51
|
Schiano E, Cappello E, Cecere D, Pompeo F, Novellino E, Stornaiuolo M, Izzo M. Increased Levels of Circulating Iron-Albumin Complexes in Peripheral Arterial Disease Patients. Antioxidants (Basel) 2023; 12:antiox12020503. [PMID: 36830061 PMCID: PMC9952351 DOI: 10.3390/antiox12020503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Under physiological conditions, extracellular iron circulates in the blood bound to transferrin. As a consequence of several pathologies, the circulating level of a Non-Transferrin Bound pool of Iron (NTBI) increases. The NTBI pool is biologically heterogeneous and represented by iron chelated either by small metabolites (citrate, amino acids, or cofactors) or by serum proteins. By promoting reactive oxygen species (ROS) and reactive nitrogen species (RNS) formation, NTBI causes oxidative stress and alteration of membrane lipids, seriously compromising the healthy state of organs and tissues. While NTBI involvement in several pathologies has been clarified, its contribution to vascular diseases remains to be investigated. Here we measure and analyze the pool of NTBI in the serum of a small group of peripheral arterial disease (PAD) patients. We show that: (i) the NTBI pool shifts from low molecular complexes to high-molecular ones in PAD patients compared to healthy controls; (ii) most of this NTBI is bound to the serum protein Albumin; (iii) this NTBI-Albumin complex can be isolated and quantitated following a simple immunoisolation procedure amenable to automation and suitable for clinical screening purposes.
Collapse
Affiliation(s)
- Elisabetta Schiano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | | | | | | | - Ettore Novellino
- Department of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Mariano Stornaiuolo
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
- Correspondence: (M.S.); (M.I.)
| | - Marcello Izzo
- MathTechMed-Department of Mathematics for Technology, Medicine and Biosciences Research Center, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: (M.S.); (M.I.)
| |
Collapse
|
52
|
Claus MA, Smart L, Raisis AL, Sharp CR, Abraham S, Gummer JPA, Mead MK, Bradley DL, Van Swelm R, Wiegerinck ETG, Litton E. Effect of Deferoxamine on Post-Transfusion Iron, Inflammation, and In Vitro Microbial Growth in a Canine Hemorrhagic Shock Model: A Randomized Controlled Blinded Pilot Study. Vet Sci 2023; 10:vetsci10020121. [PMID: 36851425 PMCID: PMC9962370 DOI: 10.3390/vetsci10020121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Red blood cell (RBC) transfusion is associated with recipient inflammation and infection, which may be triggered by excessive circulating iron. Iron chelation following transfusion may reduce these risks. The aim of this study was to evaluate the effect of deferoxamine on circulating iron and inflammation biomarkers over time and in vitro growth of Escherichia coli (E. coli) following RBC transfusion in dogs with atraumatic hemorrhage. Anesthetized dogs were subject to atraumatic hemorrhage and transfusion of RBCs, then randomized to receive either deferoxamine or saline placebo of equivalent volume (n = 10 per group) in a blinded fashion. Blood was sampled before hemorrhage and then 2, 4, and 6 h later. Following hemorrhage and RBC transfusion, free iron increased in all dogs over time (both p < 0.001). Inflammation biomarkers interleukin-6 (IL6), CXC motif chemokine-8 (CXCL8), interleukin-10 (IL10), and keratinocyte-derived chemokine (KC) increased in all dogs over time (all p < 0.001). Logarithmic growth of E. coli clones within blood collected 6 h post-transfusion was not different between groups. Only total iron-binding capacity was different between groups over time, being significantly increased in the deferoxamine group at 2 and 4 h post-transfusion (both p < 0.001). In summary, while free iron and inflammation biomarkers increased post-RBC transfusion, deferoxamine administration did not impact circulating free iron, inflammation biomarkers, or in vitro growth of E. coli when compared with placebo.
Collapse
Affiliation(s)
- Melissa A. Claus
- School of Veterinary Medicine, Murdoch University, Murdoch, WA 6150, Australia
- Perth Veterinary Specialists, Osborne Park, WA 6017, Australia
- Correspondence:
| | - Lisa Smart
- School of Veterinary Medicine, Murdoch University, Murdoch, WA 6150, Australia
- Small Animal Specialist Hospital, Tuggerah, NSW 2259, Australia
| | - Anthea L. Raisis
- School of Veterinary Medicine, Murdoch University, Murdoch, WA 6150, Australia
| | - Claire R. Sharp
- School of Veterinary Medicine, Murdoch University, Murdoch, WA 6150, Australia
| | - Sam Abraham
- School of Veterinary Medicine, Murdoch University, Murdoch, WA 6150, Australia
| | - Joel P. A. Gummer
- Forensic Sciences Laboratory, ChemCentre, Resources and Chemistry Precinct, Bentley, WA 6102, Australia
- School of Science, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Martin K. Mead
- School of Veterinary Medicine, Murdoch University, Murdoch, WA 6150, Australia
| | - Damian L. Bradley
- Intensive Care Unit, Rockingham General Hospital, Cooloongup, WA 6168, Australia
| | - Rachel Van Swelm
- Hepcidinanalysis.com, Department of Laboratory Medicine, Translational Metabolic Laboratory (TML 830), Radboud University Medical Center, 6525 Nijmegen, The Netherlands
| | - Erwin T. G. Wiegerinck
- Hepcidinanalysis.com, Department of Laboratory Medicine, Translational Metabolic Laboratory (TML 830), Radboud University Medical Center, 6525 Nijmegen, The Netherlands
| | - Edward Litton
- Intensive Care Unit, Fiona Stanley Hospital, Murdoch, WA 6150, Australia
- School of Medicine, University of Western Australia, Crawley, WA 6009, Australia
| |
Collapse
|
53
|
Abstract
The cardiovascular system requires iron to maintain its high energy demands and metabolic activity. Iron plays a critical role in oxygen transport and storage, mitochondrial function, and enzyme activity. However, excess iron is also cardiotoxic due to its ability to catalyze the formation of reactive oxygen species and promote oxidative damage. While mammalian cells have several redundant iron import mechanisms, they are equipped with a single iron-exporting protein, which makes the cardiovascular system particularly sensitive to iron overload. As a result, iron levels are tightly regulated at many levels to maintain homeostasis. Iron dysregulation ranges from iron deficiency to iron overload and is seen in many types of cardiovascular disease, including heart failure, myocardial infarction, anthracycline-induced cardiotoxicity, and Friedreich's ataxia. Recently, the use of intravenous iron therapy has been advocated in patients with heart failure and certain criteria for iron deficiency. Here, we provide an overview of systemic and cellular iron homeostasis in the context of cardiovascular physiology, iron deficiency, and iron overload in cardiovascular disease, current therapeutic strategies, and future perspectives.
Collapse
Affiliation(s)
- Konrad Teodor Sawicki
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL 60611
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Adam De Jesus
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL 60611
| | - Hossein Ardehali
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL 60611
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| |
Collapse
|
54
|
Yu X, Zhang Q, Ding H, Wang P, Feng J. Plasma Non-transferrin-Bound Iron Could Enter into Mice Duodenum and Negatively Affect Duodenal Defense Response to Virus and Immune Responses. Biol Trace Elem Res 2023; 201:786-799. [PMID: 35294743 DOI: 10.1007/s12011-022-03200-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 03/10/2022] [Indexed: 01/21/2023]
Abstract
Plasma non-transferrin-bound iron (NTBI) exists when the plasma iron content exceeds the carrying capacity of transferrin and can be quickly cleared by the liver, pancreas, and other organs. However, whether it could enter the small intestine and its effects still remain unclear. Herein, these issues were explored. Mice were intravenously administrated of ferric citrate (treatment) or citrate acid (control) 10 min after the saturation of the transferrin. Two hours later, hepatic, duodenal, and jejunal iron content and distribution were measured and duodenal transcriptome sequencing was performed. Significant increase of duodenal and hepatic iron content was detected, indicating that plasma NTBI could be absorbed by the duodenum as well as the liver. A total of 103 differentially expressed genes were identified in the duodenum of mice in the treatment group compared to the control group. Gene Ontology (GO) functional analysis of these genes showed that they were mainly involved in defense response to virus and immune response. The results of Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) analysis revealed that there were major changes in the hematopoietic cell lineage and some virus infection pathways between the two groups. Determination of 7 cytokines in the duodenum were further conducted, which demonstrated that the anti-inflammatory factors interferon (IL)-4 and IL-10 in the duodenum were significantly decreased after NTBI uptake. Our findings revealed that NTBI in plasma can enter the duodenum, which would change the duodenal hematopoietic cell lineage and have a negative impact on defense response to the virus and immune responses.
Collapse
Affiliation(s)
- Xiaonan Yu
- Key Laboratory of Animal Nutrition & Feed Science, Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Qian Zhang
- Key Laboratory of Animal Nutrition & Feed Science, Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Haoxuan Ding
- Key Laboratory of Animal Nutrition & Feed Science, Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Peng Wang
- Key Laboratory of Animal Nutrition & Feed Science, Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jie Feng
- Key Laboratory of Animal Nutrition & Feed Science, Zhejiang Province, College of Animal Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
55
|
Garcia MR, Comstock BA, Patel RM, Tolia VN, Josephson CD, Georgieff MK, Rao R, Monsell SE, Juul SE, Ahmad KA. Iron supplementation and the risk of bronchopulmonary dysplasia in extremely low gestational age newborns. Pediatr Res 2023; 93:701-707. [PMID: 35725917 PMCID: PMC9763546 DOI: 10.1038/s41390-022-02160-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/26/2022] [Accepted: 05/22/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND The aim of this study was to determine the relationship between iron exposure and the development of bronchopulmonary dysplasia (BPD). METHODS A secondary analysis of the PENUT Trial dataset was conducted. The primary outcome was BPD at 36 weeks gestational age and primary exposures of interest were cumulative iron exposures in the first 28 days and through 36 weeks' gestation. Descriptive statistics were calculated for study cohort characteristics with analysis adjusted for the factors used to stratify randomization. RESULTS Of the 941 patients, 821 (87.2%) survived to BPD evaluation at 36 weeks, with 332 (40.4%) diagnosed with BPD. The median cohort gestational age was 26 weeks and birth weight 810 g. In the first 28 days, 76% of infants received enteral iron and 55% parenteral iron. The median supplemental cumulative enteral and parenteral iron intakes at 28 days were 58.5 and 3.1 mg/kg, respectively, and through 36 weeks' 235.8 and 3.56 mg/kg, respectively. We found lower volume of red blood cell transfusions in the first 28 days after birth and higher enteral iron exposure in the first 28 days after birth to be associated with lower rates of BPD. CONCLUSIONS We find no support for an increased risk of BPD with iron supplementation. TRIAL REGISTRATION NUMBER NCT01378273. https://clinicaltrials.gov/ct2/show/NCT01378273 IMPACT: Prior studies and biologic plausibility raise the possibility that iron administration could contribute to the pathophysiology of oxidant-induced lung injury and thus bronchopulmonary dysplasia in preterm infants. For 24-27-week premature infants, this study finds no association between total cumulative enteral iron supplementation at either 28-day or 36-week postmenstrual age and the risk for developing bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Melissa R Garcia
- San Antonio Uniformed Services Health Education Consortium, San Antonio, TX, USA
| | | | - Ravi M Patel
- Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Veeral N Tolia
- Pediatrix Medical Group, Dallas, TX, USA
- Baylor University Medical Center, Dallas, TX, USA
| | - Cassandra D Josephson
- Emory University School of Medicine and Center for Transfusion and Cellular Therapies, Atlanta, GA, USA
| | | | | | | | | | - Kaashif A Ahmad
- San Antonio Uniformed Services Health Education Consortium, San Antonio, TX, USA.
- Pediatrix Medical Group, San Antonio, TX, USA.
- Baylor College of Medicine, San Antonio, TX, USA.
- Pediatrix and Obstetrix Specialists of Houston, Houston, TX, USA.
- Methodist Children's Hospital, San Antonio, TX, USA.
| |
Collapse
|
56
|
Qin Y, Huang Y, Li Y, Qin L, Wei Q, Chen X, Yang C, Zhang M. Association between systemic iron status and β-cell function and insulin sensitivity in patients with newly diagnosed type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1143919. [PMID: 37077360 PMCID: PMC10107407 DOI: 10.3389/fendo.2023.1143919] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/14/2023] [Indexed: 04/05/2023] Open
Abstract
OBJECTIVE Abnormal iron metabolism is related to the risk of diabetes, but the underlying mechanism of this association remains uncertain. This study was conducted to evaluate the contributions of systemic iron status to β-cell function and insulin sensitivity of patients with newly diagnosed T2DM. METHODS A total of 162 patients with newly diagnosed T2DM and 162 healthy controls were enrolled in the study. Basic characteristics, biochemical indicators, and iron metabolism biomarkers, including serum iron (SI), ferritin (SF), transferrin (Trf), and transferrin saturation (TS), were collected. All patients underwent a 75 g oral glucose tolerance test. A series of parameters for assessing β-cell function and insulin sensitivity were calculated. The multivariate stepwise linear regression model was used to investigate the contributions of iron metabolism to β-cell function and insulin sensitivity. RESULTS Compared with healthy controls, patients with newly diagnosed T2DM had significantly higher levels of SF. Among the diabetic patients, the SI and TS levels were higher, and the percentage of Trf levels below normal values was lower in men than in women. In all diabetic patients, SF was the independent risk factor associated with impaired β-cell function. Further stratification analysis showed that Trf was an independent protective factor for β-cell function in male patients, while SF was an independent risk factor for impaired β-cell function in female patients. However, systemic iron status did not affect insulin sensitivity. CONCLUSION Elevated SF levels and decreased Trf levels had a profound effect on impaired β-cell function in Chinese patients with newly diagnosed T2DM.
Collapse
Affiliation(s)
- Yao Qin
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yiting Huang
- Department of Clinical Nutrition, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuxiao Li
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lu Qin
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qianying Wei
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin Chen
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chuanhui Yang
- Department of Endocrinology, the First People’s Hospital of Lianyungang, Lianyungang, China
| | - Mei Zhang
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Mei Zhang,
| |
Collapse
|
57
|
Luaibi HA, Mohammed BJ. Does TNF-α 308 G/A (rs1800629) gene polymorphism associate with liver and pancreas disorders in Iraqi adults with beta thalassemia major? Hum Antibodies 2023; 31:99-105. [PMID: 38217591 DOI: 10.3233/hab-230015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2024]
Abstract
BACKGROUND TNF-α has been considered as the key regulator of inflammatory responses and is known to be participated in the pathogenesis of several diseases. OBJECTIVE The aim of this study was to explore the relationship of (rs1800629) gene polymorphism associated to liver and pancreas disorders in sample of β-thalassemia major adult Iraqi Patients. MATERIAL AND METHOD Blood samples were obtained from 40 patients suffered from beta thalassemia with pancreas disorder, along with 40 patient suffered from thalassemia with liver disorder, and 40 patient suffered from thalassemia without pancreas or liver, from Ibn Al-Baladi Hospital, Baghdad, and 40 samples from age and gender-matched apparently healthy individuals as control group, all subjects with age more than 18 years. TNF-308G/A (rs1800629) gene polymorphisms were assessed by Tetra- ARMS-PCR. RESULTS The result of showed that heterogeneous GA and homogeneous AA genotypes were higher, while GG wild genotype was lower in beta thalassemia major patients with liver and pancreas disorders compared to control group. CONCLUSION It can be concluded that the prevalence of TNF-α 308 G/A SNP plus (A) allele could be associated with risk of liver and pancreas disorders in sample of beta thalassemia major adult.
Collapse
|
58
|
Zhang J, Song Y, Li Y, Lin HB, Fang X. Iron homeostasis in the heart: Molecular mechanisms and pharmacological implications. J Mol Cell Cardiol 2023; 174:15-24. [PMID: 36375319 DOI: 10.1016/j.yjmcc.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/13/2022]
Abstract
Iron is necessary for the life of practically all living things, yet it may also harm people toxically. Accordingly, humans and other mammals have evolved an effective and tightly regulatory system to maintain iron homeostasis in healthy tissues, including the heart. Iron deficiency is common in patients with heart failure, and is associated with worse prognosis in this population; while the prevalence of iron overload-related cardiovascular disorders is also increasing. Therefore, enhancing the therapy of patients with cardiovascular disorders requires a thorough understanding of iron homeostasis. Here, we give readers an overview of the fundamental mechanisms governing systemic iron homeostasis as well as the most recent knowledge about the intake, storage, use, and export of iron from the heart. Genetic mouse models used for investigation of iron metabolism in various in vivo scenarios are summarized and highlighted. We also go through different clinical conditions and therapeutic approaches that target cardiac iron dyshomeostasis. Finally, we conclude the review by outlining the present knowledge gaps and important open questions in this field in order to guide future research on cardiac iron metabolism.
Collapse
Affiliation(s)
- Jiawei Zhang
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Yijing Song
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - You Li
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Han-Bin Lin
- Zhongshan Institute for Drug Discovery, SIMM, CAS, Zhongshan, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xuexian Fang
- Department of Nutrition and Toxicology, School of Public Health, Hangzhou Normal University, Hangzhou, China; Key Laboratory of Elemene Class Anti-cancer Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China.
| |
Collapse
|
59
|
Duan G, Li J, Duan Y, Zheng C, Guo Q, Li F, Zheng J, Yu J, Zhang P, Wan M, Long C. Mitochondrial Iron Metabolism: The Crucial Actors in Diseases. Molecules 2022; 28:29. [PMID: 36615225 PMCID: PMC9822237 DOI: 10.3390/molecules28010029] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Iron is a trace element necessary for cell growth, development, and cellular homeostasis, but insufficient or excessive level of iron is toxic. Intracellularly, sufficient amounts of iron are required for mitochondria (the center of iron utilization) to maintain their normal physiologic function. Iron deficiency impairs mitochondrial metabolism and respiratory activity, while mitochondrial iron overload promotes ROS production during mitochondrial electron transport, thus promoting potential disease development. This review provides an overview of iron homeostasis, mitochondrial iron metabolism, and how mitochondrial iron imbalances-induced mitochondrial dysfunction contribute to diseases.
Collapse
Affiliation(s)
- Geyan Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianjun Li
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yehui Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Changbing Zheng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Qiuping Guo
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fengna Li
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Zheng
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiayi Yu
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peiwen Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Mengliao Wan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Cimin Long
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
60
|
The Emerging Role of Ferroptosis in Liver Cancers. LIFE (BASEL, SWITZERLAND) 2022; 12:life12122128. [PMID: 36556493 PMCID: PMC9788082 DOI: 10.3390/life12122128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/25/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Liver cancer represents a global health challenge with worldwide growth. Hepatocellular carcinoma (HCC) is the most common type of liver cancer. Indeed, approximately 90% of HCC cases have a low survival rate. Moreover, cholangiocarcinoma (CC) is another malignant solid tumor originating from cholangiocytes, the epithelial cells of the biliary system. It is the second-most common primary liver tumor, with an increasing course in morbidity and mortality. Tumor cells always show high metabolic levels, antioxidant modifications, and an increased iron uptake to maintain unlimited growth. In recent years, alterations in iron metabolism have been shown to play an important role in the pathogenesis of HCC. Several findings show that a diet rich in iron can enhance HCC risk. Hence, elevated iron concentration inside the cell may promote the development of HCC. Growing evidence sustains that activating ferroptosis may potentially block the proliferation of HCC cells. Even in CC, it has been shown that ferroptosis plays a crucial role in the treatment of tumors. Several data confirmed the inhibitory effect in cell growth of photodynamic therapy (PDT) that can induce reactive oxygen species (ROS) in CC, leading to an increase in malondialdehyde (MDA) and a decrease in intracellular glutathione (GSH). MDA and GSH depletion/modulation are crucial in inducing ferroptosis, suggesting that PDT may have the potential to induce this kind of cell death through these ways. A selective induction of programmed cell death in cancer cells is one of the main treatments for malignant tumors; thus, ferroptosis may represent a novel therapeutic strategy against HCC and CC.
Collapse
|
61
|
Baltacı NG, Toraman E, Akyüz M, Kalın ŞN, Budak H. Tip60/Kat5 may be a novel candidate histone acetyltransferase for the regulation of liver iron localization via acetylation. Biometals 2022; 35:1187-1197. [PMID: 35986817 DOI: 10.1007/s10534-022-00435-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/10/2022] [Indexed: 12/14/2022]
Abstract
Hepcidin (HAMP), an iron regulatory hormone synthesized by liver hepatocytes, works together with ferritin (FTH) and ferroportin (FPN) in regulating the storage, transport, and utilization of iron in the cell. Epigenetic mechanisms, especially acetylation, also play an important role in the regulation of iron metabolism. However, a target protein has not been mentioned yet. With this preliminary study, we investigated the effect of histone acetyltransferase TIP60 on the expression of HAMP, FTH, and FPN. In addition, how the depletion of Tip60, which regulates the circadian system, affects the daily expression of Hamp was examined at six Zeitgeber time (ZT) points. For this purpose, liver-specific Tip60 knockout mice (mutant) were produced with tamoxifen-inducible Cre/lox recombination and an iron overload model in mice was generated. While HAMP and FTH expressions decreased, FPN expression increased in the mutant group. Interestingly, there was no change in the iron content. A significant increase was observed in the expressions of HAMP, FTH, and FPN and total liver iron content in the liver tissue of the iron overload group. Since intracellular iron concentration is involved in regulating the circadian clock, temporal expression of Hamp was investigated in control and mutant groups at six ZT points. In the control group, Hamp accumulated in a circadian manner with maximal and minimal levels reaching around ZT16 and ZT8, respectively. In the mutant group, there was a significant reduction in Hamp expression in the light phase ZT0 and ZT4 and in the dark phase ZT16. These data are the first findings demonstrating a possible relationship between Tip60 and iron metabolism.
Collapse
Affiliation(s)
- Nurdan Gönül Baltacı
- Department of Molecular Biology and Genetics, Science Faculty, Atatürk University, 25240, Erzurum, Türkiye
| | - Emine Toraman
- Department of Molecular Biology and Genetics, Science Faculty, Atatürk University, 25240, Erzurum, Türkiye
| | - Mesut Akyüz
- Department of Molecular Biology and Genetics, Science Faculty, Atatürk University, 25240, Erzurum, Türkiye
- Department of Molecular Biology and Genetics, Science Faculty, Erzurum Technical University, Erzurum, Türkiye
| | - Şeyda Nur Kalın
- Department of Molecular Biology and Genetics, Science Faculty, Atatürk University, 25240, Erzurum, Türkiye
| | - Harun Budak
- Department of Molecular Biology and Genetics, Science Faculty, Atatürk University, 25240, Erzurum, Türkiye.
| |
Collapse
|
62
|
Infanti L, Leitner G, Moe MK, Pehlic V, Benkert P, Cattaneo M, Holbro A, Passweg J, Worel N, Buser A. Indices of iron homeostasis in asymptomatic subjects with HFE mutations and moderate ferritin elevation during iron removal treatment. Blood Cells Mol Dis 2022; 97:102689. [DOI: 10.1016/j.bcmd.2022.102689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 10/31/2022]
|
63
|
Gehrer CM, Hoffmann A, Hilbe R, Grubwieser P, Mitterstiller AM, Talasz H, Fang FC, Meyron-Holtz EG, Atkinson SH, Weiss G, Nairz M. Availability of Ferritin-Bound Iron to Enterobacteriaceae. Int J Mol Sci 2022; 23:13087. [PMID: 36361875 PMCID: PMC9657528 DOI: 10.3390/ijms232113087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/10/2023] Open
Abstract
The sequestration of iron in case of infection, termed nutritional immunity, is an established strategy of host defense. However, the interaction between pathogens and the mammalian iron storage protein ferritin is hitherto not completely understood. To better characterize the function of ferritin in Gram-negative infections, we incubated iron-starved cultures of Salmonella Typhimurium and knockout mutant strains defective for major iron uptake pathways or Escherichia coli with horse spleen ferritin or ionic iron as the sole iron source. Additionally, we added bovine superoxide dismutase and protease inhibitors to the growth medium to assess the effect of superoxide and bacterial proteases, respectively, on Salmonella proliferation and reductive iron release. Compared to free ionic iron, ferritin-bound iron was less available to Salmonella, but was still sufficient to significantly enhance the growth of the bacteria. In the absence of various iron acquisition genes, the availability of ferritin iron further decreased. Supplementation with superoxide dismutase significantly reduced the growth of the ΔentC knockout strain with holoferritin as the sole iron source in comparison with ionic ferrous iron. In contrast, this difference was not observed in the wildtype strain, suggesting that superoxide dismutase undermines bacterial iron uptake from ferritin by siderophore-independent mechanisms. Ferritin seems to diminish iron availability for bacteria in comparison to ionic iron, and its iron sequestering effect could possibly be enhanced by host superoxide dismutase activity.
Collapse
Affiliation(s)
- Clemens M. Gehrer
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Alexander Hoffmann
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Richard Hilbe
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Philipp Grubwieser
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Anna-Maria Mitterstiller
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Heribert Talasz
- Biocenter, Institute of Medical Biochemistry, Medical Universitiy of Innsbruck, 6020 Innsbruck, Austria
| | - Ferric C. Fang
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98195-7110, USA
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98195-7735, USA
| | - Esther G. Meyron-Holtz
- Laboratory of Molecular Nutrition, Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Sarah H. Atkinson
- Kenya Medical Research Institute (KEMRI), Centre for Geographic Medicine Research Coast, KEMRI-Wellcome Trust Research Programme, Kilifi 80108, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7LG, UK
- Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
64
|
Oliveira PMCD, Espósito BP, Carvente C, Silva GA, Dellavance A, Baldo DC, Ferraz MLG. Transferrin saturation as a surrogate marker for assessment of labile nontransferrin bound iron in chronic liver disease. Eur J Gastroenterol Hepatol 2022; 34:1047-1052. [PMID: 36052686 DOI: 10.1097/meg.0000000000002416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND Increased transferrin saturation (TS) and ferritin are common in hereditary hemochromatosis (HH) but also in chronic liver diseases (CLD). Nontransferrin bound iron (NTBI) is believed to be associated with iron-induced cell damage. We aimed to evaluate NTBI in CLD and their relationship with liver damage. METHODS Two groups of patients were studied. Group 1 (G1): 94 CLD patients from an Outpatient Hepatology Unit. Group 2 (G2): 36 healthy individuals form a Medical Checkup Clinic. Transferrin iron-binding capacity, TS, ferritin, AST, ALT, and red cell count were performed using standard tests. NTBI was assessed as enhanced labile plasma iron (eLPi). Levels of eLPi less than 0.4 µmol/l were considered within the normal range. RESULTS Prevalence of increased iron tests (elevated TS and ferritin) was 14% in G1 and 5.5% in G2 ( P = 0.19). Positive NTBI was found in 12 patients (11 in G1 and 1 in G2). Positivity to NTBI was associated with increased iron tests ( P = 0.03), cirrhosis ( P = 0.03) and AST index (ASTI) ( P = 0.03). NTBI was associated with TS of more than 70% ( P = 0.002) but not to elevated ferritin ( P = 0.74). Variables strongly associated with a positive NTBI in univariate analysis (TS > 70%, cirrhosis and ASTI) were submitted to binary regression analysis. TS of more than 70% was the only independent predictive factor ( P = 0.049; odds ratio, 6.8). CONCLUSION NTBI was associated with TS in CLD, but not with ferritin. NTBI testing could be useful for CLD patients with increased iron tests. Alternatively, a TS of more than 70% can be used as a surrogate marker.
Collapse
Affiliation(s)
| | | | - Claudia Carvente
- Gastroenterology Division, Federal University of São Paulo (UNIFESP)
| | | | | | | | | |
Collapse
|
65
|
Jomova K, Makova M, Alomar SY, Alwasel SH, Nepovimova E, Kuca K, Rhodes CJ, Valko M. Essential metals in health and disease. Chem Biol Interact 2022; 367:110173. [PMID: 36152810 DOI: 10.1016/j.cbi.2022.110173] [Citation(s) in RCA: 323] [Impact Index Per Article: 107.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/10/2022] [Accepted: 09/05/2022] [Indexed: 11/03/2022]
Abstract
In total, twenty elements appear to be essential for the correct functioning of the human body, half of which are metals and half are non-metals. Among those metals that are currently considered to be essential for normal biological functioning are four main group elements, sodium (Na), potassium (K), magnesium (Mg), and calcium (Ca), and six d-block transition metal elements, manganese (Mn), iron (Fe), cobalt (Co), copper (Cu), zinc (Zn) and molybdenum (Mo). Cells have developed various metallo-regulatory mechanisms for maintaining a necessary homeostasis of metal-ions for diverse cellular processes, most importantly in the central nervous system. Since redox active transition metals (for example Fe and Cu) may participate in electron transfer reactions, their homeostasis must be carefully controlled. The catalytic behaviour of redox metals which have escaped control, e.g. via the Fenton reaction, results in the formation of reactive hydroxyl radicals, which may cause damage to DNA, proteins and membranes. Transition metals are integral parts of the active centers of numerous enzymes (e.g. Cu,Zn-SOD, Mn-SOD, Catalase) which catalyze chemical reactions at physiologically compatible rates. Either a deficiency, or an excess of essential metals may result in various disease states arising in an organism. Some typical ailments that are characterized by a disturbed homeostasis of redox active metals include neurological disorders (Alzheimer's, Parkinson's and Huntington's disorders), mental health problems, cardiovascular diseases, cancer, and diabetes. To comprehend more deeply the mechanisms by which essential metals, acting either alone or in combination, and/or through their interaction with non-essential metals (e.g. chromium) function in biological systems will require the application of a broader, more interdisciplinary approach than has mainly been used so far. It is clear that a stronger cooperation between bioinorganic chemists and biophysicists - who have already achieved great success in understanding the structure and role of metalloenzymes in living systems - with biologists, will access new avenues of research in the systems biology of metal ions. With this in mind, the present paper reviews selected chemical and biological aspects of metal ions and their possible interactions in living systems under normal and pathological conditions.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences and Informatics, Constantine The Philosopher University in Nitra, 949 01, Nitra, Slovakia
| | - Marianna Makova
- Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, 812 37, Bratislava, Slovakia
| | - Suliman Y Alomar
- King Saud University, Zoology Department, College of Science, Riyadh, 11451, Saudi Arabia
| | - Saleh H Alwasel
- King Saud University, Zoology Department, College of Science, Riyadh, 11451, Saudi Arabia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic; Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | | | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, 812 37, Bratislava, Slovakia; King Saud University, Zoology Department, College of Science, Riyadh, 11451, Saudi Arabia.
| |
Collapse
|
66
|
Genetic Causal Association between Iron Status and Osteoarthritis: A Two-Sample Mendelian Randomization. Nutrients 2022; 14:nu14183683. [PMID: 36145059 PMCID: PMC9501024 DOI: 10.3390/nu14183683] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Objective: Observational studies have shown the association between iron status and osteoarthritis (OA). However, due to difficulties of determining sequential temporality, their causal association is still elusive. Based on the summary data of genome-wide association studies (GWASs) of a large-scale population, this study explored the genetic causal association between iron status and OA. Methods: First, we took a series of quality control steps to select eligible instrumental SNPs which were strongly associated with exposure. The genetic causal association between iron status and OA was analyzed using the two-sample Mendelian randomization (MR). Inverse-variance weighted (IVW), MR-Egger, weighted median, simple mode, and weighted mode methods were used for analysis. The results were mainly based on IVW (random effects), followed by sensitivity analysis. IVW and MR-Egger were used for heterogeneity testing. MR-Egger was also used for pleiotropy testing. Leave-one-SNP-out analysis was used to identify single nucleotide polymorphisms (SNPs) with potential impact. Maximum likelihood, penalized weighted median, and IVW (fixed effects) were performed to further validate the reliability of results. Results: IVW results showed that transferrin saturation had a positive causal association with knee osteoarthritis (KOA), hip osteoarthritis (HOA) and KOA or HOA (p < 0.05, OR > 1), and there was a negative causal association between transferrin and HOA and KOA or HOA (p < 0.05, OR < 1). The results of heterogeneity test showed that our IVW analysis results were basically free of heterogeneity (p > 0.05). The results of the pleiotropy test showed that there was no pleiotropy in our IVW analysis (p > 0.05). The analysis results of maximum likelihood, penalized weighted median and IVW (fixed effects) were consistent with our IVW results. No genetic causal association was found between serum iron and ferritin and OA. Conclusions: This study provides evidence of the causal association between iron status and OA, which provides novel insights to the genetic research of OA.
Collapse
|
67
|
Wang JW, Jin CH, Ke JF, Ma YL, Wang YJ, Lu JX, Li MF, Li LX. Serum iron is closely associated with metabolic dysfunction-associated fatty liver disease in type 2 diabetes: A real-world study. Front Endocrinol (Lausanne) 2022; 13:942412. [PMID: 36133303 PMCID: PMC9484008 DOI: 10.3389/fendo.2022.942412] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS There is still a debate about the relationship between serum iron and metabolic dysfunction-associated fatty liver disease (MAFLD). Furthermore, few relevant studies were conducted in type 2 diabetes mellitus (T2DM). Therefore, this study aimed to explore the association of serum iron levels with MAFLD in Chinese patients with T2DM. METHODS This cross-sectional, real-world study consisted of 1,467 Chinese T2DM patients. MAFLD was diagnosed by abdominal ultrasonography. Based on serum iron quartiles, the patients were classified into four groups. Clinical characteristics were compared among the four groups, and binary logistic analyses were used to assess the associations of serum iron levels and quartiles with the presence of MAFLD in T2DM. RESULTS After adjusting for gender, age, and diabetes duration, significantly higher prevalence of MAFLD was found in the second (45.7%), third (45.2%), and fourth (47.0%) serum iron quartiles than in the first quartiles (26.8%), with the highest MAFLD prevalence in the fourth quartile (p < 0.001 for trend). Moreover, increased HOMA2-IR (p = 0.003 for trend) and decreased HOMA2-S (p = 0.003 for trend) were observed across the serum iron quartiles. Fully adjusted binary logistic regression analyses indicated that both increased serum iron levels (OR: 1.725, 95% CI: 1.427 to 2.085, p < 0.001) and quartiles (p < 0.001 for trend) were still closely associated with the presence of MAFLD in T2DM patients even after controlling for multiple confounding factors. CONCLUSIONS There is a positive correlation between the presence of MAFLD and serum iron levels in T2DM patients, which may be attributed to the close association between serum iron and insulin resistance. Serum iron levels may act as one of the indicators for evaluating the risk of MAFLD in T2DM individuals.
Collapse
Affiliation(s)
- Jun-Wei Wang
- 1Department of Endocrinology and Metabolism, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Chun-Hua Jin
- Department of Endocrinology and Metabolism, Shanghai Songjiang District Central Hospital, Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine (Preparatory Stage), Shanghai, China
| | - Jiang-Feng Ke
- 1Department of Endocrinology and Metabolism, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Yi-Lin Ma
- 1Department of Endocrinology and Metabolism, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Yu-Jie Wang
- 1Department of Endocrinology and Metabolism, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Jun-Xi Lu
- 1Department of Endocrinology and Metabolism, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Mei-Fang Li
- Department of Emergency, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lian-Xi Li
- 1Department of Endocrinology and Metabolism, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| |
Collapse
|
68
|
Venkatesan P, Ramasamy J, Vanitha S, Jacob M, Varghese J. Impaired pancreatic beta-cell function after a single dose of oral iron: a before-and-after (pre-post) study. J Hum Nutr Diet 2022; 36:1111-1120. [PMID: 36000222 DOI: 10.1111/jhn.13074] [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: 07/01/2022] [Accepted: 07/25/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Although in vitro and animal studies have shown that iron loading in pancreatic beta-cells impaired insulin secretion, no human studies have documented the acute effects of oral iron on beta-cell insulin secretory capacity. In this study, we determined beta-cell insulin secretory capacity at baseline and after a single oral dose of iron (ferrous sulphate, 120 mg elemental iron) in healthy male individuals. METHODS Fifteen healthy male volunteers underwent an oral glucose tolerance test (OGTT) to document baseline glucose tolerance and insulin secretion kinetics (baseline OGTT). One week later, the same subjects underwent a second OGTT, two hours after an oral dose of ferrous sulfate (120 mg of elemental iron) (post-iron OGTT). Changes in disposition index, insulin secretion kinetics, glucose tolerance, insulin resistance, insulin clearance, and iron-related parameters in serum were determined. RESULTS Compared to baseline OGTT, the areas under the curve (AUC) for serum iron and transferrin saturation increased by 125% and 118% respectively, in the post-iron OGTT. The disposition index decreased by 20% (p=0.009) and the AUC for glucose concentrations increased by 5.7% (p<0.001) during the post-iron OGTT. The insulin secretion rate was marginally lower during the first hour (-3.5%, p=0.63), but became significantly higher during the second hour (22%, p=0.005) of the post-iron OGTT. Insulin resistance and insulin clearance rate were not affected by iron intake. CONCLUSION The decrease in disposition index and glucose tolerance observed after the oral dose of iron points to an acute iron-induced impairment in pancreatic beta-cell insulin secretory capacity. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Padmanaban Venkatesan
- Departments of Biochemistry, Christian Medical College, Vellore, Tamil Nadu, India, 632002.,Departments of Christian Medical College, The Tamil Nadu Dr. MGR Medical University, Chennai, India
| | - Jagadish Ramasamy
- Departments of Biochemistry, Christian Medical College, Vellore, Tamil Nadu, India, 632002.,Departments of Christian Medical College, The Tamil Nadu Dr. MGR Medical University, Chennai, India
| | - S Vanitha
- Departments of Clinical Biochemistry, Christian Medical College, Vellore, Tamil Nadu, India, 632002.,Departments of Christian Medical College, The Tamil Nadu Dr. MGR Medical University, Chennai, India
| | - Molly Jacob
- Departments of Biochemistry, Christian Medical College, Vellore, Tamil Nadu, India, 632002.,Departments of Christian Medical College, The Tamil Nadu Dr. MGR Medical University, Chennai, India
| | - Joe Varghese
- Departments of Biochemistry, Christian Medical College, Vellore, Tamil Nadu, India, 632002.,Departments of Christian Medical College, The Tamil Nadu Dr. MGR Medical University, Chennai, India
| |
Collapse
|
69
|
Lokesh KN, Raichur AM. Bioactive nutraceutical ligands and their efficiency to chelate elemental iron of varying dynamic oxidation states to mitigate associated clinical conditions. Crit Rev Food Sci Nutr 2022; 64:517-543. [PMID: 35943179 DOI: 10.1080/10408398.2022.2106936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The natural bioactive or nutraceuticals exhibit several health benefits, including anti-inflammatory, anti-cancer, metal chelation, antiviral, and antimicrobial activity. The inherent limitation of nutraceuticals or bioactive ligand(s) in terms of poor pharmacokinetic and other physicochemical properties affects their overall therapeutic efficiency. The excess of iron in the physiological compartments and its varying dynamic oxidation state [Fe(II) and Fe(III)] precipitates various clinical conditions such as non-transferrin bound iron (NTBI), labile iron pool (LIP), ferroptosis, cancer, etc. Though several natural bioactive ligands are proposed to chelate iron, the efficiency of bioactive ligands is limited due to poor bioavailability, denticity, and other related physicochemical properties. The present review provides insight into the relevance of studying the dynamic oxidation state of iron(II) and iron(III) in the physiological compartments and its clinical significance for selecting diagnostics and therapeutic regimes. We suggested a three-pronged approach, i.e., diagnosis, selection of therapeutic regime (natural bioactive), and integration of novel drug delivery systems (NDDS) or nanotechnology-based principles. This systematic approach improves the overall therapeutic efficiency of natural iron chelators to manage iron overload-related clinical conditions.
Collapse
Affiliation(s)
- K N Lokesh
- Department of Biotechnology, Ramaiah Institute of Technology, Bengaluru, Karnataka, India
| | - Ashok M Raichur
- Department of Materials Engineering, Indian Institute of Science, Bengaluru, Karnataka, India
| |
Collapse
|
70
|
Fishbane S, Ganz T, Pratt RD. Ferric pyrophosphate citrate for parenteral administration of maintenance iron: structure, mechanism of action, clinical efficacy and safety. Curr Med Res Opin 2022; 38:1417-1429. [PMID: 35726771 DOI: 10.1080/03007995.2022.2092373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Anemia is a common complication in patients with hemodialysis-dependent chronic kidney disease (HDD-CKD). Anemia is principally the result of erythropoietin deficiency, inflammation, and iron deficiency. High molecular weight iron oxide nanoparticles (IONP) are routinely administered intravenously to replace iron losses and, although effective, there are lingering concerns about possible safety issues. Ferric pyrophosphate citrate (FPC, Triferic, Triferic AVNU [Triferic and Triferic AVNU are the proprietary name for ferric pyrophosphate citrate. Triferic and Triferic AVNU are registered trademarks of Rockwell medical Inc.]) is a complex iron salt that donates iron directly to plasma transferrin. FPC is devoid of any carbohydrate moiety and is administered via the dialysate or intravenously during each hemodialysis session to replace iron and maintain hemoglobin. Controlled clinical trials of up to 48 weeks in duration have demonstrated the efficacy of regular administration of dialysate FPC for maintaining hemoglobin levels and iron balance in HDD-CKD patients. Clinical data also suggest that dialysate FPC may reduce the dose requirements for and use of erythropoiesis-stimulating agents and IONPs in HDD-CKD patients. Safety data from clinical studies and post-marketing surveillance show that FPC is well tolerated and not associated with an increased risk of infection, inflammation, iron overload, or serious hypersensitivity reactions. FPC represents an effective and well-tolerated choice for iron replacement and maintenance of hemoglobin in the long-term management of HDD-CKD patients.
Collapse
Affiliation(s)
- Steven Fishbane
- Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY, USA
| | - Tomas Ganz
- University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | | |
Collapse
|
71
|
Ryan E, Mulready K, Wiegerinck E, Russell J, Swinkels DW, Stewart S. NTBI levels in C282Y homozygotes after therapeutic phlebotomy. EJHAEM 2022; 3:644-652. [PMID: 36051052 PMCID: PMC9422009 DOI: 10.1002/jha2.507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022]
Abstract
C282Y homozygotes exposed to sustained elevated transferrin saturation (TS) may develop worsening clinical symptoms. This might be related to the appearance of non-transferrin bound iron (NTBI) when TS≥50% and labile plasma iron (LPI) when TS levels reach 75-80%. In this study, NTBI levels were examined in 219 randomly selected untreated and treated C282Y homozygotes. Overall, 161 of 219 had TS ≥ 50%, 124 of whom had detectable NTBI (≥0.47 µM, 1.81 µM [0.92-2.46 µM]) with a median serum ferritin 320 µg/L (226-442 µg/L). Ninety of 219 homozygotes had TS ≥ 75%, and all had detectable NTBI (2.21 µM [1.53-2.59 µM] with a median ferritin 338 µg/L [230-447 µg/L]). Of 125 homozygotes who last had phlebotomy ≥12 months ago (42 months [25-74 months], 92 had TS levels ≥ 50%, and 70 of these had NTBI ≥ 0.47 µM (2.06 µM [1.23-2.61µM]). Twenty-six of these 70 had a normal ferritin. Fifty-five of 125 had TS ≥ 75%, and NTBI was detected in all of these (2.32 µM [1.57-2.77 µM]) with a median ferritin 344 µg/L (255-418 µg/L). Eighteen of these 55 had a normal ferritin. In summary, NTBI is frequently found in C282Y homozygotes with TS ≥ 50%. Furthermore, C282Y homozygotes in the maintenance phase often have TS ≥ 50% together with a normal ferritin. Therefore, monitoring the TS level during the maintenance phase is recommended as an accessible clinical marker of the presence of NTBI.
Collapse
Affiliation(s)
- Eleanor Ryan
- Liver CentreMater Misericordiae University HospitalDublinIreland
| | - Keith Mulready
- Department of Biochemistry and Diagnostic EndocrinologyMater Misericordiae University HospitalDublinIreland
| | - Erwin Wiegerinck
- Laboratory of Genetic, Endocrine and Metabolic Diseases, Department of Laboratory MedicineRadboud University Medical CentreNijmegenThe Netherlands
| | - Jennifer Russell
- Liver CentreMater Misericordiae University HospitalDublinIreland
| | - Dorine W. Swinkels
- Laboratory of Genetic, Endocrine and Metabolic Diseases, Department of Laboratory MedicineRadboud University Medical CentreNijmegenThe Netherlands
| | - Stephen Stewart
- Liver CentreMater Misericordiae University HospitalDublinIreland
| |
Collapse
|
72
|
Emerging Roles of the Iron Chelators in Inflammation. Int J Mol Sci 2022; 23:ijms23147977. [PMID: 35887336 PMCID: PMC9318075 DOI: 10.3390/ijms23147977] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 02/06/2023] Open
Abstract
Iron is a crucial element for mammalian cells, considering its intervention in several physiologic processes. Its homeostasis is finely regulated, and its alteration could be responsible for the onset of several disorders. Iron is closely related to inflammation; indeed, during inflammation high levels of interleukin-6 cause an increased production of hepcidin which induces a degradation of ferroportin. Ferroportin degradation leads to decreased iron efflux that culminates in elevated intracellular iron concentration and consequently iron toxicity in cells and tissues. Therefore, iron chelation could be considered a novel and useful therapeutic strategy in order to counteract the inflammation in several autoimmune and inflammatory diseases. Several iron chelators are already known to have anti-inflammatory effects, among them deferiprone, deferoxamine, deferasirox, and Dp44mT are noteworthy. Recently, eltrombopag has been reported to have an important role in reducing inflammation, acting both directly by chelating iron, and indirectly by modulating iron efflux. This review offers an overview of the possible novel biological effects of the iron chelators in inflammation, suggesting them as novel anti-inflammatory molecules.
Collapse
|
73
|
Chio JCT, Punjani N, Hejrati N, Zavvarian MM, Hong J, Fehlings MG. Extracellular Matrix and Oxidative Stress Following Traumatic Spinal Cord Injury: Physiological and Pathophysiological Roles and Opportunities for Therapeutic Intervention. Antioxid Redox Signal 2022; 37:184-207. [PMID: 34465134 DOI: 10.1089/ars.2021.0120] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: Traumatic spinal cord injury (SCI) causes significant disruption to neuronal, glial, vascular, and extracellular elements. The spinal cord extracellular matrix (ECM) comprises structural and communication proteins that are involved in reparative and regenerative processes after SCI. In the healthy spinal cord, the ECM helps maintain spinal cord homeostasis. After SCI, the damaged ECM limits plasticity and contributes to inflammation through the expression of damage-associated molecules such as proteoglycans. Recent Advances: Considerable insights have been gained by characterizing the origins of the gliotic and fibrotic scars, which not only reduce the spread of injury but also limit neuroregeneration. These properties likely limit the success of therapies used to treat patients with SCI. The ECM, which is a major contributor to the scars and normal physiological functions of the spinal cord, represents an exciting therapeutic target to enhance recovery post-SCI. Critical Issue: Various ECM-based preclinical therapies have been developed. These include disrupting scar components, inhibiting activity of ECM metalloproteinases, and maintaining iron homeostasis. Biomaterials have also been explored. However, the majority of these treatments have not experienced successful clinical translation. This could be due to the ECM and scars' polarizing roles. Future Directions: This review surveys the complexity involved in spinal ECM modifications, discusses new ECM-based combinatorial strategies, and explores the biomaterials evaluated in clinical trials, which hope to introduce new treatments that enhance recovery after SCI. These topics will incorporate oxidative species, which are both beneficial and harmful in reparative and regenerative processes after SCI, and not often assessed in pertinent literature. Antioxid. Redox Signal. 37, 184-207.
Collapse
Affiliation(s)
- Jonathon Chon Teng Chio
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Nayaab Punjani
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Nader Hejrati
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada
| | - Mohammad-Masoud Zavvarian
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - James Hong
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada
| | - Michael G Fehlings
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada.,Department of Surgery and Spine Program, University of Toronto, Toronto, Canada
| |
Collapse
|
74
|
Choi YK, Kim YM. Beneficial and Detrimental Roles of Heme Oxygenase-1 in the Neurovascular System. Int J Mol Sci 2022; 23:ijms23137041. [PMID: 35806040 PMCID: PMC9266949 DOI: 10.3390/ijms23137041] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
Heme oxygenase (HO) has both beneficial and detrimental effects via its metabolites, including carbon monoxide (CO), biliverdin or bilirubin, and ferrous iron. HO-1 is an inducible form of HO that is upregulated by oxidative stress, nitric oxide, CO, and hypoxia, whereas HO-2 is a constitutive form that regulates vascular tone and homeostasis. In brains injured by trauma, ischemia-reperfusion, or Alzheimer’s disease (AD), the long-term expression of HO-1 can be detected, which can lead to cytotoxic ferroptosis via iron accumulation. In contrast, the transient induction of HO-1 in the peri-injured region may have regenerative potential (e.g., angiogenesis, neurogenesis, and mitochondrial biogenesis) and neurovascular protective effects through the CO-mediated signaling pathway, the antioxidant properties of bilirubin, and the iron-mediated ferritin synthesis. In this review, we discuss the dual roles of HO-1 and its metabolites in various neurovascular diseases, including age-related macular degeneration, ischemia-reperfusion injury, traumatic brain injury, Gilbert’s syndrome, and AD.
Collapse
Affiliation(s)
- Yoon Kyung Choi
- Bio/Molecular Informatics Center, Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
- Correspondence: (Y.K.C.); (Y.-M.K.); Tel.: +82-2-450-0558 (Y.K.C.); +82-33-250-8831 (Y.-M.K.)
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
- Correspondence: (Y.K.C.); (Y.-M.K.); Tel.: +82-2-450-0558 (Y.K.C.); +82-33-250-8831 (Y.-M.K.)
| |
Collapse
|
75
|
Wang H, Jiang C, Yang Y, Li J, Wang Y, Wang C, Gao Y. Resveratrol ameliorates iron overload induced liver fibrosis in mice by regulating iron homeostasis. PeerJ 2022; 10:e13592. [PMID: 35698613 PMCID: PMC9188311 DOI: 10.7717/peerj.13592] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/24/2022] [Indexed: 01/17/2023] Open
Abstract
This study is intended to explore the protective effects of resveratrol (RES) on iron overload-induced liver fibrosis and its mechanism. Iron dextran (50 mg/kg) was injected intraperitoneally in all groups except the control group. Mice in the L-RES, M-RES and H-RES groups were gavaged with RES solution at 25, 50 mg/kg and 100 mg/kg, respectively, 4 h before injection of iron dextran every day; mice in the deferoxamine (DFO) group were injected with DFO intraperitoneally (100 mg/kg); mice in the control group received isovolumetric saline. After seven weeks of RES administration, serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) activities and liver hydroxyproline (Hyp) levels were reduced; the malondialdehyde (MDA) activities decreased and the levels of superoxide dismutase (SOD) and glutathione (GSH) were raised. Hematoxylin and eosin (H&E), Prussian, and Masson staining indicated that RES treatment could improve cell damage and reduce hepatic iron deposition and collagen deposition in iron-overload mice. The expression of Bcl-2 was increased, the expression levels of Bax and caspase-3 were decreased under RES treatment. Moreover, RES reduced the expression of hepcidin, ferritin (Ft), divalent metal transporter-1 (DMT-1), transferrin receptor-2 (TFR-2), and raised the expression of ferroprotein-1 (FPN-1). In conclusion, RES could ameliorate iron overload-induced liver fibrosis, and the potential mechanisms may be related to antioxidant, anti-inflammatory, anti-apoptotic, and more importantly, regulation of iron homeostasis by reducing iron uptake and increasing iron export.
Collapse
Affiliation(s)
- Hua Wang
- Hebei University of Chinese Medicine, Department of Preventive Medicine, Shijiazhuang, Hebei, China
| | - Chuan Jiang
- Hebei University of Chinese Medicine, Department of Preventive Medicine, Shijiazhuang, Hebei, China
| | - Yakun Yang
- Hebei University of Chinese Medicine, School of Pharmacy, Shijiazhuang, Hebei, China
| | - Jinghan Li
- Hebei University of Chinese Medicine, Department of Preventive Medicine, Shijiazhuang, Hebei, China
| | - Yihan Wang
- Hebei University of Chinese Medicine, Collge of Basic Medicine, Shijiazhuang, Hebei, China
| | - Chaonan Wang
- Hebei University of Chinese Medicine, Department of Preventive Medicine, Shijiazhuang, Hebei, China
| | - Yonggang Gao
- Hebei University of Chinese Medicine, Department of Preventive Medicine, Shijiazhuang, Hebei, China,Hebei Key laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, China
| |
Collapse
|
76
|
Angoro B, Motshakeri M, Hemmaway C, Svirskis D, Sharma M. Non-transferrin bound iron. Clin Chim Acta 2022; 531:157-167. [DOI: 10.1016/j.cca.2022.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 12/31/2022]
|
77
|
Park SH, Kim RS, Stiles WR, Jo M, Zeng L, Rho S, Baek Y, Kim J, Kim MS, Kang H, Choi HS. Injectable Thermosensitive Hydrogels for a Sustained Release of Iron Nanochelators. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200872. [PMID: 35343104 PMCID: PMC9130884 DOI: 10.1002/advs.202200872] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Indexed: 05/17/2023]
Abstract
Deferoxamine (DFO) is an FDA-approved iron-chelating agent which shows good therapeutic efficacy, however, its short blood half-life presents challenges such as the need for repeated injections or continuous infusions. Considering the lifelong need of chelating agents for iron overload patients, a sustained-release formulation that can reduce the number of chelator administrations is essential. Here, injectable hydrogel formulations prepared by integrating crosslinked hyaluronic acid into Pluronic F127 for an extended release of DFO nanochelators are reported. The subcutaneously injected hydrogel shows a thermosensitive sol-gel transition at physiological body temperature and provides a prolonged release of renal clearable nanochelators over 2 weeks, resulting in a half-life 47-fold longer than that of the nanochelator alone. In addition, no chronic toxicity of the nanochelator-loaded hydrogel is confirmed by biochemical and histological analyses. This injectable hydrogel formulation with DFO nanochelators has the potential to be a promising formulation for the treatment of iron overload disorders.
Collapse
Affiliation(s)
- Seung Hun Park
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
- Department of Molecular Science and TechnologyAjou UniversitySuwon16499South Korea
| | - Richard S. Kim
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Wesley R. Stiles
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Minjoo Jo
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Lingxue Zeng
- Department of Biomedical & Nutritional SciencesZuckerberg College of Health SciencesUniversity of MassachusettsLowellMA01854USA
| | - Sunghoon Rho
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Yoonji Baek
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Jonghan Kim
- Department of Biomedical & Nutritional SciencesZuckerberg College of Health SciencesUniversity of MassachusettsLowellMA01854USA
| | - Moon Suk Kim
- Department of Molecular Science and TechnologyAjou UniversitySuwon16499South Korea
| | - Homan Kang
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Hak Soo Choi
- Gordon Center for Medical ImagingDepartment of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| |
Collapse
|
78
|
Guo L, Zhang D, Liu S, Dong Z, Zhou J, Yin Y, Wan D. Maternal iron supplementation during pregnancy affects placental function and iron status in offspring. J Trace Elem Med Biol 2022; 71:126950. [PMID: 35183047 DOI: 10.1016/j.jtemb.2022.126950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/12/2021] [Accepted: 02/10/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND Iron deficiency and overload during pregnancy damage to maternal and fetal health. Placenta as an organ for the transport of nutrients between mother and fetus protects fetus from the harmful effects of iron deficiency and iron overload through regulation of placental iron homeostasis. METHODS To determine the effect of dietary iron supplementation during pregnancy on reproduction and the mechanism of placental iron regulation, we designed dietary high iron (HI: 344 mg/kg), medium iron (MI: 40 mg/kg), low iron (LI: 2 mg/kg) groups of pregnant female mice fed ferrous citrate 2 weeks before mating to 18.5 days of gestation. RESULTS We find dietary iron supplementation during pregnancy effect maternal liver iron, placental iron, hemoglobin and fetal iron. Dietary iron significantly improves reproductive performance as litter weight and fetal weight. Correlation analysis suggest placental iron increased with liver iron, higher and lower liver iron is not conducive to the accumulation of fetal iron, placental iron deficiency and excess reduce litter weight. Placental transcriptome analysis revealed DEGs with the same trend in HI and LI groups compared with MI group, dietary iron may change biology process of ion transport and gland development in placenta. Granzyme may affect the placental trophoblast structure prior to delivery with iron overload uniquely. CONCLUSION This research highlights the importance of moderate iron supplements in pregnancy due to damage of reproduction by affecting placental function under different dose of maternal iron supplementation.
Collapse
Affiliation(s)
- Liu Guo
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China; University of Chinese Academy of Sciences, Beijing, China
| | - Dongming Zhang
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China
| | - Shuan Liu
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zhenglin Dong
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China
| | - Jian Zhou
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China
| | - Dan Wan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan 410125, China.
| |
Collapse
|
79
|
Hać PJ, Cieślik BM, Konieczka P. Review of cigars and cigar-type products as potential sources of consumer exposure to heavy metals. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2022; 40:172-196. [PMID: 35895917 DOI: 10.1080/26896583.2022.2052708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The popularity of cigars, growing since 1993, has not gone hand in hand with the increased interest of researchers in these products. Although the literature widely describes the harmfulness of tobacco and the content of toxic substances in tobacco products, the topic is often treated selectively as relating primarily to cigarettes and rarely extends to other products of the broadly defined tobacco industry. However, there is no reason to marginalize the harmful effects of other nicotine products, (which include tobacco products such as cigars). The study analyzed the available literature on the content of selected heavy metals in cigar tobacco. Among the heavy metals, the following contents of elements in tobacco were recorded in cigars: Fe (420-2200 µg/g), Mn (100-370 µg/g), Zn (14-180 µg/g), Cu (15-140 µg/g), Pb (not detected-32 µg/g), Cd (nd-19 µg/g), Ni (nd-13 µg/g), Cr (nd-10 µg/g), Co (0.65-1.0 µg/g), As (nd-0.66 µg/g), Hg (18-25 ng/g). Importantly, the values often differ between cigars of different origins and types, indicating the need for more extensive research.
Collapse
Affiliation(s)
- Paweł Jacek Hać
- Faculty of Chemistry, Department of Analytical Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Bartłomiej Michał Cieślik
- Faculty of Chemistry, Department of Analytical Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Piotr Konieczka
- Faculty of Chemistry, Department of Analytical Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| |
Collapse
|
80
|
Kunachowicz D, Ściskalska M, Jakubek M, Kizek R, Kepinska M. Structural changes in selected human proteins induced by exposure to quantum dots, their biological relevance and possible biomedical applications. NANOIMPACT 2022; 26:100405. [PMID: 35560289 DOI: 10.1016/j.impact.2022.100405] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/05/2022] [Accepted: 04/27/2022] [Indexed: 06/15/2023]
Abstract
Quantum dots (QDs) are semi-conductor luminescent nanocrystals usually of 2-10 nm diameter, attracting the significant attention in biomedical studies since emerged. Due to their unique optical and electronic properties, i.e. wide absorption spectra, narrow tunable emission bands or stable, bright photoluminescence, QDs seem to be ideally suited for multi-colour, simultaneous bioimaging and cellular labeling at the molecular level as new-generation probes. A highly reactive surface of QDs allows for conjugating them to biomolecules, what enables their direct binding to areas of interest inside or outside the cell for biosensing or targeted delivery. Particularly protein-QDs conjugates are current subjects of research, as features of QDs can be combined with protein specific functionalities and therefore used as a complex in variety of biomedical applications. It is known that QDs are able to interact with cells, organelles and macromolecules of the human body after administration. QDs are reported to cause changes at proteins level, including unfolding and three-dimensional structure alterations which might hamper proteins from performing their physiological functions and thereby limit the use of QD-protein conjugates in vivo. Moreover, these changes may trigger unwanted cellular outcomes as the effect of different signaling pathways activation. In this review, characteristics of QDs interactions with certain human proteins are presented and discussed. Besides that, the following manuscript provides an overview on structural changes of specific proteins exposed to QDs and their biological and biomedical relevance.
Collapse
Affiliation(s)
- Dominika Kunachowicz
- Department of Pharmaceutical Biochemistry, Division of Biomedical and Environmental Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland
| | - Milena Ściskalska
- Department of Pharmaceutical Biochemistry, Division of Biomedical and Environmental Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| | - Rene Kizek
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| | - Marta Kepinska
- Department of Pharmaceutical Biochemistry, Division of Biomedical and Environmental Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland.
| |
Collapse
|
81
|
Martin D, Nay K, Robin F, Rebillard A, Orfila L, Martin B, Leroyer P, Guggenbuhl P, Dufresne S, Noirez P, Ropert M, Loréal O, Derbré F. Oxidative and glycolytic skeletal muscles deploy protective mechanisms to avoid atrophy under pathophysiological iron overload. J Cachexia Sarcopenia Muscle 2022; 13:1250-1261. [PMID: 35118832 PMCID: PMC8978014 DOI: 10.1002/jcsm.12897] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/18/2021] [Accepted: 11/22/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Iron excess has been proposed as an essential factor in skeletal muscle wasting. Studies have reported correlations between muscle iron accumulation and atrophy, either through ageing or by using experimental models of secondary iron overload. However, iron treatments performed in most of these studies induced an extra-pathophysiological iron overload, more representative of intoxication or poisoning. The main objective of this study was to determine the impact of iron excess closer to pathophysiological conditions on structural and metabolic adaptations (i) in differentiated myotubes and (ii) in skeletal muscle exhibiting oxidative (i.e. the soleus) or glycolytic (i.e. the gastrocnemius) metabolic phenotypes. METHODS The impact of iron excess was assessed in both in vitro and in vivo models. Murine differentiated myotubes were exposed to ferric ammonium citrate (FAC) (i.e. 10 and 50 μM) for the in vitro component. The in vivo model was achieved by a single iron dextran subcutaneous injection (1 g/kg) in mice. Four months after the injection, soleus and gastrocnemius muscles were harvested for analysis. RESULTS In vitro, iron exposure caused dose-dependent increases of iron storage protein ferritin (P < 0.01) and dose-dependent decreases of mRNA TfR1 levels (P < 0.001), which support cellular adaptations to iron excess. Extra-physiological iron treatment (50 μM FAC) promoted myotube atrophy (P = 0.018), whereas myotube size remained unchanged under pathophysiological treatment (10 μM FAC). FAC treatments, whatever the doses tested, did not affect the expression of proteolytic markers (i.e. NF-κB, MurF1, and ubiquitinated proteins). In vivo, basal iron content and mRNA TfR1 levels were significantly higher in the soleus compared with the gastrocnemius (+130% and +127%; P < 0.001, respectively), supporting higher iron needs in oxidative skeletal muscle. Iron supplementation induced muscle iron accumulation in the soleus and gastrocnemius muscles (+79%, P < 0.001 and +34%, P = 0.002, respectively), but ferritin protein expression only increased in the gastrocnemius (+36%, P = 0.06). Despite iron accumulation, muscle weight, fibre diameter, and myosin heavy chain distribution remained unchanged in either skeletal muscle. CONCLUSIONS Together, these data support that under pathophysiological conditions, skeletal muscle can protect itself from the related deleterious effects of excess iron.
Collapse
Affiliation(s)
- David Martin
- Laboratory 'Movement, Sport and Health Sciences'-EA7470, University of Rennes/ENS Rennes, Bruz, France
| | - Kévin Nay
- Laboratory 'Movement, Sport and Health Sciences'-EA7470, University of Rennes/ENS Rennes, Bruz, France.,Exercise and Nutrition Research Program, Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Vic., Australia
| | - François Robin
- INSERM, INRAe, University of Rennes, Nutrition Metabolisms and Cancer Institute (NuMeCan), Platform AEM2, CHU Rennes, Rennes, France
| | - Amélie Rebillard
- Laboratory 'Movement, Sport and Health Sciences'-EA7470, University of Rennes/ENS Rennes, Bruz, France
| | - Luz Orfila
- Laboratory 'Movement, Sport and Health Sciences'-EA7470, University of Rennes/ENS Rennes, Bruz, France
| | - Brice Martin
- Laboratory 'Movement, Sport and Health Sciences'-EA7470, University of Rennes/ENS Rennes, Bruz, France
| | - Patricia Leroyer
- INSERM, INRAe, University of Rennes, Nutrition Metabolisms and Cancer Institute (NuMeCan), Platform AEM2, CHU Rennes, Rennes, France
| | - Pascal Guggenbuhl
- INSERM, INRAe, University of Rennes, Nutrition Metabolisms and Cancer Institute (NuMeCan), Platform AEM2, CHU Rennes, Rennes, France
| | - Suzanne Dufresne
- Laboratory 'Movement, Sport and Health Sciences'-EA7470, University of Rennes/ENS Rennes, Bruz, France
| | - Philippe Noirez
- IRMES-Institute for Research in Medicine and Epidemiology of Sport, INSEP, Paris, France.,INSERM S1124, Université de Paris, Paris, France.,EA7507, Performance Health Metrology Society, Université de Reims Champagne Ardenne, Reims, France
| | | | - Olivier Loréal
- INSERM, INRAe, University of Rennes, Nutrition Metabolisms and Cancer Institute (NuMeCan), Platform AEM2, CHU Rennes, Rennes, France
| | - Frédéric Derbré
- Laboratory 'Movement, Sport and Health Sciences'-EA7470, University of Rennes/ENS Rennes, Bruz, France
| |
Collapse
|
82
|
Wu X, Qin K, Iroegbu CD, Xiang K, Peng J, Guo J, Yang J, Fan C. Genetic analysis of potential biomarkers and therapeutic targets in ferroptosis from coronary artery disease. J Cell Mol Med 2022; 26:2177-2190. [PMID: 35152560 PMCID: PMC8995456 DOI: 10.1111/jcmm.17239] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/18/2022] [Accepted: 02/01/2022] [Indexed: 01/05/2023] Open
Abstract
Ferroptosis plays a key role in the death of cells including cardiomyocytes, and it is related to a variety of cardiac diseases. However, the role of ferroptosis-related genes (FRGs) in coronary artery disease (CAD) is not well characterized. We downloaded CAD-related information and FRGs from the gene expression omnibus (GEO) database and Ferroptosis Database (FerrDb) respectively. A total of 10 CAD-related DE-FRGs were obtained, which were closely linked to autophagy regulation and immune response. Subsequently, CA9, CBS, CEBPG, HSPB1, SLC1A4, STMN1 and TRIB3 among the 10 DE-FRGs were identified as marker genes by LASSO and SVM-RFE algorithms, which had tolerable diagnostic capabilities. Subsequent functional enrichment analysis showed that these marker genes may play a corresponding role in CAD by participating in the regulation of immune response, amino acid metabolism, cell cycle and multiple pathways related to the pathogenesis of CAD. Furthermore, a total of 58 drugs targeting 7 marker genes had been obtained. On the contrary, the ceRNA network revealed a complex regulatory relationship based on the marker genes. Also, CIBERSORT analysis showed that the changes in the immune microenvironment of CAD patients may be related to CBS, HSPB1 and CEBPG. We developed a diagnostic potency and provided an insight for exploring the mechanism for CAD. Before clinical application, further research is needed to test its diagnostic value for CAD.
Collapse
Affiliation(s)
- Xun Wu
- Department of the Cardiovascular SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Kele Qin
- Department of the Cardiovascular SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Chukwuemeka Daniel Iroegbu
- Department of the Cardiovascular SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Kun Xiang
- Department of the Cardiovascular SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Jun Peng
- Hunan Provincial Key Laboratory of Cardiovascular ResearchCentral South UniversityChangshaChina
| | - Jianjun Guo
- Hunan Fangsheng Pharmaceutical Co., Ltd.ChangshaChina
| | - Jinfu Yang
- Department of the Cardiovascular SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Chengming Fan
- Department of the Cardiovascular SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaChina
- Hunan Provincial Key Laboratory of Cardiovascular ResearchCentral South UniversityChangshaChina
- Hunan Fangsheng Pharmaceutical Co., Ltd.ChangshaChina
| |
Collapse
|
83
|
The (Bio)Chemistry of Non-Transferrin-Bound Iron. Molecules 2022; 27:molecules27061784. [PMID: 35335148 PMCID: PMC8951307 DOI: 10.3390/molecules27061784] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 12/14/2022] Open
Abstract
In healthy individuals, virtually all blood plasma iron is bound by transferrin. However, in several diseases and clinical conditions, hazardous non-transferrin-bound iron (NTBI) species occur. NTBI represents a potentially toxic iron form, being a direct cause of oxidative stress in the circulating compartment and tissue iron loading. The accumulation of these species can cause cellular damage in several organs, namely, the liver, spleen, and heart. Despite its pathophysiological relevance, the chemical nature of NTBI remains elusive. This has precluded its use as a clinical biochemical marker and the development of targeted therapies. Herein, we make a critical assessment of the current knowledge of NTBI speciation. The currently accepted hypotheses suggest that NTBI is mostly iron bound to citric acid and iron bound to serum albumin, but the chemistry of this system remains fuzzy. We explore the complex chemistry of iron complexation by citric acid and its implications towards NTBI reactivity. Further, the ability of albumin to bind iron is revised and the role of protein post-translational modifications on iron binding is discussed. The characterization of the NTBI species structure may be the starting point for the development of a standardized analytical assay, the better understanding of these species’ reactivity or the identification of NTBI uptake mechanisms by different cell types, and finally, to the development of new therapies.
Collapse
|
84
|
Carter PW, Dunham AJ. Modelling haemoglobin incremental loss on chronic red blood cell transfusions. Vox Sang 2022; 117:831-838. [PMID: 35238052 DOI: 10.1111/vox.13261] [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: 12/12/2021] [Revised: 01/20/2022] [Accepted: 02/09/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVES Understanding the impact of red blood cell (RBC) lifespan, initial RBC removal, and transfusion intervals on patient haemoglobin (Hb) levels and total iron exposure is not accessible for chronic transfusion scenarios. This article introduces the first model to help clinicians optimize chronic transfusion intervals to minimize transfusion frequency. MATERIALS AND METHODS Hb levels and iron exposure from multiple transfusions were calculated from Weibull residual lifespan distributions, the fraction effete RBC removed within 24-h (Xe ) and the nominal Hb increment. Two-unit transfusions of RBCs initiated at patient [Hb] = 7 g/dl were modelled for different RBC lifespans and transfusion intervals from 18 to 90 days, and Xe from 0.1 to 0.5. RESULTS Increased Xe requires shorter transfusion intervals to achieve steady-state [Hb] of 9 g/dl as follows: 30 days between transfusions at Xe = 0.5, 36 days at Xe = 0.4, 42 days at Xe = 0.3, 48 days at Xe = 0.2 and 54 days at Xe = 0.1. The same transfusion interval/Xe pairs result in a steady-state [Hb] = 8 g/dl when the RBC lifespan was halved. By reducing transfused RBC increment loss from 30% to 10%, annual transfusions were decreased by 22% with iron addition decreased by 24%. Acute dosing of iron occurs at the higher values of Xe on the day after a transfusion event. CONCLUSION Systematic trends in fractional Hb incremental loss Xe have been modelled and have a significant and calculatable impact on transfusion intervals and associated introduction of iron.
Collapse
|
85
|
Van Coillie S, Van San E, Goetschalckx I, Wiernicki B, Mukhopadhyay B, Tonnus W, Choi SM, Roelandt R, Dumitrascu C, Lamberts L, Dams G, Weyts W, Huysentruyt J, Hassannia B, Ingold I, Lele S, Meyer E, Berg M, Seurinck R, Saeys Y, Vermeulen A, van Nuijs ALN, Conrad M, Linkermann A, Rajapurkar M, Vandenabeele P, Hoste E, Augustyns K, Vanden Berghe T. Targeting ferroptosis protects against experimental (multi)organ dysfunction and death. Nat Commun 2022; 13:1046. [PMID: 35210435 PMCID: PMC8873468 DOI: 10.1038/s41467-022-28718-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 02/07/2022] [Indexed: 12/26/2022] Open
Abstract
The most common cause of death in the intensive care unit (ICU) is the development of multiorgan dysfunction syndrome (MODS). Besides life-supporting treatments, no cure exists, and its mechanisms are still poorly understood. Catalytic iron is associated with ICU mortality and is known to cause free radical-mediated cellular toxicity. It is thought to induce excessive lipid peroxidation, the main characteristic of an iron-dependent type of cell death conceptualized as ferroptosis. Here we show that the severity of multiorgan dysfunction and the probability of death are indeed associated with plasma catalytic iron and lipid peroxidation. Transgenic approaches underscore the role of ferroptosis in iron-induced multiorgan dysfunction. Blocking lipid peroxidation with our highly soluble ferrostatin-analogue protects mice from injury and death in experimental non-septic multiorgan dysfunction, but not in sepsis-induced multiorgan dysfunction. The limitations of the experimental mice models to mimic the complexity of clinical MODS warrant further preclinical testing. In conclusion, our data suggest ferroptosis targeting as possible treatment option for a stratifiable subset of MODS patients.
Collapse
Affiliation(s)
- Samya Van Coillie
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Emily Van San
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Ines Goetschalckx
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Bartosz Wiernicki
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Banibrata Mukhopadhyay
- Department of Nephrology, Muljibhai Patel Society for Research in Nephro-Urology, Nadiad, India
| | - Wulf Tonnus
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus, the Technische Universität Dresden, Dresden, Germany
| | - Sze Men Choi
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Ria Roelandt
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Catalina Dumitrascu
- Department of Pharmaceutical Sciences, Toxicological Centre, University of Antwerp, Antwerp, Belgium
| | - Ludwig Lamberts
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Geert Dams
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Wannes Weyts
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
| | - Jelle Huysentruyt
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Behrouz Hassannia
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Irina Ingold
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany.,Department of Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Suhas Lele
- Department of Nephrology, Muljibhai Patel Society for Research in Nephro-Urology, Nadiad, India
| | - Evelyne Meyer
- Department of Pharmacology, Toxicology and Biochemistry, Ghent University, Merelbeke, Belgium
| | - Maya Berg
- Department of Pharmaceutical Sciences, Toxicological Centre, University of Antwerp, Antwerp, Belgium
| | - Ruth Seurinck
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Yvan Saeys
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - An Vermeulen
- Department of Bioanalysis, Ghent University, Ghent, Belgium
| | - Alexander L N van Nuijs
- Department of Pharmaceutical Sciences, Toxicological Centre, University of Antwerp, Antwerp, Belgium
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany.,National Research Medical University, Laboratory of Experimental Oncology, Moscow, Russia
| | - Andreas Linkermann
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus, the Technische Universität Dresden, Dresden, Germany.,Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Mohan Rajapurkar
- Department of Nephrology, Muljibhai Patel Society for Research in Nephro-Urology, Nadiad, India
| | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.,Methusalem program, Ghent University, Ghent, Belgium
| | - Eric Hoste
- Intensive Care Unit, Ghent University Hospital; Ghent University, Ghent, Belgium
| | - Koen Augustyns
- Department of Pharmaceutical Sciences, Toxicological Centre, University of Antwerp, Antwerp, Belgium
| | - Tom Vanden Berghe
- VIB-UGent Center for Inflammation Research, Ghent, Belgium. .,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium. .,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
86
|
Borella E, Oosterholt S, Magni P, Pasqua OD. Characterisation of individual ferritin response in patients receiving chelation therapy. Br J Clin Pharmacol 2022; 88:3683-3694. [PMID: 35199367 PMCID: PMC9544664 DOI: 10.1111/bcp.15290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 12/21/2021] [Accepted: 01/07/2022] [Indexed: 11/26/2022] Open
Abstract
Aims To develop a drug–disease model describing iron overload and its effect on ferritin response in patients affected by transfusion‐dependent haemoglobinopathies and investigate the contribution of interindividual differences in demographic and clinical factors on chelation therapy with deferiprone or deferasirox. Methods Individual and mean serum ferritin data were retrieved from 13 published studies in patients affected by haemoglobinopathies receiving deferiprone or deferasirox. A nonlinear mixed effects modelling approach was used to characterise iron homeostasis and serum ferritin production taking into account annual blood consumption, baseline demographic and clinical characteristics. The effect of chelation therapy was parameterised as an increase in the iron elimination rate. Internal and external validation procedures were used to assess model performance across different study populations. Results An indirect response model was identified, including baseline ferritin concentrations and annual blood consumption as covariates. The effect of chelation on iron elimination rate was characterised by a linear function, with different slopes for each drug (0.0109 [90% CI: 0.0079–0.0131] vs. 0.0013 [90% CI: 0.0008–0.0018] L/mg mo). In addition to drug‐specific differences in the magnitude of the ferritin response, simulation scenarios indicate that ferritin elimination rates depend on ferritin concentrations at baseline. Conclusion Modelling of serum ferritin following chronic blood transfusion enabled the evaluation of drug‐induced changes in iron elimination rate and ferritin production. The use of a semi‐mechanistic parameterisation allowed us to disentangle disease‐specific factors from drug‐specific properties. Despite comparable chelation mechanisms, deferiprone appears to have a significantly larger effect on the iron elimination rate than deferasirox.
Collapse
Affiliation(s)
- Elisa Borella
- Department of Industrial Engineering and Informatics, Università degli Studi di Pavia, Pavia, Italy
| | - Sean Oosterholt
- Clinical Pharmacology & Therapeutics Group, University College London, London, United Kingdom
| | - Paolo Magni
- Department of Industrial Engineering and Informatics, Università degli Studi di Pavia, Pavia, Italy
| | - Oscar Della Pasqua
- Clinical Pharmacology & Therapeutics Group, University College London, London, United Kingdom.,Clinical Pharmacology Modelling & Simulation, GlaxoSmithKline, Brentford, United Kingdom
| |
Collapse
|
87
|
Raha AA, Biswas A, Henderson J, Chakraborty S, Holland A, Friedland RP, Mukaetova-Ladinska E, Zaman S, Raha-Chowdhury R. Interplay of Ferritin Accumulation and Ferroportin Loss in Ageing Brain: Implication for Protein Aggregation in Down Syndrome Dementia, Alzheimer’s, and Parkinson’s Diseases. Int J Mol Sci 2022; 23:ijms23031060. [PMID: 35162984 PMCID: PMC8834792 DOI: 10.3390/ijms23031060] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/05/2022] [Accepted: 01/14/2022] [Indexed: 12/24/2022] Open
Abstract
Iron accumulates in the ageing brain and in brains with neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and Down syndrome (DS) dementia. However, the mechanisms of iron deposition and regional selectivity in the brain are ill-understood. The identification of several proteins that are involved in iron homeostasis, transport, and regulation suggests avenues to explore their function in neurodegenerative diseases. To uncover the molecular mechanisms underlying this association, we investigated the distribution and expression of these key iron proteins in brain tissues of patients with AD, DS, PD, and compared them with age-matched controls. Ferritin is an iron storage protein that is deposited in senile plaques in the AD and DS brain, as well as in neuromelanin-containing neurons in the Lewy bodies in PD brain. The transporter of ferrous iron, Divalent metal protein 1 (DMT1), was observed solely in the capillary endothelium and in astrocytes close to the ventricles with unchanged expression in PD. The principal iron transporter, ferroportin, is strikingly reduced in the AD brain compared to age-matched controls. Extensive blood vessel damage in the basal ganglia and deposition of punctate ferritin heavy chain (FTH) and hepcidin were found in the caudate and putamen within striosomes/matrix in both PD and DS brains. We suggest that downregulation of ferroportin could be a key reason for iron mismanagement through disruption of cellular entry and exit pathways of the endothelium. Membrane damage and subsequent impairment of ferroportin and hepcidin causes oxidative stress that contributes to neurodegeneration seen in DS, AD, and in PD subjects. We further propose that a lack of ferritin contributes to neurodegeneration as a consequence of failure to export toxic metals from the cortex in AD/DS and from the substantia nigra and caudate/putamen in PD brain.
Collapse
Affiliation(s)
- Animesh Alexander Raha
- John van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge CB2 2PY, UK; (A.A.R.); (J.H.); (S.C.)
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Anwesha Biswas
- Department of Biochemistry, The M. S. University of Baroda, Vadodara 39002, India;
| | - James Henderson
- John van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge CB2 2PY, UK; (A.A.R.); (J.H.); (S.C.)
| | - Subhojit Chakraborty
- John van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge CB2 2PY, UK; (A.A.R.); (J.H.); (S.C.)
- NIHR Biomedical Research Centre, Moorfields Eye Hospital and UCL Institute of Ophthalmology, London EC1V 9EL, UK
| | - Anthony Holland
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8AH, UK; (A.H.); (S.Z.)
| | - Robert P. Friedland
- Department of Neurology, School of Medicine University of Louisville, Louisville, KY 40292, USA;
| | | | - Shahid Zaman
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8AH, UK; (A.H.); (S.Z.)
| | - Ruma Raha-Chowdhury
- John van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge CB2 2PY, UK; (A.A.R.); (J.H.); (S.C.)
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge CB2 8AH, UK; (A.H.); (S.Z.)
- Correspondence: ; Tel.: +44-1223-465262; Fax: +44-1223-746033
| |
Collapse
|
88
|
Shang X, Zhang R, Wang X, Yao J, Zhao X, Li H. The Relationship of Hyperferritinemia to Metabolism and Chronic Complications in Type 2 Diabetes. Diabetes Metab Syndr Obes 2022; 15:175-182. [PMID: 35068935 PMCID: PMC8769058 DOI: 10.2147/dmso.s348232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 12/20/2021] [Indexed: 11/30/2022] Open
Abstract
AIM Elevated serum ferritin has been found to be closely related to type 2 diabetes mellitus. This study aimed to explore the relationship of high serum ferritin to metabolism and chronic complications in type 2 diabetes. METHODS This was a cross-sectional study. A total of 330 type 2 diabetes patients who visited an endocrine clinic were included for the analysis. Serum ferritin and metabolic parameters were recorded. The prevalence of chronic diabetic complications was evaluated. Based on serum ferritin, participants were divided into hyperferritinemia and normal-ferritin groups. Metabolic parameters and prevalence of chronic diabetic complications were compared. The relationship between hyperferritinemia and chronic diabetic complications was explored with multivariate logistic regression models. Data were statistically analyzed by sex. RESULTS Compared with the normal-ferritin group, the hyperferritinemia group showed higher levels of the serum inflammatory marker CRP and higher prevalence of diabetic retinopathy (DR) and coronary heart disease (CHD), regardless of sex (p<0.05). Moreover, male patients with hyperferritinemia had increased serum triglyceride, alanine transferase, γ-glutamyltranspeptidase, urea nitrogen, creatinine, and uric acid and higher prevalence of microalbuminuria (p<0.01). After controlling for demographics and metabolic profiles, hyperferritinemia remained an independent risk factor of DR (male OR 3.957, 95% CI 1.559-10.041, p=0.004; female OR 2.474, 95% CI 1.127-5.430, p=0.024) and CHD (male OR 2.607, 95% CI 1.087-6.257, p=0.032; female OR 2.293, 95% CI 1.031-5.096, p=0.042). CONCLUSION This study found that hyperferritinemia was associated with increased CRP and higher prevalence of DR and CHD in type 2 diabetes. In men, high serum ferritin was also associated with dyslipidemia, hepatic dysfunction, and microalbuminuria.
Collapse
Affiliation(s)
- Xiaojing Shang
- Department of Endocrinology, Tianjin Fourth Central Hospital, Tianjin, People’s Republic of China
| | - Rui Zhang
- Division of Health Management, Tianjin Fourth Central Hospital, Tianjin, People’s Republic of China
| | - Xiaolai Wang
- Department of Endocrinology, Tianjin Fourth Central Hospital, Tianjin, People’s Republic of China
| | - Junxin Yao
- Department of Endocrinology, Tianjin Fourth Central Hospital, Tianjin, People’s Republic of China
| | - Xiaoying Zhao
- Department of Endocrinology, Tianjin Fourth Central Hospital, Tianjin, People’s Republic of China
| | - Huanming Li
- Department of Cardiology, Tianjin Fourth Central Hospital, Tianjin, People’s Republic of China
- Correspondence: Huanming Li Email
| |
Collapse
|
89
|
Jayakumar D, S Narasimhan KK, Periandavan K. Triad role of hepcidin, ferroportin, and Nrf2 in cardiac iron metabolism: From health to disease. J Trace Elem Med Biol 2022; 69:126882. [PMID: 34710708 DOI: 10.1016/j.jtemb.2021.126882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/29/2021] [Accepted: 10/19/2021] [Indexed: 11/28/2022]
Abstract
Iron is an essential trace element required for several vital physiological and developmental processes, including erythropoiesis, bone, and neuronal development. Iron metabolism and oxygen homeostasis are interlinked to perform a vital role in the functionality of the heart. The metabolic machinery of the heart utilizes almost 90 % of oxygen through the electron transport chain. To handle this tremendous level of oxygen, the iron metabolism in the heart is utmost crucial. Iron availability to the heart is therefore tightly regulated by (i) the hepcidin/ferroportin axis, which controls dietary iron absorption, storage, and recycling, and (ii) iron regulatory proteins 1 and 2 (IRP1/2) via hypoxia inducible factor 1 (HIF1) pathway. Despite iron being vital to the heart, recent investigations have demonstrated that iron imbalance is a common manifestation in conditions of heart failure (HF), since free iron readily transforms between Fe2+ and Fe3+via the Fenton reaction, leading to reactive oxygen species (ROS) production and oxidative damage. Therefore, to combat iron-mediated oxidative stress, targeting Nrf2/ARE antioxidant signaling is rational. The involvement of Nrf2 in regulating several genes engaged in heme synthesis, iron storage, and iron export is beginning to be uncovered. Consequently, it is possible that Nrf2/hepcidin/ferroportin might act as an epicenter connecting iron metabolism to redox alterations. However, the mechanism bridging the two remains obscure. In this review, we tried to summarize the contemporary insight of how cardiomyocytes regulate intracellular iron levels and discussed the mechanisms linking cardiac dysfunction with iron imbalance. Further, we emphasized the impact of Nrf2 on the interplay between systemic/cardiac iron control in the context of heart disease, particularly in myocardial ischemia and HF.
Collapse
Affiliation(s)
- Deepthy Jayakumar
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute for Basic Medical Sciences, University of Madras, Chennai, 600113, Tamil Nadu, India
| | - Kishore Kumar S Narasimhan
- Department of Pharmacology and Neurosciences, Creighton University, 2500 California Plaza, Omaha, NE, USA
| | - Kalaiselvi Periandavan
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute for Basic Medical Sciences, University of Madras, Chennai, 600113, Tamil Nadu, India.
| |
Collapse
|
90
|
Cabantchik IZ, Hershko C. Plasma nontransferrin bound iron-nontransferrin bound iron revisited: Implications for systemic iron overload and in iv iron supplementation. Am J Hematol 2022; 97:7-9. [PMID: 34637551 DOI: 10.1002/ajh.26374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/24/2022]
Affiliation(s)
- Ioav Z Cabantchik
- Institute of Life Sciences, Faculty of Natural Sciences, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Chaim Hershko
- Institute of Medicine, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
91
|
Rombout-Sestrienkova E, Brandts L, Koek GH, van Deursen CTBM. Patients with hereditary hemochromatosis reach safe range of transferrin saturation sooner with erythrocytaphereses than with phlebotomies. J Clin Apher 2021; 37:100-105. [PMID: 34897777 PMCID: PMC9299622 DOI: 10.1002/jca.21956] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 01/01/2023]
Abstract
Introduction For the maintenance treatment of patients with hereditary hemochromatosis (HH), it is advised to keep the transferrin saturation (TSAT) <70% to prevent formation of non‐transferrin‐bound iron and labile plasma iron. The period of the initial iron depletion may last up to 1 year or longer and during this period, the patient is exposed to elevated TSAT levels. Therapeutic erythrocytapheresis (TE) is a modality which has proven to reduce treatment duration of patients with iron overload from HH. In this study, we investigated the time to reach TSAT <70% for both treatment modalities. Methods From a previous randomized controlled trial comparing erythrocytaphereses with phlebotomies (PBMs), we performed an analysis in a subgroup of patients who presented with TSAT >70%. Mann‐Whitney U tests were performed to compare the number of treatments and the number of weeks to reach the interim goal of a persistent level of <70% for TSAT between TE and PBM. Results The period to reach TSAT levels of <70% was statistically significant shorter for the TE group compared to the PBM treatment group (median treatment procedures [IQR] 2.0 (5) vs 16.0 (23), P‐value: <.001, and median treatment duration [IQR]: 5.5 (11) vs 19.0 (29) weeks, P‐value: .007). Conclusion Patients with HH reach a safe TSAT <70% significantly sooner and with less treatment procedures with TE compared to PBM.
Collapse
Affiliation(s)
- Eva Rombout-Sestrienkova
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Department of Transfusion Medicine, Sanquin Blood Supply, Maastricht, The Netherlands
| | - Lloyd Brandts
- Clinical Epidemiology and Medical Technology Assessment (KEMTA), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ger H Koek
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre+, Maastricht, The Netherlands.,School of Nutrition and Translational Research in Metabolism (NUTRIM), University Maastricht, Maastricht, The Netherlands
| | - Cees Th B M van Deursen
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Department of Internal Medicine Gastroenterology and Clinical Geriatrics, Zuyderland Medical Center, Heerlen, The Netherlands
| |
Collapse
|
92
|
Yu S, Jia J, Zheng J, Zhou Y, Jia D, Wang J. Recent Progress of Ferroptosis in Lung Diseases. Front Cell Dev Biol 2021; 9:789517. [PMID: 34869391 PMCID: PMC8635032 DOI: 10.3389/fcell.2021.789517] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/31/2021] [Indexed: 01/08/2023] Open
Abstract
Ferroptosis is a new form of programmed cell death due to iron-dependent excess accumulation of lipid peroxides and differs from other programmed cell deaths in morphological and biochemical characteristics. The process of ferroptosis is precisely regulated by iron metabolism, lipid metabolism, amino acid metabolism, and numerous signaling pathways, and plays a complex role in many pathophysiological processes. Recent studies have found that ferroptosis is closely associated with the development and progression of many lung diseases, including acute lung injury, pulmonary ischemia-reperfusion injury, lung cancer, chronic obstructive pulmonary disease, and pulmonary fibrosis. Here, we present a review of the main regulatory mechanisms of ferroptosis and its research progress in the pathogenesis and treatment of lung diseases, with the aim of providing new ideas for basic and clinical research of lung-related diseases.
Collapse
Affiliation(s)
- Shangjiang Yu
- Department of Clinical Medicine, Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinqiu Jia
- Department of Pediatrics, Taizhou Women and Children's Hospital of Wenzhou Medical University, Taizhou, China
| | - Jinyu Zheng
- Department of Clinical Medicine, Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiyang Zhou
- Department of Clinical Medicine, Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Danyun Jia
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Junlu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
93
|
The redox mechanism of ferrocene and its phytochemical and biochemical compounds in anticancer therapy: A mini review. INORG CHEM COMMUN 2021. [DOI: 10.1016/j.inoche.2021.109044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
94
|
Zeidan RS, Han SM, Leeuwenburgh C, Xiao R. Iron homeostasis and organismal aging. Ageing Res Rev 2021; 72:101510. [PMID: 34767974 DOI: 10.1016/j.arr.2021.101510] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/21/2022]
Abstract
Iron is indispensable for normal body functions across species because of its critical roles in red blood cell function and many essential proteins and enzymes required for numerous physiological processes. Regulation of iron homeostasis is an intricate process involving multiple modulators at the systemic, cellular, and molecular levels. Interestingly, emerging evidence has demonstrated that many modulators of iron homeostasis contribute to organismal aging and longevity. On the other hand, the age-related dysregulation of iron homeostasis is often associated with multiple age-related pathologies including bone resorption and neurodegenerative diseases such as Alzheimer's disease. Thus, a thorough understanding on the interconnections between systemic and cellular iron balance and organismal aging may help decipher the etiologies of multiple age-related diseases, which could ultimately lead to developing therapeutic strategies to delay aging and treat various age-related diseases. Here we present the current understanding on the mechanisms of iron homeostasis. We also discuss the impacts of aging on iron homeostatic processes and how dysregulated iron metabolism may affect aging and organismal longevity.
Collapse
|
95
|
Sakajiri T, Nakatsuji M, Teraoka Y, Furuta K, Ikuta K, Shibusa K, Sugano E, Tomita H, Inui T, Yamamura T. Zinc mediates the interaction between ceruloplasmin and apo-transferrin for the efficient transfer of Fe(III) ions. Metallomics 2021; 13:6427378. [PMID: 34791391 DOI: 10.1093/mtomcs/mfab065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 11/02/2021] [Indexed: 11/14/2022]
Abstract
Fe(II) exported from cells is oxidized to Fe(III), possibly by a multi-copper ferroxidase (MCF) such as ceruloplasmin (CP), to efficiently bind with the plasma iron transport protein transferrin (TF). As unbound Fe(III) is highly insoluble and reactive, its release into the blood during the transfer from MCF to TF must be prevented. A likely mechanism for preventing the release of unbound Fe(III) is via direct interaction between MCF and TF; however, the occurrence of this phenomenon remains controversial. This study aimed to reveal the interaction between these proteins, possibly mediated by zinc. Using spectrophotometric, isothermal titration calorimetric, and surface plasmon resonance methods, we found that Zn(II)-bound CP bound to iron-free TF (apo-TF) with a Kd of 4.2 μM and a stoichiometry CP:TF of ∼2:1. Computational modeling of the complex between CP and apo-TF predicted that each of the three Zn(II) ions that bind to CP further binds to acidic amino acid residues of apo-TF to play a role as a cross-linker connecting both proteins. Domain 4 of one CP molecule and domain 6 of the other CP molecule fit tightly into the clefts in the N- and C-lobes of apo-TF, respectively. Upon the binding of two Fe(III) ions to apo-TF, the resulting diferric TF [Fe(III)2TF] dissociated from CP by conformational changes in TF. In human blood plasma, zinc deficiency reduced the production of Fe(III)2TF and concomitantly increased the production of non-TF-bound iron. Our findings suggest that zinc may be involved in the transfer of iron between CP and TF.
Collapse
Affiliation(s)
- Tetsuya Sakajiri
- Laboratory of Biological Macromolecules, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan.,Faculty of Nutritional Sciences, the University of Morioka, 808 Sunakomi, Takizawa, Iwate 020-0694, Japan.,Qualtec Co. Ltd., 4-230 Sambo-cho, Sakai, Osaka 590-0906, Japan.,Department of Nutrition, Kyushu Nutrition Welfare University, 5-1-1 Shimoitozu, Kitakyushu Kokurakita-ku, Fukuoka 803-0846, Japan
| | - Masatoshi Nakatsuji
- Laboratory of Biological Macromolecules, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Yoshiaki Teraoka
- Laboratory of Biological Macromolecules, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Kosuke Furuta
- Laboratory of Biological Macromolecules, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Katsuya Ikuta
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan.,Japanese Red Cross Hokkaido Blood Center, 2-1 Nijuyonken, Nishi-ku, Sapporo, Hokkaido 063-0802, Japan
| | - Kotoe Shibusa
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan.,Hokkaido System Science Co., Ltd., 2-1 Shinkawa Nishi, Kita-ku, Sapporo, Hokkaido 001-0932, Japan
| | - Eriko Sugano
- Department of Chemistry and Biological Sciences, Faculty of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| | - Hiroshi Tomita
- Department of Chemistry and Biological Sciences, Faculty of Science and Engineering, Iwate University, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| | - Takashi Inui
- Laboratory of Biological Macromolecules, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Takaki Yamamura
- Laboratory of Biological Macromolecules, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan.,Faculty of Nutritional Sciences, the University of Morioka, 808 Sunakomi, Takizawa, Iwate 020-0694, Japan
| |
Collapse
|
96
|
Ali S, Mumtaz S, Shakir HA, Khan M, Tahir HM, Mumtaz S, Mughal TA, Hassan A, Kazmi SAR, Sadia, Irfan M, Khan MA. Current status of beta-thalassemia and its treatment strategies. Mol Genet Genomic Med 2021; 9:e1788. [PMID: 34738740 PMCID: PMC8683628 DOI: 10.1002/mgg3.1788] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/15/2020] [Accepted: 08/13/2021] [Indexed: 01/05/2023] Open
Abstract
Background Thalassemia is an inherited hematological disorder categorized by a decrease or absence of one or more of the globin chains synthesis. Beta‐thalassemia is caused by one or more mutations in the beta‐globin gene. The absence or reduced amount of beta‐globin chains causes ineffective erythropoiesis which leads to anemia. Methods Beta‐thalassemia has been further divided into three main forms: thalassemia major, intermedia, and minor/silent carrier. A more severe form among these is thalassemia major in which individuals depend upon blood transfusion for survival. The high level of iron deposition occurs due to regular blood transfusion therapy. Results Overloaded iron raises the synthesis of reactive oxygen species (ROS) that are noxious and prompting the injury to the hepatic, endocrine, and vascular system. Thalassemia can be analyzed and diagnosed via prenatal testing (genetic testing of amniotic fluid), blood smear, complete blood count, and DNA analysis (genetic testing). Treatment of thalassemia intermediate is symptomatic; however; it can also be accomplished by folic supplementation and splenectomy. Conclusion Thalassemia major can be cured through regular transfusion of blood, transplantation of bone marrow, iron chelation management, hematopoietic stem cell transplantation, stimulation of fetal hemoglobin production, and gene therapy.
Collapse
Affiliation(s)
- Shaukat Ali
- Applied Entomology and Medical Toxicology, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shumaila Mumtaz
- Applied Entomology and Medical Toxicology, Department of Zoology, Government College University, Lahore, Pakistan
| | | | - Muhammad Khan
- Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Hafiz Muhammad Tahir
- Applied Entomology and Medical Toxicology, Department of Zoology, Government College University, Lahore, Pakistan
| | - Samaira Mumtaz
- Applied Entomology and Medical Toxicology, Department of Zoology, Government College University, Lahore, Pakistan
| | - Tafail Akbar Mughal
- Applied Entomology and Medical Toxicology, Department of Zoology, Government College University, Lahore, Pakistan
| | - Ali Hassan
- Applied Entomology and Medical Toxicology, Department of Zoology, Government College University, Lahore, Pakistan
| | | | - Sadia
- Department of Biotechnology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Muhammad Irfan
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| | - Muhammad Adeeb Khan
- Department of Zoology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| |
Collapse
|
97
|
Albalat E, Cavey T, Leroyer P, Ropert M, Balter V, Loréal O. H fe Gene Knock-Out in a Mouse Model of Hereditary Hemochromatosis Affects Bodily Iron Isotope Compositions. Front Med (Lausanne) 2021; 8:711822. [PMID: 34722560 PMCID: PMC8554230 DOI: 10.3389/fmed.2021.711822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
Hereditary hemochromatosis is a genetic iron overload disease related to a mutation within the HFE gene that controls the expression of hepcidin, the master regulator of systemic iron metabolism. The natural stable iron isotope composition in whole blood of control subjects is different from that of hemochromatosis patients and is sensitive to the amount of total iron removed by the phlebotomy treatment. The use of stable isotopes to unravel the pathological mechanisms of iron overload diseases is promising but hampered by the lack of data in organs involved in the iron metabolism. Here, we use Hfe -/- mice, a model of hereditary hemochromatosis, to study the impact of the knock-out on iron isotope compositions of erythrocytes, spleen and liver. Iron concentration increases in liver and red blood cells of Hfe -/- mice compared to controls. The iron stable isotope composition also increases in liver and erythrocytes, consistent with a preferential accumulation of iron heavy isotopes in Hfe -/- mice. In contrast, no difference in the iron concentration nor isotope composition is observed in spleen of Hfe -/- and control mice. Our results in mice suggest that the observed increase of whole blood isotope composition in hemochromatosis human patients does not originate from, but is aggravated by, bloodletting. The subsequent rapid increase of whole blood iron isotope composition of treated hemochromatosis patients is rather due to the release of hepatic heavy isotope-enriched iron than augmented iron dietary absorption. Further research is required to uncover the iron light isotope component that needs to balance the accumulation of hepatic iron heavy isotope, and to better understand the iron isotope fractionation associated to metabolism dysregulation during hereditary hemochromatosis.
Collapse
Affiliation(s)
- Emmanuelle Albalat
- CNRS UMR 5276, LGL-TPE, ENS de Lyon, Université de Lyon, Villeurbanne, France
| | - Thibault Cavey
- INSERM, Univ Rennes, INRAe, UMR 1241, Plateforme AEM2, CHU Pontchaillou, Institut Nutrition Metabolisms et Cancer (NuMeCan), Rennes, France
| | - Patricia Leroyer
- INSERM, Univ Rennes, INRAe, UMR 1241, Plateforme AEM2, CHU Pontchaillou, Institut Nutrition Metabolisms et Cancer (NuMeCan), Rennes, France
| | - Martine Ropert
- INSERM, Univ Rennes, INRAe, UMR 1241, Plateforme AEM2, CHU Pontchaillou, Institut Nutrition Metabolisms et Cancer (NuMeCan), Rennes, France
| | - Vincent Balter
- CNRS UMR 5276, LGL-TPE, ENS de Lyon, Université de Lyon, Villeurbanne, France
| | - Olivier Loréal
- INSERM, Univ Rennes, INRAe, UMR 1241, Plateforme AEM2, CHU Pontchaillou, Institut Nutrition Metabolisms et Cancer (NuMeCan), Rennes, France
| |
Collapse
|
98
|
Dong H, Xia Y, Jin S, Xue C, Wang Y, Hu R, Jiang H. Nrf2 attenuates ferroptosis-mediated IIR-ALI by modulating TERT and SLC7A11. Cell Death Dis 2021; 12:1027. [PMID: 34716298 PMCID: PMC8556385 DOI: 10.1038/s41419-021-04307-1] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/18/2021] [Accepted: 10/13/2021] [Indexed: 12/18/2022]
Abstract
Acute lung injury (ALI) carries a mortality rate of ~50% and is a hot topic in the world of critical illness research. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a critical modulator of intracellular oxidative homeostasis and serves as an antioxidant. The Nrf2-related anti-oxidative stress is strongly associated with ferroptosis suppression. Meanwhile, telomerase reverse transcriptase (TERT), the catalytic portion of the telomerase protein, is reported to travel to the mitochondria to alleviate ROS. In our study, we found that TERT was significantly reduced in lung tissue of Nrf2-/- mice in the model of intestinal ischemia/reperfusion-induced acute lung injury (IIR-ALI). In addition, MDA levels showed marked increase, whereas GSH and GPX4 levels fell drastically in ALI models. Moreover, typical-related structural changes were observed in the type II alveolar epithelial cells in the IIR model. We further employed the scanning transmission X-ray microscopy (STXM) to examine Fe levels and distribution within cells. Based on our observations, massive aggregates of Fe were found in the MLE-12 cells upon OGD/R (oxygen and glucose deprivation/reperfusion) induction. Additionally, Nrf2 silencing dramatically reduced TERT and SLC7A11 levels, and further exacerbated cellular injuries. In contrast, TERT-overexpressing cells exhibited marked elevation in SLC7A11 levels and thereby inhibited ferroptosis. Collectively, these data suggest that Nrf2 can negatively regulate ferroptosis via modulation of TERT and SLC7A11 levels. The conclusion from this study brings insight into new candidates that can be targeted in future IIR-ALI therapy.
Collapse
Affiliation(s)
- Hui Dong
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, China
| | - Yangyang Xia
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, China
| | - Shanliang Jin
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, China
| | - Chaofan Xue
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Yanjun Wang
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, China
| | - Rong Hu
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, China.
| | - Hong Jiang
- Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai, China.
| |
Collapse
|
99
|
Palumbo GA, Galimberti S, Barcellini W, Cilloni D, Di Renzo N, Elli EM, Finelli C, Maurillo L, Ricco A, Musto P, Russo R, Latagliata R. From Biology to Clinical Practice: Iron Chelation Therapy With Deferasirox. Front Oncol 2021; 11:752192. [PMID: 34692534 PMCID: PMC8527180 DOI: 10.3389/fonc.2021.752192] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/08/2021] [Indexed: 01/19/2023] Open
Abstract
Iron chelation therapy (ICT) has become a mainstay in heavily transfused hematological patients, with the aim to reduce iron overload (IOL) and prevent organ damage. This therapeutic approach is already widely used in thalassemic patients and in low-risk Myelodysplastic Syndrome (MDS) patients. More recently, ICT has been proposed for high-risk MDS, especially when an allogeneic bone marrow transplantation has been planned. Furthermore, other hematological and hereditary disorders, characterized by considerable transfusion support to manage anemia, could benefit from this therapy. Meanwhile, data accumulated on how iron toxicity could exacerbate anemia and other clinical comorbidities due to oxidative stress radical oxygen species (ROS) mediated by free iron species. Taking all into consideration, together with the availability of approved oral iron chelators, we envision a larger use of ICT in the near future. The aim of this review is to better identify those non-thalassemic patients who can benefit from ICT and give practical tips for management of this therapeutic strategy.
Collapse
Affiliation(s)
- Giuseppe A. Palumbo
- Department of Scienze Mediche Chirurgiche e Tecnologie Avanzate “G.F. Ingrassia, ” University of Catania, Catania, Italy
| | - Sara Galimberti
- Section of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Wilma Barcellini
- Hematology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico di Milano and University of Milan, Milan, Italy
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Nicola Di Renzo
- Hematology and Transplant Unit, Ospedale Vito Fazzi, Lecce, Italy
| | - Elena Maria Elli
- Division of Hematology and Bone Marrow Unit, Ospedale San Gerardo, Aziende Socio Sanitarie Territoriali (ASST), Monza, Italy
| | - Carlo Finelli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, Bologna, Italy
| | - Luca Maurillo
- Department of Onco-hematology, Fondazione Policlinico Tor Vergata, Rome, Italy
| | - Alessandra Ricco
- Unit of Hematology and Stem Cell Transplantation, Azienda Ospedaliera Universitaria (AOU) Consorziale Policlinico, Bari, Italy
| | - Pellegrino Musto
- Unit of Hematology and Stem Cell Transplantation, Azienda Ospedaliera Universitaria (AOU) Consorziale Policlinico, Bari, Italy
- Department of Emergency and Organ Transplantation, “Aldo Moro” University School of Medicine, Bari, Italy
| | - Rodolfo Russo
- Clinica Nefrologica, Dialisi e Trapianto, Department of Integrated Medicine with the Territory, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Roberto Latagliata
- Unità Operativa Complessa (UOC) Ematologia, Ospedale Belcolle, Viterbo and Division of Cellular Biotechnology and Hematology, Sapienza University, Rome, Italy
| |
Collapse
|
100
|
Karoor V, Swindle D, Pak DI, Strassheim D, Fini MA, Dempsey E, Stenmark KR, Hassell K, Nuss R, Buehler PW, Irwin DC. Evidence supporting a role for circulating macrophages in the regression of vascular remodeling following sub-chronic exposure to hemoglobin plus hypoxia. Pulm Circ 2021; 11:20458940211056806. [PMID: 34777787 PMCID: PMC8573496 DOI: 10.1177/20458940211056806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/12/2021] [Indexed: 11/15/2022] Open
Abstract
Macrophages are a heterogeneous population with both pro- and anti-inflammatory functions play an essential role in maintaining tissue homeostasis, promoting inflammation under pathological conditions, and tissue repair after injury. In pulmonary hypertension, the M1 phenotype is more pro-inflammatory compared to the M2 phenotype, which is involved in tissue repair. The role of macrophages in the initiation and progression of pulmonary hypertension is well studied. However, their role in the regression of established pulmonary hypertension is not well known. Rats chronically exposed to hemoglobin (Hb) plus hypoxia (HX) share similarities to humans with pulmonary hypertension associated with hemolytic disease, including the presence of a unique macrophage phenotype surrounding distal vessels that are associated with vascular remodeling. These lung macrophages are characterized by high iron content, HO-1, ET-1, and IL-6, and are recruited from the circulation. Depletion of macrophages in this model prevents the development of pulmonary hypertension and vascular remodeling. In this study, we specifically investigate the regression of pulmonary hypertension over a four-week duration after rats were removed from Hb + HX exposure with and without gadolinium chloride administration. Withdrawal of Hb + HX reversed systolic pressures and right ventricular function after Hb + Hx exposure in four weeks. Our data show that depleting circulating monocytes/macrophages during reversal prevents complete recovery of right ventricular systolic pressure and vascular remodeling in this rat model of pulmonary hypertension at four weeks post exposure. The data presented offer a novel insight into the role of macrophages in the processes of pulmonary hypertension regression in a rodent model of Hb + Hx-driven disease.
Collapse
Affiliation(s)
- Vijaya Karoor
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Delaney Swindle
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - David I Pak
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Derek Strassheim
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Mehdi A Fini
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Edward Dempsey
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Kurt R Stenmark
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Kathryn Hassell
- Division of Hematology Colorado Sickle Cell Treatment and Research Center, School of Medicine, Anschutz Medical Campus, University of Colorado-Denver School of Medicine, Aurora, CO, USA
| | - Rachelle Nuss
- Division of Hematology Colorado Sickle Cell Treatment and Research Center, School of Medicine, Anschutz Medical Campus, University of Colorado-Denver School of Medicine, Aurora, CO, USA
| | - Paul W. Buehler
- Department of Pathology, University of Maryland, Baltimore, MD, USA
- The Center for Blood Oxygen Transport, Department of Pediatrics, School of Medicine, Baltimore, MD, USA
| | - David C. Irwin
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|