear factor erythroid 2-related factor 2. The potential role of DJ-1 inactivation in the precipitation of ferroptosis and the assessment of lipid peroxidation are described. Finally, a rational approach to therapy is considered, with a discussion on the roles of coenzyme Q10, iron chelation therapy, in the form of deferiprone, deferoxamine (desferrioxamine) and deferasirox, and N-acetylcysteine.
Collapse
|
35
|
Shinohara N, Zhang G, Oshima Y, Kobayashi T, Imatanaka N, Nakai M, Sasaki T, Kawaguchi K, Gamo M. Kinetics and dissolution of intratracheally administered nickel oxide nanomaterials in rats. Part Fibre Toxicol 2017; 14:48. [PMID: 29183341 PMCID: PMC5706298 DOI: 10.1186/s12989-017-0229-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 11/10/2017] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND The toxicokinetics of nanomaterials are an important factor in toxicity, which may be affected by slow clearance and/or distribution in the body. METHODS Four types of nickel oxide (NiO) nanoparticles were single-administered intratracheally to male F344 rats at three doses of 0.67-6.0 mg/kg body weight. The rats were sacrificed under anesthesia and the lung, thoracic lymph nodes, bronchoalveolar lavage fluid, liver, and other organs were sampled for Ni burden measurement 3, 28, and 91 days post-administration; Ni excretion was measured 6 and 24 h after administration. Solubility of NiO nanoparticles was determined using artificial lysosomal fluid, artificial interstitial fluid, hydrogen peroxide solution, pure water, and saline. In addition, macrophage migration to trachea and phagosome-lysosome-fusion rate constants were estimated using pulmonary clearance and dissolution rate constants. RESULTS The wire-like NiO nanoparticles were 100% dissolved by 24 h when mixed with artificial lysosomal fluid (dissolution rate coefficient: 0.18/h); spherical NiO nanoparticles were 12% and 35% dissolved after 216 h when mixed with artificial lysosomal fluid (1.4 × 10-3 and 4.9 × 10-3/h). The largest irregular-shaped NiO nanoparticles hardly dissolved in any solution, including artificial lysosomal fluid (7.8 × 10-5/h). Pulmonary clearance rate constants, estimated using a one-compartment model, were much higher for the NiO nanoparticles with a wire-shape (0.069-0.078/day) than for the spherical and irregular-shaped NiO nanoparticles (0-0.012/day). Pulmonary clearance rate constants of the largest irregular-shaped NiO nanoparticles showed an inverse correlation with dose. Translocation of NiO from the lungs to the thoracic lymph nodes increased in a time- and dose-dependent manner for three spherical and irregular-shaped NiO nanoparticles, but not for the wire-like NiO nanoparticles. Thirty-five percent of the wire-like NiO nanoparticles were excreted in the first 24 h after administration; excretion was 0.33-3.6% in that time frame for the spherical and irregular-shaped NiO nanoparticles. CONCLUSION These findings suggest that nanomaterial solubility differences can result in variations in their pulmonary clearance. Nanoparticles with moderate lysosomal solubility may induce persistent pulmonary inflammation.
Collapse
Affiliation(s)
- Naohide Shinohara
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, 305-8569, Japan.
| | - Guihua Zhang
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, 305-8569, Japan
| | - Yutaka Oshima
- Chemicals Evaluation and Research Institute (CERI), Hita, Oita, 877-0061, Japan
| | - Toshio Kobayashi
- Chemicals Evaluation and Research Institute (CERI), Hita, Oita, 877-0061, Japan
| | - Nobuya Imatanaka
- Chemicals Evaluation and Research Institute (CERI), Bunkyo, Tokyo, 112-0004, Japan
| | - Makoto Nakai
- Chemicals Evaluation and Research Institute (CERI), Bunkyo, Tokyo, 112-0004, Japan
| | - Takeshi Sasaki
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, 305-8565, Japan
| | - Kenji Kawaguchi
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, 305-8565, Japan
| | - Masashi Gamo
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, 305-8569, Japan
| |
Collapse
|
36
|
Martinez Guimera A, Welsh CM, Proctor CJ, McArdle A, Shanley DP. 'Molecular habituation' as a potential mechanism of gradual homeostatic loss with age. Mech Ageing Dev 2017; 169:53-62. [PMID: 29146308 PMCID: PMC5846846 DOI: 10.1016/j.mad.2017.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/26/2017] [Accepted: 11/10/2017] [Indexed: 12/17/2022]
Abstract
Constitutive signals indicate homeostatic dysregulation but their effect on signal transduction remains largely unexplored. A theoretical approach is undertaken to examine how oxidative stress may affect redox signal transduction. Constitutive signals can result in a ‘molecular habituation’ effect that interferes with information transmission. The robustness of such a theoretical observation to the underlying methodology hints at the generality of this principle.
The ability of reactive oxygen species (ROS) to cause molecular damage has meant that chronic oxidative stress has been mostly studied from the point of view of being a source of toxicity to the cell. However, the known duality of ROS molecules as both damaging agents and cellular redox signals implies another perspective in the study of sustained oxidative stress. This is a perspective of studying oxidative stress as a constitutive signal within the cell. In this work, we adopt a theoretical perspective as an exploratory and explanatory approach to examine how chronic oxidative stress can interfere with signal processing by redox signalling pathways in the cell. We report that constitutive signals can give rise to a ‘molecular habituation’ effect that can prime for a gradual loss of biological function. This is because a constitutive signal in the environment has the potential to reduce the responsiveness of a signalling pathway through the prolonged activation of negative regulators. Additionally, we demonstrate how this phenomenon is likely to occur in different signalling pathways exposed to persistent signals and furthermore at different levels of biological organisation.
Collapse
Affiliation(s)
- Alvaro Martinez Guimera
- Institute for Cell and Molecular Biosciences (ICaMB), Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, Newcastle Upon Tyne, NE4 5PL,United Kingdom; MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom
| | - Ciaran M Welsh
- Institute for Cell and Molecular Biosciences (ICaMB), Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, Newcastle Upon Tyne, NE4 5PL,United Kingdom
| | - Carole J Proctor
- Institute of Cellular Medicine, Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, Newcastle Upon Tyne, NE4 5PL, United Kingdom; MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom
| | - Anne McArdle
- Department of Musculoskeletal Biology, University of Liverpool (University, Not-for-profit), Institute of Ageing and Chronic Disease,William Duncan Building, 6 West Derby Street, Liverpool L7 8TX, United Kingdom; MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom
| | - Daryl P Shanley
- Institute for Cell and Molecular Biosciences (ICaMB), Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, Newcastle Upon Tyne, NE4 5PL,United Kingdom; MRC/Arthritis Research UK Centre for Musculoskeletal Ageing (CIMA), United Kingdom.
| |
Collapse
|
37
|
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
|
38
|
O'Sullivan SA, Velasco-Estevez M, Dev KK. Demyelination induced by oxidative stress is regulated by sphingosine 1-phosphate receptors. Glia 2017; 65:1119-1136. [PMID: 28375547 DOI: 10.1002/glia.23148] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 03/02/2017] [Accepted: 03/20/2017] [Indexed: 01/14/2023]
Abstract
Oxidative stress is a pathological condition defined as an imbalance between production and removal of reactive oxygen species. This process causes structural cell damage, disrupts DNA repair and induces mitochondrial dysfunction. Many in vitro studies have used direct bolus application of H2 O2 to investigate the role of oxidative stress in cell culture. In this study, using mouse organotypic cerebellar slice cultures, the effects of H2 O2 -induced oxidative stress on myelination state were examined, using bolus concentrations of H2 O2 (0.1-1 mM) and low-continuous H2 O2 (∼20 μM) generated from glucose oxidase and catalase (GOX-CAT). Using these models, the potential therapeutic effects of pFTY720, an oral therapy used in multiple sclerosis, was also examined. We found bolus treatment of H2 O2 (0.5 mM) and, for the first time, low-continuous H2 O2 (GOX-CAT) to induce demyelination in organotypic slices. Both bolus H2 O2 and GOX-CAT treatments significantly decreased vimentin expression in these slice cultures as well as increased cell death in isolated astrocyte cultures. Importantly, pre-treatment with pFTY720 significantly attenuated both bolus H2 O2 and GOX-CAT-induced demyelination and the GOX-CAT-induced decrease in vimentin in cerebellar slices, without altering levels of the proinflammatory cytokines such as IL-6 and CX3CL1. We also observed increased SMI-32 immunoreactivity in the white matter tract induced by GOX-CAT indicating axonal damage, which was remarkably attenuated by pFTY720. Taken together, this data establishes a novel GOX-CAT model of demyelination and demonstrates that pFTY720 can act independently of inflammatory cytokines to attenuate decreases in vimentin, as well as axonal damage and demyelination induced by oxidative stress.
Collapse
Affiliation(s)
- Sinead A O'Sullivan
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | | | - Kumlesh K Dev
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
39
|
Abstract
Impaired iron homeostasis and the suppressive effects of proinflammatory cytokines on erythropoiesis, together with alterations of the erythrocyte membrane that impair its survival, cause anemia of inflammation. Recent epidemiologic studies have connected inflammatory anemia with critical illness, obesity, aging, kidney failure, cancer, chronic infection, and autoimmune disease. The proinflammatory cytokine, interleukin-6, the iron regulatory hormone, hepcidin, and the iron exporter, ferroportin, interact to cause iron sequestration in the setting of inflammation. Although severe anemia is associated with adverse outcomes in critical illness, experimental models suggest that iron sequestration is part of a natural defense against pathogens.
Collapse
Affiliation(s)
- Paula G Fraenkel
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
40
|
Singh SK, Thirumalai A, Pathak A, Ngwa DN, Agrawal A. Functional Transformation of C-reactive Protein by Hydrogen Peroxide. J Biol Chem 2017; 292:3129-3136. [PMID: 28096464 PMCID: PMC5336149 DOI: 10.1074/jbc.m116.773176] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 01/11/2017] [Indexed: 12/21/2022] Open
Abstract
C-reactive protein (CRP) is present at sites of inflammation including amyloid plaques, atherosclerotic lesions, and arthritic joints. CRP, in its native pentameric structural conformation, binds to cells and molecules that have exposed phosphocholine (PCh) groups. CRP, in its non-native pentameric structural conformation, binds to a variety of deposited, denatured, and aggregated proteins, in addition to binding to PCh-containing substances. In this study, we investigated the effects of H2O2, a prototypical reactive oxygen species that is also present at sites of inflammation, on the ligand recognition function of CRP. Controlled H2O2 treatment of native CRP did not monomerize CRP and did not affect the PCh binding activity of CRP. In solid phase ELISA-based ligand binding assays, purified pentameric H2O2-treated CRP bound to a number of immobilized proteins including oxidized LDL, IgG, amyloid β peptide 1-42, C4b-binding protein, and factor H, in a CRP concentration- and ligand concentration-dependent manner. Using oxidized LDL as a representative protein ligand for H2O2-treated CRP, we found that the binding occurred in a Ca2+-independent manner and did not involve the PCh-binding site of CRP. We conclude that H2O2 is a biological modifier of the structure and ligand recognition function of CRP. Overall, the data suggest that the ligand recognition function of CRP is dependent on the presence of an inflammatory microenvironment. We hypothesize that one of the functions of CRP at sites of inflammation is to sense the inflammatory microenvironment, change its own structure in response but remain pentameric, and then bind to pathogenic proteins deposited at those sites.
Collapse
Affiliation(s)
- Sanjay K Singh
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614
| | - Avinash Thirumalai
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614
| | - Asmita Pathak
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614
| | - Donald N Ngwa
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614
| | - Alok Agrawal
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614.
| |
Collapse
|
41
|
Wang H, Shi P, Huang C, Liu Q. Maresin 1 ameliorates iron-deficient anemia in IL-10(-/-) mice with spontaneous colitis by the inhibition of hepcidin expression though the IL-6/STAT3 pathway. Am J Transl Res 2016; 8:2758-2766. [PMID: 27398158 PMCID: PMC4931169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/08/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Approximately 50% of patients with inflammatory bowel disease (IBD) suffer from anemia, which is prevalently caused by iron deficiency. Maresin 1 (MaR1) is a novel docosahexaenoic acid-derived pro-resolving agent that promotes the resolution of inflammation. The aim of the present study was to investigate the therapeutic effects of MaR1 on iron-deficient anemia in IL-10 knockout (IL-10(-/-)) mice with spontaneous chronic colitis. METHODS IL-10(-/-) mice of 16 weeks of age with established colitis were used for the experiments with MaR1 treatment for 2 weeks. Histologic injury, CD4+ lymphocyte values in the lamina propria, blood hemoglobin, hematocrit, serum iron concentrations, transferrin saturation, splenic iron stores, levels of inflammatory cytokines, expression of liver hepcidin mRNA, and western blotting of STAT3 were analyzed in this study. RESULTS MaR1 treatment (0.3 ng/mouse) effectively attenuated histological colitis typically associated with decreased CD4+ lymphocytes in the lamina propria as well as the concentrations of MPO, TNF-α, IFN-γ, IL-6 and IL-17 (P<0.05). Furthermore, reduced expression of liver hepcidin mRNA and p-STAT3 expression, as well as increased hemoglobin concentration, hematocrit, levels of serum iron, transferrin saturation and splenic iron stores were found in IL-10(-/-) mice after MaR1 treatment (P<0.05). CONCLUSIONS These results indicate that MaR1 treatment ameliorates iron-deficient anemia by reducing colonic inflammation and inhibiting hepcidin expression though the IL-6/STAT3 pathway.
Collapse
Affiliation(s)
- Honggang Wang
- Department of General Surgery, Taizhou People’s Hospital Affiliated to Nantong UniversityNo. 210 Yingchun Road, Taizhou, Jiangsu Province 225300, China
| | - Peiliang Shi
- Model Animal Research Center of Nanjing UniversityNo. 12 Xuefu Road, Nanjing, Jiangsu Province, China
| | - Chuanjiang Huang
- Department of General Surgery, Taizhou People’s Hospital Affiliated to Nantong UniversityNo. 210 Yingchun Road, Taizhou, Jiangsu Province 225300, China
| | - Qinghong Liu
- Department of General Surgery, Taizhou People’s Hospital Affiliated to Nantong UniversityNo. 210 Yingchun Road, Taizhou, Jiangsu Province 225300, China
| |
Collapse
|
42
|
Hepcidin Plays a Key Role in 6-OHDA Induced Iron Overload and Apoptotic Cell Death in a Cell Culture Model of Parkinson's Disease. PARKINSONS DISEASE 2016; 2016:8684130. [PMID: 27298749 PMCID: PMC4889865 DOI: 10.1155/2016/8684130] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/05/2016] [Indexed: 01/11/2023]
Abstract
Background. Elevated brain iron levels have been implicated in the pathogenesis of Parkinson's disease (PD). However, the precise mechanism underlying abnormal iron accumulation in PD is not clear. Hepcidin, a hormone primarily produced by hepatocytes, acts as a key regulator in both systemic and cellular iron homeostasis. Objective. We investigated the role of hepcidin in 6-hydroxydopamine (6-OHDA) induced apoptosis in a cell culture model of PD. Methods. We downregulated hepcidin using siRNA interference in N27 dopaminergic neuronal cells and made a comparison with control siRNA transfected cells to investigate the role of hepcidin in 6-OHDA induced neurodegeneration. Results. Hepcidin knockdown (32.3%, P < 0.0001) upregulated ferroportin 1 expression and significantly (P < 0.05) decreased intracellular iron by 25%. Hepcidin knockdown also reduced 6-OHDA induced caspase-3 activity by 42% (P < 0.05) and DNA fragmentation by 29% (P = 0.086) and increased cell viability by 22% (P < 0.05). In addition, hepcidin knockdown significantly attenuated 6-OHDA induced protein carbonyls by 52% (P < 0.05) and intracellular iron by 28% (P < 0.01), indicating the role of hepcidin in oxidative stress. Conclusions. Our results demonstrate that hepcidin knockdown protected N27 cells from 6-OHDA induced apoptosis and that hepcidin plays a major role in reducing cellular iron burden and oxidative damage by possibly regulating cellular iron export mediated by ferroportin 1.
Collapse
|
43
|
Abstract
Abstract
The anemia of chronic disease is an old disease concept, but contemporary research in the role of proinflammatory cytokines and iron biology has shed new light on the pathophysiology of the condition. Recent epidemiologic studies have connected the anemia of chronic disease with critical illness, obesity, aging, and kidney failure, as well as with the well-established associations of cancer, chronic infection, and autoimmune disease. Functional iron deficiency, mediated principally by the interaction of interleukin-6, the iron regulatory hormone hepcidin, and the iron exporter ferroportin, is a major contributor to the anemia of chronic disease. Although anemia is associated with adverse outcomes, experimental models suggest that iron sequestration is desirable in the setting of severe infection. Experimental therapeutic approaches targeting interleukin-6 or the ferroportin–hepcidin axis have shown efficacy in reversing anemia in either animal models or human patients, although these agents have not yet been approved for the treatment of the anemia of chronic disease.
Collapse
|
44
|
|
45
|
Schmidt PJ. Regulation of Iron Metabolism by Hepcidin under Conditions of Inflammation. J Biol Chem 2015; 290:18975-83. [PMID: 26055723 DOI: 10.1074/jbc.r115.650150] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Iron is a redox-active metal required as a cofactor in multiple metalloproteins essential for a host of life processes. The metal is highly toxic when present in excess and must be strictly regulated to prevent tissue and organ damage. Hepcidin, a molecule first characterized as an antimicrobial peptide, plays a critical role in the regulation of iron homeostasis. Multiple stimuli positively influence the expression of hepcidin, including iron, inflammation, and infection by pathogens. In this Minireview, I will discuss how inflammation regulates hepcidin transcription, allowing for sufficient concentrations of iron for organismal needs while sequestering the metal from infectious pathogens.
Collapse
Affiliation(s)
- Paul J Schmidt
- From the Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
46
|
Harrison-Findik DD, Lu S. The effect of alcohol and hydrogen peroxide on liver hepcidin gene expression in mice lacking antioxidant enzymes, glutathione peroxidase-1 or catalase. Biomolecules 2015; 5:793-807. [PMID: 25955433 PMCID: PMC4496697 DOI: 10.3390/biom5020793] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/22/2015] [Accepted: 04/27/2015] [Indexed: 01/04/2023] Open
Abstract
This study investigates the regulation of hepcidin, the key iron-regulatory molecule, by alcohol and hydrogen peroxide (H2O2) in glutathione peroxidase-1 (gpx-1(-/-)) and catalase (catalase(-/-)) knockout mice. For alcohol studies, 10% ethanol was administered in the drinking water for 7 days. Gpx-1(-/-) displayed significantly higher hepatic H2O2 levels than catalase(-/-) compared to wild-type mice, as measured by 2'-7'-dichlorodihydrofluorescein diacetate (DCFH-DA). The basal level of liver hepcidin expression was attenuated in gpx-1(-/-) mice. Alcohol increased H2O2 production in catalase(-/-) and wild-type, but not gpx-1(-/-), mice. Hepcidin expression was inhibited in alcohol-fed catalase(-/-) and wild-type mice. In contrast, alcohol elevated hepcidin expression in gpx-1(-/-) mice. Gpx-1(-/-) mice also displayed higher level of basal liver CHOP protein expression than catalase(-/-) mice. Alcohol induced CHOP and to a lesser extent GRP78/BiP expression, but not XBP1 splicing or binding of CREBH to hepcidin gene promoter, in gpx-1(-/-) mice. The up-regulation of hepatic ATF4 mRNA levels, which was observed in gpx-1(-/-) mice, was attenuated by alcohol. In conclusion, our findings strongly suggest that H2O2 inhibits hepcidin expression in vivo. Synergistic induction of CHOP by alcohol and H2O2, in the absence of gpx-1, stimulates liver hepcidin gene expression by ER stress independent of CREBH.
Collapse
Affiliation(s)
- Duygu Dee Harrison-Findik
- Department of Internal Medicine, Division of Gastroenterology/Hepatology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Sizhao Lu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
47
|
The role of iron in alcohol-mediated hepatocarcinogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 815:89-112. [PMID: 25427903 DOI: 10.1007/978-3-319-09614-8_6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alcoholic liver disease (ALD) is the major liver disease in the developed world and characterized by hepatic iron overload in ca. 50% of all patients. This iron overload is an independent factor of disease progression, hepatocellular carcinoma and it determines survival. Since simple phlebotomy does not allow the efficient removal of excess iron in ALD, a better understanding of the underlying mechanisms is urgently needed to identify novel targeted treatment strategies. This review summarizes the present knowledge on iron overload in patients with ALD. Although multiple sides of the cellular and systemic iron homeostasis may be affected during alcohol consumption, most studies have focused on potential hepatic causes. However, it should not be overlooked that more than 90% of the major iron pool, the hemoglobin-associated iron, is efficiently recycled within the human body and it is also strongly affected by alcohol. The few available studies suggest various molecular mechanisms that involve iron regulatory protein (IRP1), transferrin receptor 1 (TfR1), and the systemic iron master switch hepcidin, but not classical mutations of the HFE gene. Notably, reactive oxygen species (ROS), namely, hydrogen peroxide (H2O2), are powerful modulators of these iron-steering proteins. For instance, depending on the level, H2O2 may both strongly suppress and induce the expression of hepcidin that could partly explain the anemia and iron overload observed in these patients. More studies with appropriate ROS models such as the novel GOX/CAT system are required to unravel the mechanisms of iron overload in ALD to consequently identify molecular-targeted therapies in the future.
Collapse
|
48
|
The iron-regulatory hormone hepcidin: A possible therapeutic target? Pharmacol Ther 2015; 146:35-52. [DOI: 10.1016/j.pharmthera.2014.09.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 09/02/2014] [Indexed: 01/19/2023]
|
49
|
Abstract
Iron and oxygen metabolism are intimately linked with one another.
Collapse
Affiliation(s)
- Robert J. Simpson
- Diabetes and Nutritional Sciences
- School of Medicine
- Kings College London
- , UK
| | - Andrew T. McKie
- Diabetes and Nutritional Sciences
- School of Medicine
- Kings College London
- , UK
| |
Collapse
|
50
|
Macciò A, Madeddu C, Gramignano G, Mulas C, Tanca L, Cherchi MC, Floris C, Omoto I, Barracca A, Ganz T. The role of inflammation, iron, and nutritional status in cancer-related anemia: results of a large, prospective, observational study. Haematologica 2015; 100:124-132. [PMID: 25239265 PMCID: PMC4281325 DOI: 10.3324/haematol.2014.112813] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 09/16/2014] [Indexed: 12/21/2022] Open
Abstract
Anemia in oncology patients is often considered a side effect of cancer therapy; however, it may occur before any antineoplastic treatment (cancer-related anemia). This study was aimed to evaluate the prevalence of cancer-related anemia in a large cohort of oncology patients and whether inflammation and malnutrition were predictive of its development and severity. The present study included 888 patients with cancer at different sites between May 2011 and January 2014. Patients were assessed at diagnosis before any cancer treatment. The prevalence of anemia according to the main clinical factors (tumor site, stage and performance status) was analyzed. In each patient markers of inflammation, iron metabolism, malnutrition and oxidative stress as well as the modified Glasgow prognostic score, a combined index of malnutrition and inflammation, were assessed and their role in predicting hemoglobin level was evaluated. The percentage of anemic patients was 63% with the lowest hemoglobin levels being found in the patients with most advanced cancer and compromised performance status. Hemoglobin concentration differed by tumor site and was lowest in patients with ovarian cancer. Hemoglobin concentration was inversely correlated with inflammatory markers, hepcidin, ferritin, erythropoietin and reactive oxygen species, and positively correlated with leptin, albumin, cholesterol and antioxidant enzymes. In multivariate analysis, stage, interleukin-6 and leptin were independent predictors of hemoglobin concentration. Furthermore, hemoglobin was inversely dependent on modified Glasgow Prognostic Score. In conclusion, cancer-related anemia is a multifactorial problem with immune, nutritional and metabolic components that affect its severity. Only a detailed assessment of the pathogenesis of cancer-related anemia may enable clinicians to provide safe and effective individualized treatment.
Collapse
Affiliation(s)
- Antonio Macciò
- Department of Gynecological Oncology, A. Businco Hospital, Regional Referral Center for Cancer Disease, Cagliari, Italy
| | - Clelia Madeddu
- Department of Medical Science "Mario Aresu", University of Cagliari, Italy
| | | | - Carlo Mulas
- Medical Oncology Unit, Sirai Hospital, Carbonia, Italy
| | - Luciana Tanca
- Department of Medical Oncology, A. Businco Hospital, Regional Referral Center for Cancer Disease, Cagliari, Italy
| | - Maria Cristina Cherchi
- Department of Medical Oncology, A. Businco Hospital, Regional Referral Center for Cancer Disease, Cagliari, Italy
| | - Carlo Floris
- Medical Oncology Unit, "Nuova Casa di Cura", Decimomannu, Cagliari, Italy
| | - Itaru Omoto
- Department of Digestive Surgery, Breast and Thyroid Surgery, Kagoshima University School of Medicine, Kagoshima, Japan
| | - Antonio Barracca
- Department of Gynecological Oncology, A. Businco Hospital, Regional Referral Center for Cancer Disease, Cagliari, Italy
| | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|