1
|
Fahoum L, Moshe-Belisowski S, Zaydel K, Ghatpande N, Guttmann-Raviv N, Zhang W, Li K, Tong WH, Nyska A, Waterman M, Weisshof R, Zuckerman A, Meyron-Holtz EG. Iron regulatory protein 1 is required for the propagation of inflammation in inflammatory bowel disease. J Biol Chem 2024; 300:107639. [PMID: 39122013 PMCID: PMC11408829 DOI: 10.1016/j.jbc.2024.107639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 07/02/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
Inflammatory bowel diseases (IBDs) are complex disorders. Iron accumulates in the inflamed tissue of IBD patients, yet neither a mechanism for the accumulation nor its implication on the course of inflammation is known. We hypothesized that the inflammation modifies iron homeostasis, affects tissue iron distribution, and that this in turn perpetuates the inflammation. This study analyzed human biopsies, animal models, and cellular systems to decipher the role of iron homeostasis in IBD. We found inflammation-mediated modifications of iron distribution, and iron-decoupled activation of the iron regulatory protein (IRP) 1. To understand the role of IRP1 in the course of this inflammation-associated iron pattern, a novel cellular coculture model was established, which replicated the iron-pattern observed in vivo, and supported involvement of nitric oxide in the activation of IRP1 and the typical iron pattern in inflammation. Importantly, deletion of IRP1 from an IBD mouse model completely abolished both, the misdistribution of iron and intestinal inflammation. These findings suggest that IRP1 plays a central role in the coordination of the inflammatory response in the intestinal mucosa and that it is a viable candidate for therapeutic intervention in IBD.
Collapse
Affiliation(s)
- Lulu Fahoum
- Laboratory of Molecular Nutrition, Department of Biotechnology and Food Engineering, Technion- Israel Institute of Technology, Haifa, Israel
| | - Shirly Moshe-Belisowski
- Laboratory of Molecular Nutrition, Department of Biotechnology and Food Engineering, Technion- Israel Institute of Technology, Haifa, Israel
| | - Kristina Zaydel
- Laboratory of Molecular Nutrition, Department of Biotechnology and Food Engineering, Technion- Israel Institute of Technology, Haifa, Israel
| | - Niraj Ghatpande
- Laboratory of Molecular Nutrition, Department of Biotechnology and Food Engineering, Technion- Israel Institute of Technology, Haifa, Israel
| | - Noga Guttmann-Raviv
- Laboratory of Molecular Nutrition, Department of Biotechnology and Food Engineering, Technion- Israel Institute of Technology, Haifa, Israel
| | - Wenxin Zhang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Kuanyu Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Wing-Hang Tong
- Molecular Medicine Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | - Abraham Nyska
- Department of Biotechnology and Food Engineering, Tel Aviv University and Consultant in Toxicologic Pathology, Tel Aviv, Israel
| | - Matti Waterman
- Department of Biotechnology and Food Engineering, Rambam/Technion- Israel Institute of Technology, Haifa, Israel
| | - Ronni Weisshof
- Department of Biotechnology and Food Engineering, Rambam/Technion- Israel Institute of Technology, Haifa, Israel
| | - Avi Zuckerman
- Department of Biotechnology and Food Engineering, Aviv Projects, Ness Ziona, Israel
| | - Esther G Meyron-Holtz
- Laboratory of Molecular Nutrition, Department of Biotechnology and Food Engineering, Technion- Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
2
|
Lakhal-Littleton S, Cleland JGF. Iron deficiency and supplementation in heart failure. Nat Rev Cardiol 2024; 21:463-486. [PMID: 38326440 DOI: 10.1038/s41569-024-00988-1] [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] [Accepted: 01/08/2024] [Indexed: 02/09/2024]
Abstract
Non-anaemic iron deficiency (NAID) is a strategic target in cardiovascular medicine because of its association with a range of adverse effects in various conditions. Endeavours to tackle NAID in heart failure have yielded mixed results, exposing knowledge gaps in how best to define 'iron deficiency' and the handling of iron therapies by the body. To address these gaps, we harness the latest understanding of the mechanisms of iron homeostasis outside the erythron and integrate clinical and preclinical lines of evidence. The emerging picture is that current definitions of iron deficiency do not assimilate the multiple influences at play in patients with heart failure and, consequently, fail to identify those with a truly unmet need for iron. Additionally, current iron supplementation therapies benefit only certain patients with heart failure, reflecting differences in the nature of the unmet need for iron and the modifying effects of anaemia and inflammation on the handling of iron therapies by the body. Building on these insights, we identify untapped opportunities in the management of NAID, including the refinement of current approaches and the development of novel strategies. Lessons learned from NAID in cardiovascular disease could ultimately translate into benefits for patients with other chronic conditions such as chronic kidney disease, chronic obstructive pulmonary disease and cancer.
Collapse
Affiliation(s)
| | - John G F Cleland
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| |
Collapse
|
3
|
Frydrych A, Frankowski M, Jurowski K. The toxicological analysis and assessment of essential elements (Cu, Fe, Mn, Zn) in Food for Special Medical Purposes (FSMP) dedicated to oncological patients available in Polish pharmacies. Food Chem Toxicol 2024; 189:114768. [PMID: 38810942 DOI: 10.1016/j.fct.2024.114768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Foods for Special Medical Purposes (FSMP) for oncology patients, available in pharmacies, play a crucial role in providing nutrition and supplementation. However, the scientific literature lacks comprehensive research on the safety of essential trace elements in these products. This study aimed to assess Cu, Fe, Mn and Zn levels in commonly prescribed FSMPs (n = 23) from Polish pharmacies. Using ICP-MS after microwave-induced digestion (using concentrated nitric acid and hydrogen peroxide), we evaluated element levels. Our research used three approaches: the raw score for Cu, Fe, Mn, and Zn; single intake per serving; and the daily ration, compared with the reference values of the European Food Safety Authority. Discrepancies were found between the actual and declared product compositions, influenced by the route of administration and the recommended intake. Despite variations, all products were considered safe for oncological patients based on current evidence. However, it is recommended to have clear guidelines for FSMPs in cancer care. This pioneering study evaluates the safety and quality of prescription FSMPs for cancer patients from toxicological and nutritional perspectives, highlighting the need for standardised protocols in pharmacy-dispensed FSMPs.
Collapse
Affiliation(s)
- Adrian Frydrych
- Laboratory of Innovative Toxicological Research and Analyzes, Institute of Medical Sciences, Medical College, Rzeszów University, Al. mjr. W. Kopisto 2a, 35-959, Rzeszów, Poland
| | - Marcin Frankowski
- Department of Analytical and Environmental Chemistry, Faculty of Chemistry, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 8, 61-614, Poznań, Poland
| | - Kamil Jurowski
- Laboratory of Innovative Toxicological Research and Analyzes, Institute of Medical Sciences, Medical College, Rzeszów University, Al. mjr. W. Kopisto 2a, 35-959, Rzeszów, Poland; Department of Regulatory and Forensic Toxicology, Institute of Medical Expertises in Łódź, ul. Aleksandrowska 67/93, 91-205, Łódź, Poland.
| |
Collapse
|
4
|
Gete DG, Doust J, Mortlock S, Montgomery G, Mishra GD. Risk of Iron Deficiency in Women With Endometriosis: A Population-Based Prospective Cohort Study. Womens Health Issues 2024; 34:317-324. [PMID: 38658289 DOI: 10.1016/j.whi.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/25/2024] [Accepted: 03/15/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Endometriosis may be linked to the risk of iron deficiency through chronic systemic inflammation or heavy menstrual bleeding. No longitudinal studies, however, have examined the relationship between endometriosis and the risk of iron deficiency. METHODS This study included 3,294 participants born from 1973 to 1978 and followed as part of the Australian Longitudinal Study on Women's Health from 2000 to 2018. Participants with endometriosis were identified using self-reported longitudinal surveys linked to administrative health records. During each survey, participants were also asked to report the diagnosis of iron deficiency, and we validated diagnoses using an administrative health database. Generalized estimating equations for binary responses with an autoregressive correlation matrix were used to examine the association between endometriosis and the risk of iron deficiency over the seven time points. FINDINGS We found that women with endometriosis had a significantly higher risk of iron deficiency than those without endometriosis after adjusting for sociodemographic, lifestyle, reproductive, and nutrition factors (adjusted odds ratio [aOR] = 1.46; 95% confidence interval [CI] [1.29, 1.66]; p < .0001). Women with a surgically confirmed diagnosis and those with clinically suspected endometriosis had a higher risk of iron deficiency (aOR = 1.38; 95% CI [1.17, 1.64] and aOR = 1.53; 95% CI [1.30, 1.81]), respectively. These associations, however, were slightly attenuated (by 8%) when adjusted for the presence of heavy menstrual bleeding. CONCLUSIONS Women with endometriosis are at a higher risk of developing iron deficiency than those without endometriosis. The findings suggest that iron deficiency should be concomitantly addressed during initial diagnosis and successive management of endometriosis.
Collapse
Affiliation(s)
- Dereje G Gete
- Australian Women and Girls' Health Research Centre, School of Public Health, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| | - Jenny Doust
- Australian Women and Girls' Health Research Centre, School of Public Health, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Sally Mortlock
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Grant Montgomery
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Gita D Mishra
- Australian Women and Girls' Health Research Centre, School of Public Health, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
5
|
Zeidan RS, Martenson M, Tamargo JA, McLaren C, Ezzati A, Lin Y, Yang JJ, Yoon HS, McElroy T, Collins JF, Leeuwenburgh C, Mankowski RT, Anton S. Iron homeostasis in older adults: balancing nutritional requirements and health risks. J Nutr Health Aging 2024; 28:100212. [PMID: 38489995 DOI: 10.1016/j.jnha.2024.100212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/17/2024]
Abstract
Iron plays a crucial role in many physiological processes, including oxygen transport, bioenergetics, and immune function. Iron is assimilated from food and also recycled from senescent red blood cells. Iron exists in two dietary forms: heme (animal based) and non-heme (mostly plant based). The body uses iron for metabolic purposes, and stores the excess mainly in splenic and hepatic macrophages. Physiologically, iron excretion in humans is inefficient and not highly regulated, so regulation of intestinal absorption maintains iron homeostasis. Iron losses occur at a steady rate via turnover of the intestinal epithelium, blood loss, and exfoliation of dead skin cells, but overall iron homeostasis is tightly controlled at cellular and systemic levels. Aging can have a profound impact on iron homeostasis and induce a dyshomeostasis where iron deficiency or overload (sometimes both simultaneously) can occur, potentially leading to several disorders and pathologies. To maintain physiologically balanced iron levels, reduce risk of disease, and promote healthy aging, it is advisable for older adults to follow recommended daily intake guidelines and periodically assess iron levels. Clinicians can evaluate body iron status using different techniques but selecting an assessment method primarily depends on the condition being examined. This review provides a comprehensive overview of the forms, sources, and metabolism of dietary iron, associated disorders of iron dyshomeostasis, assessment of iron levels in older adults, and nutritional guidelines and strategies to maintain iron balance in older adults.
Collapse
Affiliation(s)
- Rola S Zeidan
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA; Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Matthew Martenson
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Javier A Tamargo
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Christian McLaren
- Department of Clinical and Health Psychology, College of Health and Health Professions, University of Florida, Gainesville, Florida, USA
| | - Armin Ezzati
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA; Department of Food, Nutrition, Dietetics and Health, Kansas State University, Manhattan, KS, USA
| | - Yi Lin
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jae Jeong Yang
- UF Health Cancer Center, Gainesville, FL, USA; Department of Surgery, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Hyung-Suk Yoon
- UF Health Cancer Center, Gainesville, FL, USA; Department of Surgery, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Taylor McElroy
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA; Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - James F Collins
- Department of Food Science & Human Nutrition, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, USA
| | - Christiaan Leeuwenburgh
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Robert T Mankowski
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Stephen Anton
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA; Department of Clinical and Health Psychology, College of Health and Health Professions, University of Florida, Gainesville, Florida, USA.
| |
Collapse
|
6
|
Shao M, Cheng H, Li X, Qiu Y, Zhang Y, Chang Y, Fu J, Shen M, Xu X, Feng D, Han Y, Yue S, Zhou Y, Luo Z. Abnormal mitochondrial iron metabolism damages alveolar type II epithelial cells involved in bleomycin-induced pulmonary fibrosis. Theranostics 2024; 14:2687-2705. [PMID: 38773980 PMCID: PMC11103499 DOI: 10.7150/thno.94072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/08/2024] [Indexed: 05/24/2024] Open
Abstract
Rationale: Pulmonary fibrosis is a chronic progressive lung disease with limited therapeutic options. We previously revealed that there is iron deposition in alveolar epithelial type II cell (AECII) in pulmonary fibrosis, which can be prevented by the iron chelator deferoxamine. However, iron in the cytoplasm and the mitochondria has two relatively independent roles and regulatory systems. In this study, we aimed to investigate the role of mitochondrial iron deposition in AECII injury and pulmonary fibrosis, and to find potential therapeutic strategies. Methods: BLM-treated mice, MLE-12 cells, and primary AECII were employed to establish the mouse pulmonary fibrosis model and epithelial cells injury model, respectively. Mitochondrial transplantation, siRNA and plasmid transfection, western blotting (WB), quantitative real-time polymerase chain reaction (RT-qPCR), polymerase chain reaction (PCR), immunofluorescence, immunoprecipitation (IP), MitoSOX staining, JC-1 staining, oxygen consumption rate (OCR) measurement, and Cell Counting Kit-8 (CCK8) assay were utilized to elucidate the role of mitochondrial iron deposition in cell and lung fibrosis and determine its mechanism. Results: This study showed that prominent mitochondrial iron deposition occurs within AECII in bleomycin (BLM)-induced pulmonary fibrosis mouse model and in BLM-treated MLE-12 epithelial cells. Further, the study revealed that healthy mitochondria rescue BLM-damaged AECII mitochondrial iron deposition and cell damage loss. Mitoferrin-2 (MFRN2) is the main transporter that regulates mitochondrial iron metabolism by transferring cytosolic iron into mitochondria, which is upregulated in BLM-treated MLE-12 epithelial cells. Direct overexpression of MFRN2 causes mitochondrial iron deposition and cell damage. In this study, decreased ubiquitination of the ubiquitin ligase F-box/LRR-repeat protein 5 (FBXL5) degraded iron-reactive element-binding protein 2 (IREB2) and promoted MFRN2 expression as well as mitochondrial iron deposition in damaged AECII. Activation of the prostaglandin E2 receptor EP4 subtype (EP4) receptor signaling pathway counteracted mitochondrial iron deposition by downregulating IREB2-MFRN2 signaling through upregulation of FBXL5. This intervention not only reduced mitochondrial iron content but also preserved mitochondrial function and protected against AECII damage after BLM treatment. Conclusion: Our findings highlight the unexplored roles, mechanisms, and regulatory approaches of abnormal mitochondrial iron metabolism of AECII in pulmonary fibrosis. Therefore, this study deepens the understanding of the mechanisms underlying pulmonary fibrosis and offers a promising strategy for developing effective therapeutic interventions using the EP4 receptor activator.
Collapse
Affiliation(s)
- Min Shao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Haipeng Cheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China
| | - Xiaohong Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410000, China
| | - Yujia Qiu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yunna Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yanfen Chang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Jiafeng Fu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Mengxia Shen
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xinxin Xu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Dandan Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yang Han
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - ShaoJie Yue
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Yan Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Ziqiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, Hunan, 410013, China
| |
Collapse
|
7
|
Rogez J, Urbanski G, Vinatier E, Lavigne C, Emmanuel L, Dupin I, Ravaiau C, Lacombe V. Iron deficiency in pernicious anemia: Specific features of iron deficient patients and preliminary data on response to iron supplementation. Clin Nutr 2024; 43:1025-1032. [PMID: 38527394 DOI: 10.1016/j.clnu.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/09/2024] [Accepted: 03/16/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND & AIMS While vitamin B12 (B12) deficiency is considered as the hallmark of pernicious anemia (PA), iron deficiency (ID) is also prevalent. Indeed, this auto immune gastritis is responsible for parietal cell atrophy and increase in gastric pH, leading to impaired iron absorption. We compared PA patients' features according to their iron status at PA diagnosis, and we assessed the iron status recovery after oral or intravenous iron supplementation. METHODS We prospectively included patients presenting with a newly diagnosed PA in a tertiary referral hospital between November 2018 and October 2020. Iron status was assessed at PA diagnosis then regularly during a standardized follow-up. In case of ID, the decision of treatment with oral and/or intravenous iron supplementation was left to the clinician convenience. RESULTS We included 28 patients with newly diagnosed PA. ID was observed in 21/28 (75.0%) patients: from the PA diagnosis in 13 patients, or during the follow-up in 8 patients. Iron deficient PA patients had higher plasma B12 (p = 0.04) and lower homocysteine levels (p = 0.04). Also, ID was independently associated with the 'APCA (anti-parietal cell antibodies) alone' immunological status (absence of anti-intrinsic factor antibodies) after adjustment for age, gender and B12 level (aOR 12.1 [1.1-141.8], p = 0.04). High level of APCA was associated with lower ferritin level. After 3 months of supplementation, 3/11 PA patients normalized the iron status with oral iron supplementation, versus 7/8 with intravenous iron supplementation (p = 0.02). CONCLUSION The high frequency of iron deficiency in PA highlights the interest of regular assessment of iron status in this condition. ID was associated with a profile including APCA alone and less pronounced B12 deficiency. Intravenous iron supplementation seemed to be more efficient than an oral supplementation in these preliminary data.
Collapse
Affiliation(s)
- Juliette Rogez
- Department of Internal Medicine and Clinical Immunology, Angers University Hospital, Angers, France
| | - Geoffrey Urbanski
- Department of Internal Medicine and Clinical Immunology, Angers University Hospital, Angers, France; Department of Immunology and Allergology, Geneva University Hospital, Geneve, Switzerland; Department of Orofacial Sciences, School of Dentistry, University of California, San Francisco, CA, USA
| | - Emeline Vinatier
- Laboratory of Immunology, Angers University Hospital, Angers, France
| | - Christian Lavigne
- Department of Internal Medicine and Clinical Immunology, Angers University Hospital, Angers, France
| | - Léa Emmanuel
- Department of Internal Medicine and Clinical Immunology, Angers University Hospital, Angers, France
| | - Iris Dupin
- Department of Internal Medicine and Clinical Immunology, Angers University Hospital, Angers, France
| | - Camille Ravaiau
- Department of Internal Medicine and Clinical Immunology, Angers University Hospital, Angers, France
| | - Valentin Lacombe
- Department of Internal Medicine and Clinical Immunology, Angers University Hospital, Angers, France; Univ Angers, MitoLab, Unité MITOVASC, UMR CNRS 6015, INSERM U1083, SFR ICAT, Angers, France.
| |
Collapse
|
8
|
Matsuoka T, Abe M, Kobayashi H. Iron Metabolism and Inflammatory Mediators in Patients with Renal Dysfunction. Int J Mol Sci 2024; 25:3745. [PMID: 38612557 PMCID: PMC11012052 DOI: 10.3390/ijms25073745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Chronic kidney disease (CKD) affects around 850 million people worldwide, posing significant challenges in healthcare due to complications like renal anemia, end-stage kidney disease, and cardiovascular diseases. This review focuses on the intricate interplay between iron metabolism, inflammation, and renal dysfunction in CKD. Renal anemia, prevalent in CKD, arises primarily from diminished erythropoietin (EPO) production and iron dysregulation, which worsens with disease progression. Functional and absolute iron deficiencies due to impaired absorption and chronic inflammation are key factors exacerbating erythropoiesis. A notable aspect of CKD is the accumulation of uremic toxins, such as indoxyl sulfate (IS), which hinder iron metabolism and worsen anemia. These toxins directly affect renal EPO synthesis and contribute to renal hypoxia, thus playing a critical role in the pathophysiology of renal anemia. Inflammatory cytokines, especially TNF-α and IL-6, further exacerbate CKD progression and disrupt iron homeostasis, thereby influencing anemia severity. Treatment approaches have evolved to address both iron and EPO deficiencies, with emerging therapies targeting hepcidin and employing hypoxia-inducible factor (HIF) stabilizers showing potential. This review underscores the importance of integrated treatment strategies in CKD, focusing on the complex relationship between iron metabolism, inflammation, and renal dysfunction to improve patient outcomes.
Collapse
Affiliation(s)
| | | | - Hiroki Kobayashi
- Division of Nephrology, Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine, Tokyo 173-8610, Japan
| |
Collapse
|
9
|
Zhao R, Liu JH, Qiu ZL. Value of serum iron and urine neutrophil gelatinase-associated lipocalin in predicting the mortality of critically ill patients with sepsis. CLIN INVEST MED 2024; 47:4-12. [PMID: 38546383 DOI: 10.3138/cim-2024-2672] [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: 04/04/2024]
Abstract
INTRODUCTION We aimed to investigate the association of iron metabolism-related parameters with 60-day mortality in critically ill patients with sepsis. METHODS Serum or urine concentrations of iron metabolism-related parameters on intensive care unit admission were measured in a prospective cohort of 133 eligible patients with sepsis according to the Sepsis-3 criteria, and these values were compared between survivors and nonsurvivors, categorized according to their 60-day survival status. Cox regression analyses were performed to examine the association between iron parameters and 60-day mortality. Kaplan-Meier methods were used to illustrate the differences in survival between different iron parameters. RESULTS Of the 133 patients included in the study, 61 (45.8%) had died by day 60. After adjusting for confounding variables, higher concentrations of serum iron (cut-off 9.5 μmol/mL) and higher concentrations of urine neutrophil gelatinase-associated lipocalin (uNGAL; cut-off 169.3 ng/mL) were associated with a significantly greater risk of death in the Cox regression analysis. These two biomarkers combined with Sequential Organ Failure Assessment (SOFA) scores increased the area under the receiver operating characteristic (AUROC) curve to 0.85. DISCUSSION These findings suggest that higher concentrations of serum iron and uNGAL are each associated with higher 60-day mortality, and they add significant accuracy to this prediction in combination with SOFA. Abbreviations: uNGAL: urine neutrophil gelatinase-associated lipocalin; ICU: intensive care unit; SOFA: Sequential Organ Failure Assessment; APACHE II: the Acute Physiology and Chronic Health Evaluation II; ELISA: enzyme-linked immunosorbent assay; HR: hazard ratio; CIs: confidence intervals; WBC: white blood cell; TBIL: total bilirubin.
Collapse
Affiliation(s)
- Rui Zhao
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- These authors contributed equally to this work
| | - Jiang-Hua Liu
- Department of Medical Records and Statistics, Shanghai PuDong Guangming Hospital of Traditional Chinese Medicine, Shanghai, China
- These authors contributed equally to this work
| | - Ze-Liang Qiu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Emergency, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Sun B, Tan B, Zhang P, Zhu L, Wei H, Huang T, Li C, Yang W. Iron deficiency anemia: a critical review on iron absorption, supplementation and its influence on gut microbiota. Food Funct 2024; 15:1144-1157. [PMID: 38235788 DOI: 10.1039/d3fo04644c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Iron deficiency anemia (IDA) caused by micronutrient iron deficiency has attracted global attention due to its adverse health effects. The regulation of iron uptake and metabolism is finely controlled by various transporters and hormones in the body. Dietary iron intake and regulation are essential in maintaining human health and iron requirements. The review aims to investigate literature concerning dietary iron intake and systemic regulation. Besides, recent IDA treatment and dietary iron supplementation are discussed. Considering the importance of the gut microbiome, the interaction between bacteria and micronutrient iron in the gut is also a focus of this review. The iron absorption efficiency varies considerably according to iron type and dietary factors. Iron fortification remains the cost-effective strategy, although challenges exist in developing suitable iron fortificants and food vehicles regarding bioavailability and acceptability. Iron deficiency may alter the microbiome structure and promote the growth of pathogenic bacteria in the gut, affecting immune balance and human health.
Collapse
Affiliation(s)
- Bolun Sun
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
- School of Nursing, Wenzhou Medical University, Wenzhou 325035, China
| | - Beibei Tan
- School of Agriculture and Food, Faculty of Science, University of Melbourne, Australia
| | - Panxue Zhang
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| | - Lianlian Zhu
- School of Nursing, Wenzhou Medical University, Wenzhou 325035, China
| | - Huamao Wei
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| | - Tao Huang
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| | - Chao Li
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| | - Wenge Yang
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
11
|
Lüersen K, Jöckel T, Chin D, Demetrowitsch T, Schwarz K, Rimbach G. Reduced iron and cobalt levels in response to curcumin supplementation are not responsible for the prolonged larval development and do not affect the oxidative stress tolerance and polyamine status of D. melanogaster. Biofactors 2024; 50:161-180. [PMID: 37597249 DOI: 10.1002/biof.2000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/23/2023] [Indexed: 08/21/2023]
Abstract
Recent reports indicated that the phytochemical curcumin possesses iron-chelating activity. Here, by employing the fruit fly Drosophila melanogaster, we conducted feeding studies supplementing curcumin or, as a control, the iron chelator bathophenanthroline (BPA). First, the absorption and further metabolization of dietary curcuminoids were proved by metabolomics analyses. Next, we found that 0.2% dietary curcumin, similar to BPA, lowered the iron but also the cobalt content, and to a lesser extent affected the manganese and zinc status. Supplementation during larval stages was required and sufficient for both compounds to elicit these alterations in adult animals. However, curcumin-induced retarded larval development was not attributable to the changed trace metal status. In addition, a reduction in the iron content of up to 70% by curcumin or BPA supplementation did not reduce heme-dependent catalase activity and tolerance toward H2 O2 in D. melanogaster. Moreover, polyamines were not influenced by curcumin treatment and decreased iron levels. This was confirmed for selected organs from 0.2% curcumin-treated mice, except for the spleen. Here, elevated spermidine level and concomitant upregulation of genes involved in polyamine production were associated with a putatively anemia-derived increased spleen mass. Our data underline that the metal-chelating property of curcumin needs to be considered in feeding studies.
Collapse
Affiliation(s)
- Kai Lüersen
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Tobias Jöckel
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Dawn Chin
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Tobias Demetrowitsch
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Karin Schwarz
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Gerald Rimbach
- Division of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| |
Collapse
|
12
|
Long D, Mao C, Liu Y, Zhou T, Xu Y, Zhu Y. Evolving trends and burden of iron deficiency among children, 1990-2019: a systematic analysis for the global burden of disease study 2019. Front Nutr 2023; 10:1275291. [PMID: 38130442 PMCID: PMC10734639 DOI: 10.3389/fnut.2023.1275291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
Objectives We aimed to provide a timely, comprehensive, and reliable assessment of the burden of iron deficiency (ID) in children between 1990 and 2019 at the global, regional, and national levels to inform policymakers in developing locally appropriate health policies. Methods Data related to ID among children younger than 15 years old were analyzed by sex, age, year, socio-demographic index (SDI), and location according to the Global Burden of Disease Study 2019 (GBD 2019). Age-standardized rates were used to compare the burden between different regions and countries. Furthermore, the Joinpoint regression model was used to assess temporal trends from 1990 to 2019. Results In 2019, the number of prevalent cases and disability-adjusted life years (DALYs) for ID in children were 391,491,699 and 13,620,231, respectively. The global age-standardized prevalence and DALY rates for childhood ID in 2019 were 20,146.35 (95% confidence interval: 19,407.85 to 20,888.54) and 698.90 (466.54 to 1015.31) per 100,000, respectively. Over the past 30 years, the global prevalence of ID among children has been highest in low-SDI regions, particularly in Western Sub-Saharan Africa, South Asia, and Eastern Sub-Saharan Africa. Since 1990, the prevalence and DALY of ID in children have been declining in most geographic regions. Nationally, Ecuador, China, and Chile have shown the most significant decreases in prevalence. The greatest decline in age-standardized DALY rate was observed in Ecuador, while Burkina Faso experienced the highest increase. Bhutan had the highest prevalence and DALY rates in 2019. On the age level, the prevalence was relatively higher among the <5 years age group. At the gender dimension, the prevalence of ID in children overall was more pronounced in girls than in boys, as was the case for DALY. Conclusion Although the burden of ID in children has been declining, this disease remains a major public health problem, especially in countries with low SDI. Children younger than 5 years of age are an important group for whom targeted measures are needed to reduce the burden of ID.
Collapse
Affiliation(s)
- Dan Long
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chenhan Mao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yaxuan Liu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Tao Zhou
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yin Xu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ying Zhu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
13
|
Liu Y, Li G, Lu F, Guo Z, Cai S, Huo T. Excess iron intake induced liver injury: The role of gut-liver axis and therapeutic potential. Biomed Pharmacother 2023; 168:115728. [PMID: 37864900 DOI: 10.1016/j.biopha.2023.115728] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/23/2023] Open
Abstract
Excessive iron intake is detrimental to human health, especially to the liver, which is the main organ for iron storage. Excessive iron intake can lead to liver injury. The gut-liver axis (GLA) refers to the bidirectional relationship between the gut and its microbiota and the liver, which is a combination of signals generated by dietary, genetic and environmental factors. Excessive iron intake disrupts the GLA at multiple interconnected levels, including the gut microbiota, gut barrier function, and the liver's innate immune system. Excessive iron intake induces gut microbiota dysbiosis, destroys gut barriers, promotes liver exposure to gut microbiota and its derived metabolites, and increases the pro-inflammatory environment of the liver. There is increasing evidence that excess iron intake alters the levels of gut microbiota-derived metabolites such as secondary bile acids (BAs), short-chain fatty acids, indoles, and trimethylamine N-oxide, which play an important role in maintaining homeostasis of the GLA. In addition to iron chelators, antioxidants, and anti-inflammatory agents currently used in iron overload therapy, gut barrier intervention may be a potential target for iron overload therapy. In this paper, we review the relationship between excess iron intake and chronic liver diseases, the regulation of iron homeostasis by the GLA, and focus on the effects of excess iron intake on the GLA. It has been suggested that probiotics, fecal microbiota transfer, farnesoid X receptor agonists, and microRNA may be potential therapeutic targets for iron overload-induced liver injury by protecting gut barrier function.
Collapse
Affiliation(s)
- Yu Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China
| | - Guangyan Li
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China
| | - Fayu Lu
- School of Public Health, China Medical University, Shenyang, Liaoning 110122, China
| | - Ziwei Guo
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China
| | - Shuang Cai
- The First Affiliated Hospital of China Medical University, Shenyang 110001, China.
| | - Taoguang Huo
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
14
|
Kim EY, Park SE, Hong TH. What we should consider to facilitate recovery of the hematological profile in all patients after pancreaticoduodenectomy: the role of preoperative intravenous iron treatment. BMC Surg 2023; 23:308. [PMID: 37828447 PMCID: PMC10571369 DOI: 10.1186/s12893-023-02217-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 10/04/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND In pancreaticoduodenectomy (PD), the duodenum and upper jejunum responsible for iron absorption are removed, which can lead to massive hemorrhage during surgery and cause iron deficiency anemia after PD. The aim of this study was to evaluate overall changes in hematologic profiles of patients who underwent pancreaticoduodenectomy. Effect of preoperative intravenous iron treatment on recovery of anemia after surgery was also investigated. METHODS From March 2021 to December 2021, patients who underwent curative PD at our institution due to periampullary lesions were enrolled. They were divided into two groups according to whether or not iron was administered before surgery. In the IV iron group, all patients had been routinely administered with 1000 mg of ferric carboxymaltose intravenously once about 3-7 days before the operation day. Contrarily, patients in the control group did not receive intravenous iron before PD. Changes in hematological profile were measured preoperatively and at 5, 14, and 30 days postoperatively. Clinical results of the two groups were compared and analyzed. Additionally, a subgroup analysis was performed for selected non-anemic patients who had preoperative hemoglobin level of 12.0 g/dl or higher to compare changes in hematologic profiles between the two groups. RESULTS Thirty patients of the IV iron group and 34 patients of the control group were analyzed. Although no difference was observed in postoperative complications or mortality, hemoglobin and iron levels were recovered significantly faster at two weeks postoperatively in the IV iron group than in the control group. Iron levels were significantly higher in the IV iron group throughout the postoperative period. In subgroup analysis conducted for non-anemic patients, hemoglobin levels were recovered significantly faster and maintained higher in the IV iron group throughout the postoperative period, although baseline levels of hemoglobin were similar between the two groups. In addition, the length of intensive care unit stay was significantly shorter in the IV iron group than in the control group. CONCLUSIONS Preoperative intravenous iron treatment might be effective in facilitating recovery of hematologic profiles of patients during the recovery period after PD regardless of the presence of preoperative anemia, thus preventing postoperative iron deficiency anemia.
Collapse
Affiliation(s)
- Eun Young Kim
- Division of Trauma and Surgical Critical Care, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung Eun Park
- Division of Hepato-biliary and Pancreas Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Tae Ho Hong
- Division of Hepato-biliary and Pancreas Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| |
Collapse
|
15
|
Walter S, Mertens C, Muckenthaler MU, Ott C. Cardiac iron metabolism during aging - Role of inflammation and proteolysis. Mech Ageing Dev 2023; 215:111869. [PMID: 37678569 DOI: 10.1016/j.mad.2023.111869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/01/2023] [Accepted: 09/03/2023] [Indexed: 09/09/2023]
Abstract
Iron is the most abundant trace element in the human body. Since iron can switch between its 2-valent and 3-valent form it is essential in various physiological processes such as energy production, proliferation or DNA synthesis. Especially high metabolic organs such as the heart rely on iron-associated iron-sulfur and heme proteins. However, due to switches in iron oxidation state, iron overload exhibits high toxicity through formation of reactive oxygen species, underlining the importance of balanced iron levels. Growing evidence demonstrates disturbance of this balance during aging. While age-associated cardiovascular diseases are often related to iron deficiency, in physiological aging cardiac iron accumulates. To understand these changes, we focused on inflammation and proteolysis, two hallmarks of aging, and their role in iron metabolism. Via the IL-6-hepcidin axis, inflammation and iron status are strongly connected often resulting in anemia accompanied by infiltration of macrophages. This tight connection between anemia and inflammation highlights the importance of the macrophage iron metabolism during inflammation. Age-related decrease in proteolytic activity additionally affects iron balance due to impaired degradation of iron metabolism proteins. Therefore, this review accentuates alterations in iron metabolism during aging with regards to inflammation and proteolysis to draw attention to their implications and associations.
Collapse
Affiliation(s)
- Sophia Walter
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Molecular Toxicology, Nuthetal, Germany; TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Wuppertal, Germany; DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Christina Mertens
- Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Immunology, and Hematology, University of Heidelberg, Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Heidelberg, Mannheim, Germany
| | - Martina U Muckenthaler
- Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Immunology, and Hematology, University of Heidelberg, Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Heidelberg, Mannheim, Germany; Molecular Medicine Partnership Unit, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Christiane Ott
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Molecular Toxicology, Nuthetal, Germany; TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena, Wuppertal, Germany; DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| |
Collapse
|
16
|
Fahoum L, Belisowski S, Ghatpande N, Guttmann-Raviv N, Zhang W, Li K, Tong WH, Nyska A, Waterman M, Weisshof R, Zuckerman A, Meyron-Holtz E. Iron Regulatory Protein 1 is Required for the Propagation of Inflammation in Inflammatory Bowel Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.27.525690. [PMID: 36789413 PMCID: PMC9928023 DOI: 10.1101/2023.01.27.525690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Objective Inflammatory bowel diseases (IBD) are complex disorders. Iron accumulates in the inflamed tissue of IBD patients, yet neither a mechanism for the accumulation nor its implication on the course of inflammation are known. We hypothesized that the inflammation modifies iron homeostasis, affects tissue iron distribution and that this in turn perpetuates the inflammation. Design This study analyzed human biopsies, animal models and cellular systems to decipher the role of iron homeostasis in IBD. Results We found inflammation-mediated modifications of iron distribution, and iron-decoupled activation of the iron regulatory protein (IRP)1. To understand the role of IRP1 in the course of this inflammation-associated iron pattern, a novel cellular co-culture model was established, that replicated the iron-pattern observed in vivo, and supported involvement of nitric oxide in the activation of IRP1 and the typical iron pattern in inflammation. Importantly, deletion of IRP1 from an IBD mouse model completely abolished both, the misdistribution of iron and intestinal inflammation. Conclusion These findings suggest that IRP1 plays a central role in the coordination of the inflammatory response in the intestinal mucosa and that it is a viable candidate for therapeutic intervention in IBD.
Collapse
Affiliation(s)
- L. Fahoum
- Laboratory of Molecular Nutrition, Department of Biotechnology and Food Engineering, Technion– Israel Institute of Technology, Haifa, Israel
| | - S. Belisowski
- Laboratory of Molecular Nutrition, Department of Biotechnology and Food Engineering, Technion– Israel Institute of Technology, Haifa, Israel
| | - N. Ghatpande
- Laboratory of Molecular Nutrition, Department of Biotechnology and Food Engineering, Technion– Israel Institute of Technology, Haifa, Israel
| | - N. Guttmann-Raviv
- Laboratory of Molecular Nutrition, Department of Biotechnology and Food Engineering, Technion– Israel Institute of Technology, Haifa, Israel
| | - W. Zhang
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
| | - K. Li
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
| | - W-H. Tong
- Molecular Medicine Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - A. Nyska
- Tel Aviv University and Consultant in Toxicologic Pathology, Tel Aviv, Israel
| | - M. Waterman
- Rambam / Technion– Israel Institute of Technology, Haifa, Israel
| | - R. Weisshof
- Rambam / Technion– Israel Institute of Technology, Haifa, Israel
| | | | - E.G. Meyron-Holtz
- Laboratory of Molecular Nutrition, Department of Biotechnology and Food Engineering, Technion– Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
17
|
Sama SO, Taiwe GS, Teh RN, Njume GE, Chiamo SN, Sumbele IUN. Anaemia, iron deficiency and inflammation prevalence in children in the Mount Cameroon area and the contribution of inflammatory cytokines on haemoglobin and ferritin concentrations: a cross sectional study. BMC Nutr 2023; 9:94. [PMID: 37507740 PMCID: PMC10375674 DOI: 10.1186/s40795-023-00748-3] [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: 11/12/2022] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Iron deficiency (ID) and anaemia of inflammation (AI) coexist where infections and nutritional deficiencies are common. The aim of this study was to determine burden of ID, anaemia, inflammation and AI in children in malaria endemic Limbe, Mount Cameroon as well as decipher the contribution of some inflammatory cytokines on the concentration of haemoglobin and ferritin. METHODS A total of 520 children aged ≤ 15 years old from the Limbe Health District (LHD) were randomly selected and examined in a cross-sectional study for iron deficiency, anaemia, inflammation and inflammation anaemia. Collected blood samples were used for full blood count and inflammatory marker analyses with the aid of a haemoanalyzer and ELISA machine, respectively. Spearman's rank correlation analysis was used to determine the correlation between cytokines and haemoglobin while multiple linear regression analysis was used to evaluate the effects of inflammatory cytokines on haemoglobin and ferritin concentrations. RESULTS The overall prevalence of anaemia, ID, IDA, inflammation and AI were respectively, 67.5%, 34.6%, 12.9%, 63.1% and 30.2%. Children aged 12‒15 years (P = 0.001), enrolled from the community (P < 0.001), whose parents are civil servants (P < 0.001), living in a home with 6‒10 occupants (P = 0.016), afebrile (P < 0.001) and malaria negative (P = 0.007) had the highest prevalence of ID while, children ≤ 5 years old (P = 0.001), with a family size of 1‒5 occupants (P = 0.033) had the highest prevalence of AI. Haemoglobin concentration positively correlated with concentrations of IFN-γ (P < 0.001), TNF-α (0.045) and ferritin (P < 0.001) while a negative correlation was observed with IL-10 (P = 0.003). In the multiple linear regression analysis only IL-6 significantly (P = 0.030) influenced haemoglobin concentration. CONCLUSIONS While IL-6 is of significance in the pathology of anaemia, iron deficiency and anaemia of inflammation are of moderate public health concerns in the Mount Cameroon area. Hence, appropriate intervention against anaemia, ID and AI should be directed at children ≤ 5 years and counterparts > 10 years old that bear the highest burden.
Collapse
Affiliation(s)
- Sharon Odmia Sama
- Department of Animal Biology and Conservation, University of Buea, Buea, Cameroon
| | | | - Rene Ning Teh
- Department of Animal Biology and Conservation, University of Buea, Buea, Cameroon.
| | | | | | | |
Collapse
|
18
|
Liu Y, Wang Z, Kelimu A, Korma SA, Cacciotti I, Xiang H, Cui C. Novel iron-chelating peptide from egg yolk: Preparation, characterization, and iron transportation. Food Chem X 2023; 18:100692. [PMID: 37151212 PMCID: PMC10154770 DOI: 10.1016/j.fochx.2023.100692] [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: 01/31/2023] [Revised: 04/09/2023] [Accepted: 04/21/2023] [Indexed: 05/09/2023] Open
Abstract
In this work, an egg yolk protein hydrolysate (EYPH) with a high iron-chelating ability (87.32%) was prepared. The fractionation using 60% (v/v) ethanol concentration (E3 fraction) led to the efficiently accumulating the iron-chelating peptides in EYPH. The characterization results showed that iron mainly chelated with carboxyl, amino and phosphate groups of peptides. From E3 fraction, six iron-chelating peptides with MW ranging from 1372.36 to 2937.04 Da were identified and a hypothesized molecular model of DDSSSpSpSpSpSpSVLSK-Fe was simulated. In vitro stability determination showed that E3-Fe chelate owned a good heat, alkalinity and digestion tolerance, but a relatively bad acid tolerance. Finally, iron transport analysis showed that iron in the E3-Fe would be absorbed in caco-2 cell membrane more effectively than that of iron salts, indicating that it was possible to apply the E3-Fe complex as iron supplements.
Collapse
Affiliation(s)
- Ying Liu
- College of Food Science and Engineering, South China University of Technology, Wushan Road 381, 510640 Guangzhou, Guangdong, China
| | - Zhuo Wang
- College of Food Science and Engineering, South China University of Technology, Wushan Road 381, 510640 Guangzhou, Guangdong, China
| | - Abulimiti Kelimu
- College of Food Science and Pharmacy, Xinjiang Agricultural University, Nongda East Road 311, 830052 Urumqi, Xinjiang, China
| | - Sameh A. Korma
- Department of Food Science, Faculty of Agriculture, Zagazig University, El-Zeraa Road 114, 44519 Zagazig, Sharkia, Egypt
| | - Ilaria Cacciotti
- Department of Engineering, INSTM RU, University of Rome “Niccolò Cusano”, 3 via Don Carlo Gnocchi, 3 00166 Roma, Italy
| | - Huan Xiang
- College of Food Science and Engineering, South China University of Technology, Wushan Road 381, 510640 Guangzhou, Guangdong, China
| | - Chun Cui
- College of Food Science and Engineering, South China University of Technology, Wushan Road 381, 510640 Guangzhou, Guangdong, China
- Corresponding author.
| |
Collapse
|
19
|
Olivier RMR, Macke M, Müller JC, Schrader L, Eveslage M, Rauer M, Wempe C, Martens S, Zarbock A, Wagner NM, Karst U, Dogan DY, Steinbicker AU. Perioperative Tracking of Intravenous Iron in Patients Undergoing On-Pump Cardiac Surgery: A Prospective, Single-Center Pilot Trial. Anesth Analg 2023; 136:578-587. [PMID: 36811991 DOI: 10.1213/ane.0000000000006372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
BACKGROUND Preoperative intravenous iron administration is a frequently used patient blood management procedure. If the timeframe of intravenous iron administration before surgery is short, (1) the concentration of the intravenous iron compound might still be high in patients' plasma when undergoing surgery and (2) this iron in patients' plasma is at risk to be lost due to blood loss. The aim of the current study was, therefore, to track the iron compound ferric carboxymaltose (FCM) before, during, and after cardiac surgery requiring cardiopulmonary bypass, with an emphasis on intraoperative iron losses in shed blood and potential recovery through autologous cell salvage. METHODS Concentrations of FCM were analyzed in patients' blood using a hyphenation of liquid chromatography and inductively coupled plasma-mass spectrometry to distinguish between pharmaceutical compound FCM and serum iron. In this prospective, single-center pilot trial, 13 anemic and 10 control patients were included. Anemic patients with hemoglobin levels ≤12/13 g/dL in women and men were treated with 500 milligrams (mg) intravenous FCM 12 to 96 hours before elective on-pump cardiac surgery. Patients' blood samples were collected before surgery and at days 0, 1, 3, and 7 after surgery. One sample each was taken of the cardiopulmonary bypass, the autologous red blood cell concentrate generated by cell salvage, and the cell salvage disposal bag. RESULTS Patients who had received FCM <48 hours before surgery had higher FCM serum levels (median [Q1-Q3], 52.9 [13.0-91.6]) compared to ≥48 hours (2.1 [0.7-5.1] µg/mL, P = .008). Of 500-mg FCM administered <48 hours, 327.37 (257.96-402.48) mg were incorporated compared to administration ≥48 hours with 493.60 (487.78-496.70) mg. After surgery, patients' plasma FCM concentration in the FCM <48 hours group was decreased (-27.1 [-30 to -5.9] µg/mL). Little FCM was found in the cell salvage disposal bag (<48 hours, 4.2 [3.0-25.8] µg/mL, equivalent to 29.0 [19.0-40.7] mg total; equivalent to 5.8% or 1/17th of the 500 mg FCM initially administered), almost none in the autologous red blood cell concentrate (<48 hours, 0.1 [0.0-0.43] µg/mL). CONCLUSIONS The data generate the hypotheses that nearly all FCM is incorporated into iron stores with administration ≥48 hours before surgery. When FCM is given <48 hours of surgery, the majority is incorporated into iron stores by the time of surgery, although a small amount may be lost during surgical bleeding with limited recovery by cell salvage.
Collapse
Affiliation(s)
- Roman M R Olivier
- From the Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Marcel Macke
- Institute of Inorganic and Analytical Chemistry, University of Muenster, Muenster, Germany
| | - Jennifer C Müller
- Institute of Inorganic and Analytical Chemistry, University of Muenster, Muenster, Germany
| | - Lisa Schrader
- From the Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Maria Eveslage
- Institute of Biostatistics and Clinical Research, University of Muenster, Muenster, Germany
| | - Marcel Rauer
- From the Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Carola Wempe
- From the Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Sven Martens
- Department of Thoracic, Heart and Vascular Surgery, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Alexander Zarbock
- From the Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Nana-Maria Wagner
- From the Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Muenster, Muenster, Germany
| | - Deniz Y Dogan
- From the Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany.,Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Andrea U Steinbicker
- From the Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany.,Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| |
Collapse
|
20
|
Dalto DB, Audet I, Matte JJ, Lapointe J. Effects of high levels of zinc oxide and dietary zinc/copper ratios on the metabolism of iron in weaned pigs. J Anim Sci 2023; 101:skad391. [PMID: 38006248 PMCID: PMC10718792 DOI: 10.1093/jas/skad391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/24/2023] [Indexed: 11/26/2023] Open
Abstract
The present study compares the use different levels of dietary zinc oxide and zinc/copper ratios on the metabolism of iron (Fe) in weaned pigs. Two experiments were conducted using 120 and 160 weanling piglets (7.96 ± 1.17 kg and 7.81 ± 0.25 kg body weight, respectively) that were randomly assigned to the experimental treatments. Experiment I: diets supplemented with 100, 1,000, and 3,000 mg/kg of zinc (Zn) as ZnO (LZn, MZn, HZn) and 130 mg/kg of copper (Cu) as CuSO4; experiment II: diets supplemented with 100 or 3,000 mg/kg of Zn as ZnO (LZn and HZn) in combination with 6 or 130 mg/kg of Cu as CuSO4 (LCu and HCu). In both experiments, diets had similar levels of supplemental Fe (100 mg/kg of Fe as FeSO4). Piglets were slaughtered at d21 (weaning), d23 (experiment I), d28 (experiment II), d35, and d42 to assess whole blood, serum, and liver Fe concentrations, hemoglobin concentration, and the relative expression of key genes associated with Fe metabolism in jejunum and liver. Whole blood Fe and hemoglobin concentrations (experiment I) as well as serum Fe concentrations (experiments I and II) were not affected by dietary treatments (P ≥ 0.11). Liver Fe concentrations (experiment II) and total liver Fe content (experiments I and II) were lower (P ≤ 0.05) in HZn compared to LZn groups at d42. In both experiments, the mRNA expression of jejunal DMT1 was lowest and that of jejunal FTH1 was highest at d42 (P ≤ 0.04) for HZn piglets. In experiment II only, jejunal FTH1 and FPN1 expression were greater (P ≤ 0.04) in HCu compared to LCu groups at d42. The highest expression of hepatic FTH1 and FPN1 at d35 and d42 (P ≤ 0.02) was detected in HZn piglets in both experiments. For hepatic HAMP, expression values were greater (P = 0.04) at d42 in HZn groups. In conclusion, high dietary ZnO levels impair Fe metabolism but the effects are not intense enough to impact circulating Fe and hemoglobin concentrations.
Collapse
Affiliation(s)
- Danyel Bueno Dalto
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, Quebec J1M 0C8, Canada
| | - Isabelle Audet
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, Quebec J1M 0C8, Canada
| | - Jean-Jacques Matte
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, Quebec J1M 0C8, Canada
| | - Jérôme Lapointe
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, Quebec J1M 0C8, Canada
| |
Collapse
|
21
|
De Souza LV, Hoffmann A, Fischer C, Petzer V, Asshoff M, Theurl I, Tymoszuk P, Seifert M, Brigo N, Hilbe R, Demetz E, Von Raffay L, Berger S, Barros-Pinkelnig M, Weiss G. Comparative analysis of oral and intravenous iron therapy in rat models of inflammatory anemia and iron deficiency. Haematologica 2023; 108:135-149. [PMID: 35796011 PMCID: PMC9827174 DOI: 10.3324/haematol.2022.281149] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/29/2022] [Indexed: 02/05/2023] Open
Abstract
Anemia is a major health issue and associated with increased morbidity. Iron deficiency anemia (IDA) is the most prevalent, followed by anemia of chronic disease (ACD). IDA and ACD often co-exist, challenging diagnosis and treatment. While iron supplementation is the first-line therapy for IDA, its optimal route of administration and the efficacy of different repletion strategies in ACD are elusive. Female Lewis rats were injected with group A streptococcal peptidoglycan-polysaccharide (PG-APS) to induce inflammatory arthritis with associated ACD and/or repeatedly phlebotomized and fed with a low iron diet to induce IDA, or a combination thereof (ACD/IDA). Iron was either supplemented by daily oral gavage of ferric maltol or by weekly intravenous (i.v.) injection of ferric carboxymaltose for up to 4 weeks. While both strategies reversed IDA, they remained ineffective to improve hemoglobin (Hb) levels in ACD, although oral iron showed slight amelioration of various erythropoiesis-associated parameters. In contrast, both iron treatments significantly increased Hb in ACD/IDA. In ACD and ACD/IDA animals, i.v. iron administration resulted in iron trapping in liver and splenic macrophages, induction of ferritin expression and increased circulating levels of the iron hormone hepcidin and the inflammatory cytokine interleukin-6, while oral iron supplementation reduced interleukin-6 levels. Thus, oral and i.v. iron resulted in divergent effects on systemic and tissue iron homeostasis and inflammation. Our results indicate that both iron supplements improve Hb in ACD/IDA, but are ineffective in ACD with pronounced inflammation, and that under the latter condition, i.v. iron is trapped in macrophages and may enhance inflammation.
Collapse
Affiliation(s)
- Lara Valente De Souza
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia research, Medical University of Innsbruck
| | - Alexander Hoffmann
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia research, Medical University of Innsbruck
| | - Christine Fischer
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Verena Petzer
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Malte Asshoff
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Igor Theurl
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Markus Seifert
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia research, Medical University of Innsbruck
| | - Natascha Brigo
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Richard Hilbe
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Egon Demetz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Laura Von Raffay
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Sylvia Berger
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Marina Barros-Pinkelnig
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Guenter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia research, Medical University of Innsbruck.
| |
Collapse
|
22
|
Hof L, Old O, Steinbicker A, Meybohm P, Choorapoikayil S, Zacharowski K. Iron deficiency in cardiac surgical patients. ACTA ANAESTHESIOLOGICA BELGICA 2022. [DOI: 10.56126/73.4.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Iron is an essential element and involved in a variety of metabolic processes including oxygen transport, cellular energy production, energy metabolism of heart muscles, brain function, cell growth and cell differentiation. Preoperative anaemia is an independent risk factor for poor outcome. Recently, iron deficiency was considered only in the context of anaemia. However, negative consequences of iron deficiency in the absence of anaemia have been described for patients undergoing cardiac surgery. To date, the benefit of intravenous iron supplementation in these patients has been controversially debated. In this review, we discuss the latest progress in studies of intravenous iron supplementation in iron deficient cardiac surgical patients.
Collapse
|
23
|
Blum LV, Schmitt E, Choorapoikayil S, Baumhove O, Bayer A, Friederich P, Friedrich J, Geisen C, Gruenewald M, Gutjahr M, Herrmann E, Müller M, Narita D, Raadts A, Schwendner K, Seifried E, Stark P, Thoma J, Weigt H, Wiesenack C, Steinbicker AU, Zacharowski K, Meybohm P. Association of anaemia, co-morbidities and red blood cell transfusion according to age groups: multicentre sub-analysis of the German Patient Blood Management Network Registry. BJS Open 2022; 6:6794769. [PMID: 36326235 PMCID: PMC9631974 DOI: 10.1093/bjsopen/zrac128] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/24/2022] [Indexed: 11/06/2022] Open
Abstract
Background Blood transfusions are common medical procedures and every age group requires detailed insights and treatment bundles. The aim of this study was to examine the association of anaemia, co-morbidities, complications, in-hospital mortality, and transfusion according to age groups to identify patient groups who are particularly at risk when undergoing surgery. Methods Data from 21 Hospitals of the Patient Blood Management Network Registry were analysed. Patients were divided into age subgroups. The incidence of preoperative anaemia, co-morbidities, surgical disciplines, hospital length of stay, complications, in-hospital mortality rate, and transfusions were analysed by descriptive and multivariate regression analysis. Results A total of 1 117 919 patients aged 18–108 years were included. With increasing age, the number of co-morbidities and incidence of preoperative anaemia increased. Complications, hospital length of stay, and in-hospital mortality increased with age and were higher in patients with preoperative anaemia. The mean number of transfused red blood cells (RBCs) peaked, whereas the transfusion rate increased continuously. Multivariate regression analysis showed that increasing age, co-morbidities, and preoperative anaemia were independent risk factors for complications, longer hospital length of stay, in-hospital mortality, and the need for RBC transfusion. Conclusion Increasing age, co-morbidities, and preoperative anaemia are independent risk factors for complications, longer hospital length of stay, in-hospital mortality, and the need for RBC transfusion. Anaemia diagnosis and treatment should be established in all patients.
Collapse
Affiliation(s)
- Lea Valeska Blum
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Elke Schmitt
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany.,Department of Medicine, Institute of Biostatistics and Mathematical Modelling, Goethe University, Frankfurt, Germany
| | - Suma Choorapoikayil
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Olaf Baumhove
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, Klinikum Westmuensterland, Bocholt, Germany
| | - Alexandra Bayer
- Department of Anaesthesiology and Intensive Care Medicine, Agatharied Hospital, Hausham, Germany
| | - Patrick Friederich
- Department of Anaesthesiology, Operative Intensive Care Medicine and Pain Therapy, Munich, Germany
| | - Jens Friedrich
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum Leverkusen, Leverkusen, Germany
| | - Christof Geisen
- German Red Cross, Institute for Transfusion Medicine and Immunohaematology, German Red Cross Baden-Wuertemberg-Hessen, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Matthias Gruenewald
- Department of Anaesthesiology and Operative Intensive Care Medicine, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Martin Gutjahr
- Department of Anaesthesiology, Marienhaus, Ottweiler, Germany
| | - Eva Herrmann
- Department of Medicine, Institute of Biostatistics and Mathematical Modelling, Goethe University, Frankfurt, Germany
| | - Markus Müller
- German Red Cross, Institute for Transfusion Medicine and Immunohaematology, German Red Cross Baden-Wuertemberg-Hessen, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Diana Narita
- Institute for Laboratory Diagnostics and Transfusion Medicine, Donauisarklinikum, Deggendorf, Germany
| | - Ansgar Raadts
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Jena, Jena, Germany
| | - Klaus Schwendner
- Department of Anaesthesiology and Operative Intensive Care Medicine, Diakonie Hospital Martha-Maria, Nuremberg, Germany
| | - Erhard Seifried
- German Red Cross, Institute for Transfusion Medicine and Immunohaematology, German Red Cross Baden-Wuertemberg-Hessen, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Patrick Stark
- Department of Visceral and Vascular Surgery, Klinikum Mittelmosel, Zell, Germany
| | - Josef Thoma
- Department of Anaesthesiology and Operative Intensive Care Medicine, Ortenauklinikum, Gengenbach, Germany
| | - Henry Weigt
- Department of Anaesthesiology, SLK-Kliniken, Heilbronn, Germany
| | - Christoph Wiesenack
- Department of Anaesthesiology, Evangelisches Diakoniekrankenhaus Freiburg, Freiburg, Germany
| | - Andrea Ulrike Steinbicker
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany.,Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Kai Zacharowski
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Patrick Meybohm
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany.,Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
| | | |
Collapse
|
24
|
Kathamuthu GR, Rajamanickam A, Sridhar R, Baskaran D, Babu S. Strongyloidiasis stercoralis coinfection is associated with altered iron status biomarkers in tuberculous lymphadenitis. Front Immunol 2022; 13:999614. [PMID: 36341407 PMCID: PMC9632344 DOI: 10.3389/fimmu.2022.999614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/07/2022] [Indexed: 11/26/2022] Open
Abstract
Soil-transmitted helminth [mainly Strongyloidiasis stercoralis (Ss)] and tuberculous lymphadenitis (TBL) coinfection in humans is a significant public health problem. We have previously shown that TBL+Ss+ coinfection significantly alters diverse cytokine, matrix metalloproteinase, and tissue inhibitors of metalloproteinase profiles. However, no data is available to understand the influence of Ss coinfection in TBL disease with respect to iron status biomarkers. Hence, we have studied the effect of Ss coinfection on the circulating levels of iron status (ferritin, transferrin [TF], apotransferrin [ApoT], hepcidin, hemopexin) biomarkers in TBL disease. Our results show that TBL+Ss+ and/or TBL+Ss- individuals are associated with significantly altered biochemical and hematological (red blood cell (RBC) counts, hemoglobin (Hb), hematocrit (HCT), mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH) were decreased, and platelets were increased) parameters compared to TBL-Ss+ individuals. Our results also show that TBL+Ss+ coinfection is associated with diminished circulating levels of ferritin, ApoT, hepcidin, and hemopexin compared to TBL+Ss- individuals. TBL+Ss+ and TBL+Ss- groups are associated with altered iron status biomarkers (decreased ferritin [TBL+Ss+ alone] and increased TF, ApoT, hepcidin and hemopexin [TBL+Ss- alone]) compared to TBL-Ss+ group. The heat map expression profile and principal component analysis (PCA) analysis of iron status biomarkers were significantly altered in TBL+Ss+ compared to TBL+Ss- and/or TBL-Ss+ individuals. A significant correlation (positive/negative) was obtained among the biochemical and hematological parameters (white blood cells (WBC)/ferritin, TF, and hepcidin, mean corpuscular hemoglobin concentration (MCHC)/ferritin and hemopexin) with iron status biomarkers. Finally, receiver operating characteristic (ROC) analysis revealed that hemopexin was significantly associated with greater specificity and sensitivity in discriminating TBL+Ss+ and TBL+Ss- coinfected individuals. Thus, our data conclude that Ss coinfection is associated with altered iron status biomarkers indicating that coinfection might alter the host-Mtb interface and could influence the disease pathogenesis.
Collapse
Affiliation(s)
- Gokul Raj Kathamuthu
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India
- Indian Council of Medical Research-National Institute for Research in Tuberculosis (ICMR-NIRT), Chennai, India
- *Correspondence: Gokul Raj Kathamuthu,
| | - Anuradha Rajamanickam
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India
| | | | - Dhanaraj Baskaran
- Indian Council of Medical Research-National Institute for Research in Tuberculosis (ICMR-NIRT), Chennai, India
| | - Subash Babu
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
25
|
Zhou Z, Wu J, Yang Y, Gao P, Wang L, Wu Z. Hepcidin as a prognostic biomarker in clear cell renal cell carcinoma. Am J Cancer Res 2022; 12:4120-4139. [PMID: 36225649 PMCID: PMC9548002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/22/2022] [Indexed: 06/16/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common malignancy of urologic neoplasms. Hepcidin is a pivotal modulator of iron metabolism involved in human cancers; however, the biological significance of hepcidin in ccRCC remains to be fully understood. Therefore, in this study, we evaluated the expression profiles of hepcidin in ccRCC from several public databases and found that hepcidin expression was upregulated in ccRCC, which was further validated in ccRCC cell lines, clinical samples, and tissue microarray (TMA) quantitative real-time PCR and immunohistochemistry. In addition, we found that the expression level of hepcidin was correlated with the age, T stage and pathologic stage of patients. Furthermore, hepcidin promoter methylation was significantly associated with the worse poor clinical parameters of ccRCC patients, and hepcidin was an independent prognostic factor. Mechanistically, enrichment analysis revealed that hepcidin participated in the immune-related and metabolism-related pathways. Hepcidin was positively correlated with not only immune infiltration and immune checkpoints but also tumor mutation burden and cytotoxic T lymphocyte. Finally, we validated the positive correlation of hepcidin with the marker of macrophage (CD68) in the TMA. Our findings provide insights into understanding the function and its underlying mechanism of hepcidin in ccRCC and suggest that hepcidin might serve as a potential predictive biomarker of response to immunotherapy and the prognosis of patients with ccRCC.
Collapse
Affiliation(s)
- Zijian Zhou
- Department of Urology, Huashan Hospital, Fudan UniversityShanghai 200040, PR China
- Institute of Urology, Fudan UniversityShanghai 200040, PR China
| | - Jiajin Wu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical UniversityNanjing 210029, PR China
| | - Yuanyuan Yang
- Department of Urology, Huashan Hospital, Fudan UniversityShanghai 200040, PR China
- Institute of Urology, Fudan UniversityShanghai 200040, PR China
| | - Peng Gao
- Department of Urology, Huashan Hospital, Fudan UniversityShanghai 200040, PR China
- Institute of Urology, Fudan UniversityShanghai 200040, PR China
| | - Lujia Wang
- Department of Urology, Huashan Hospital, Fudan UniversityShanghai 200040, PR China
- Institute of Urology, Fudan UniversityShanghai 200040, PR China
| | - Zhong Wu
- Department of Urology, Huashan Hospital, Fudan UniversityShanghai 200040, PR China
- Institute of Urology, Fudan UniversityShanghai 200040, PR China
| |
Collapse
|
26
|
Balusikova K, Dostalikova-Cimburova M, Tacheci I, Kovar J. Expression profiles of iron transport molecules along the duodenum. J Cell Mol Med 2022; 26:2995-3004. [PMID: 35445529 PMCID: PMC9097835 DOI: 10.1111/jcmm.17313] [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: 12/16/2021] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 11/30/2022] Open
Abstract
Duodenal biopsies are considered a suitable source of enterocytes for studies of dietary iron absorption. However, the expression level of molecules involved in iron absorption may vary along the length of duodenum. We aimed to determine whether the expression of molecules involved in the absorption of heme and non-heme iron differs depending on the location in the duodenum. Analysis was performed with samples of duodenal biopsies from 10 individuals with normal iron metabolism. Samples were collected at the following locations: (a) immediately post-bulbar, (b) 1-2 cm below the papilla of Vater and (c) in the distal duodenum. The gene expression was analyzed at the mRNA and protein level using real-time PCR and Western blot analysis. At the mRNA level, significantly different expression of HCP1, DMT1, ferroportin and Zip8 was found at individual positions of duodenum. Position-dependent expression of other molecules, especially of FLVCR1, HMOX1 and HMOX2 was also detected but with no statistical significances. At the protein level, we observed statistically significantly decreasing expression of transporters HCP1, FLVCR1, DMT1, ferroportin, Zip14 and Zip8 with advancing positions of duodenum. Our results are consistent with a gradient of diminishing iron absorption along the duodenum for both heme and non-heme iron.
Collapse
Affiliation(s)
- Kamila Balusikova
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marketa Dostalikova-Cimburova
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ilja Tacheci
- 2nd Department of Internal Medicine - Gastroenterology, University Hospital and Charles University in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jan Kovar
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
27
|
Pecorelli A, Franceschi P, Braccischi L, Izzo F, Renzulli M, Golfieri R. MRI Appearance of Focal Lesions in Liver Iron Overload. Diagnostics (Basel) 2022; 12:diagnostics12040891. [PMID: 35453939 PMCID: PMC9029711 DOI: 10.3390/diagnostics12040891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 11/16/2022] Open
Abstract
Liver iron overload is defined as an accumulation of the chemical element Fe in the hepatic parenchyma that exceeds the normal storage. When iron accumulates, it can be toxic for the liver by producing inflammation and cell damage. This can potentially lead to cirrhosis and hepatocellular carcinoma, as well as to other liver lesions depending on the underlying condition associated to liver iron overload. The correct assessment of liver iron storage is pivotal to drive the best treatment and prevent complication. Nowadays, magnetic resonance imaging (MRI) is the best non-invasive modality to detect and quantify liver iron overload. However, due to its superparamagnetic properties, iron provides a natural source of contrast enhancement that can make challenging the differential diagnosis between different focal liver lesions (FLLs). To date, a fully comprehensive description of MRI features of liver lesions commonly found in iron-overloaded liver is lacking in the literature. Through an extensive review of the published literature, we aim to summarize the MRI signal intensity and enhancement pattern of the most common FLLs that can occur in liver iron overload.
Collapse
|
28
|
Li LX, Guo FF, Liu H, Zeng T. Iron overload in alcoholic liver disease: underlying mechanisms, detrimental effects, and potential therapeutic targets. Cell Mol Life Sci 2022; 79:201. [PMID: 35325321 PMCID: PMC11071846 DOI: 10.1007/s00018-022-04239-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023]
Abstract
Alcoholic liver disease (ALD) is a global public health challenge due to the high incidence and lack of effective therapeutics. Evidence from animal studies and ALD patients has demonstrated that iron overload is a hallmark of ALD. Ethanol exposure can promote iron absorption by downregulating the hepcidin expression, which is probably mediated by inducing oxidative stress and promoting erythropoietin (EPO) production. In addition, ethanol may enhance iron uptake in hepatocytes by upregulating the expression of transferrin receptor (TfR). Iron overload in the liver can aggravate ethanol-elicited liver damage by potentiating oxidative stress via Fenton reaction, promoting activation of Kupffer cells (KCs) and hepatic stellate cells (HSCs), and inducing a recently discovered programmed iron-dependent cell death, ferroptosis. This article reviews the current knowledge of iron metabolism, regulators of iron homeostasis, the mechanism of ethanol-induced iron overload, detrimental effects of iron overload in the liver, and potential therapeutic targets.
Collapse
Affiliation(s)
- Long-Xia Li
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Fang-Fang Guo
- Department of Pharmacy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Hong Liu
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
29
|
Zhang X, Song T, Zhao M, Tao X, Zhang B, Sun C, Wang P, Wang K, Zhao L. Sirtuin 2 Alleviates Chronic Neuropathic Pain by Suppressing Ferroptosis in Rats. Front Pharmacol 2022; 13:827016. [PMID: 35401208 PMCID: PMC8984153 DOI: 10.3389/fphar.2022.827016] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Neuropathic pain (NP) is chronic and associated with poor effects of general analgesia. It affects patients’ health and quality of life. The apoptotic process of lipid peroxidation caused by iron overload is called ferroptosis, which may be associated with nervous system disease. A recent study has found that sirtuin 2 (SIRT2) achieves a neuroprotective effect by suppressing ferroptosis. Herein, we aimed to examine whether SIRT2 regulated spared nerve injury (SNI)-induced NP by suppressing ferroptosis in rats. A rat model of NP was induced in adult male Sprague-Dawley rats weighing 200–250 g. Mechanical allodynia was observed from the first day after SNI and continued for 14 days. Compared with age-matched control rats, the expression of SIRT2 and ferroportin 1 (FPN1) decreased in the L4-6 spinal cord of the SNI-induced NP rats. In addition, we observed that the levels of both iron and anti-acyl-coenzyme A synthetase long-chain family member 4 (ACSL4) were significantly increased in the spinal cord after SNI, while the expression of glutathione peroxidase 4 (GPX4) was decreased. Furthermore, an intrathecal injection of SIRT2 overexpressed recombinant adenovirus, which upregulated the expression of SIRT2, attenuated mechanical allodynia, enhanced the level of FPN1, inhibited intracellular iron accumulation, and reduced oxidant stress levels, thereby reversing the changes to ACSL4 and GPX4 expression in the SNI rats. This evidence suggests that SIRT2-targeted therapeutics may help relieve the symptoms of chronic NP.
Collapse
|
30
|
Al-Daghri NM, Yakout S, Ghaleb A, Hussain SD, Sabico S. Iron and 25-hydroxyvitamin D in postmenopausal women with osteoporosis. Am J Transl Res 2022; 14:1387-1405. [PMID: 35422903 PMCID: PMC8991132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/22/2022] [Indexed: 06/14/2023]
Abstract
Iron and vitamin D deficiencies are some of the most common health problems in the world. Iron is essential in oxygen transport and participates in many enzymatic systems in the body, with important roles in vitamin D metabolism. Osteoporosis is one of the most prevalent chronic disease of the elderly in the world as well as in the Saudi population. The relationship between iron, vitamin D deficiency and bone health comes from clinical observations in iron overload patients who suffered bone loss. The opposite scenario, whether iron and vitamin D deficiencies affect bone metabolism, has not been fully addressed. This is of great interest, as this nutrient deficiency is a worldwide public health problem and at the same time osteoporosis and bone alterations are highly prevalent. The relationship between 25(OH)D and iron deficiencies with osteoporosis is unknown up to date. This review presents the current knowledge on nutritional iron and vitamin D deficiencies in bone remodeling, and discuss the link between iron and bone metabolism among postmenopausal women. Finally, it is hypothesized that chronic iron and vitamin D deficiencies induces bone resorption and risk of osteoporosis, thus complete recovery from anemia and its prevention should be promoted in order to improve quality of life including bone health. Several mechanisms are implicated; hence, further investigation on the possible impact of iron and vitamin D deficiencies on the development of osteoporosis is needed.
Collapse
Affiliation(s)
- Nasser M Al-Daghri
- Chair for Biomarkers of Chronic Diseases, Biochemistry Department, College of Science, King Saud University Riyadh 11451, Saudi Arabia
| | - Sobhy Yakout
- Chair for Biomarkers of Chronic Diseases, Biochemistry Department, College of Science, King Saud University Riyadh 11451, Saudi Arabia
| | - Afnan Ghaleb
- Chair for Biomarkers of Chronic Diseases, Biochemistry Department, College of Science, King Saud University Riyadh 11451, Saudi Arabia
| | - Syed Danish Hussain
- Chair for Biomarkers of Chronic Diseases, Biochemistry Department, College of Science, King Saud University Riyadh 11451, Saudi Arabia
| | - Shaun Sabico
- Chair for Biomarkers of Chronic Diseases, Biochemistry Department, College of Science, King Saud University Riyadh 11451, Saudi Arabia
| |
Collapse
|
31
|
Polak KZ, Schaffer P, Donaghy D, Zenk MC, Olver CS. Iron, hepcidin, and microcytosis in canine hepatocellular carcinoma. Vet Clin Pathol 2022; 51:208-215. [PMID: 35274348 DOI: 10.1111/vcp.13085] [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: 05/06/2021] [Revised: 10/01/2021] [Accepted: 10/14/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Erythrocyte microcytosis in some dogs with hepatocellular carcinoma (HCC) suggests a derangement in systemic iron. Hepcidin, the master regulator of iron, is secreted by the liver in response to interleukin 6 (IL-6) and/or bone morphogenetic protein 6 (BMP6) and can cause microcytosis. OBJECTIVES Pilot study to compare the quantities of hepcidin, IL-6, and BMP6 RNA molecules in archival tumoral (HCC) and adjacent peritumoral (non-HCC) hepatic tissue to determine if they are different between tissue types or associated with microcytosis. METHODS RNA was isolated from formalin-fixed, paraffin-embedded HCC and non-HCC tissue from seven microcytic dogs and four normocytic dogs. Digital RNA counts of hepcidin, IL-6, or BMP6, and six other iron-regulatory genes were determined using the Nanostring nCounter system. The area of blue on each section was digitally evaluated to measure the extent of Prussian blue staining objectively. Parameters were compared between HCC and non-HCC tissue and between microcytic and normocytic groups. RESULTS Hepcidin was decreased, and transferrin receptor 1 (TfR1) was increased in HCC tissue compared with non-HCC tissue. Non-HCC hepcidin RNA counts correlated negatively with MCV and positively with the extent of iron staining. Hepcidin expression was higher in non-HCC tissue of microcytic cases than in normocytic cases. CONCLUSIONS Canine HCC cases showed relatively increased iron staining in non-HCC tissue and decreased hepcidin RNA in HCC tissue. Microcytic cases had higher hepcidin RNA in non-HCC tissue than normocytic cases. Future studies may extend these findings to protein quantification, cellular localization of RNA changes, and determining if iron loading in canine liver is a predisposing factor for HCC.
Collapse
Affiliation(s)
- Klaudia Z Polak
- College of Veterinary Medicine and Biomedical Sciences, Clinical Pathology Section, Colorado State University, Fort Collins, Colorado, USA
| | - Paula Schaffer
- College of Veterinary Medicine and Biomedical Sciences, Clinical Pathology Section, Colorado State University, Fort Collins, Colorado, USA
| | - Dillon Donaghy
- College of Veterinary Medicine and Biomedical Sciences, Clinical Pathology Section, Colorado State University, Fort Collins, Colorado, USA
| | - Madison C Zenk
- College of Veterinary Medicine and Biomedical Sciences, Clinical Pathology Section, Colorado State University, Fort Collins, Colorado, USA
| | - Christine S Olver
- College of Veterinary Medicine and Biomedical Sciences, Clinical Pathology Section, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
32
|
Diaz S, Polania J, Ariza-Suarez D, Cajiao C, Grajales M, Raatz B, Beebe SE. Genetic Correlation Between Fe and Zn Biofortification and Yield Components in a Common Bean ( Phaseolus vulgaris L.). FRONTIERS IN PLANT SCIENCE 2022; 12:739033. [PMID: 35046970 PMCID: PMC8761845 DOI: 10.3389/fpls.2021.739033] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/08/2021] [Indexed: 05/05/2023]
Abstract
Common bean (Phaseolus vulgaris L.) is the most important legume for direct human consumption worldwide. It is a rich and relatively inexpensive source of proteins and micronutrients, especially iron and zinc. Bean is a target for biofortification to develop new cultivars with high Fe/Zn levels that help to ameliorate malnutrition mainly in developing countries. A strong negative phenotypic correlation between Fe/Zn concentration and yield is usually reported, posing a significant challenge for breeders. The objective of this study was to investigate the genetic relationship between Fe/Zn. We used Quantitative Trait Loci (QTLs) mapping and Genome-Wide Association Studies (GWAS) analysis in three bi-parental populations that included biofortified parents, identifying genomic regions associated with yield and micromineral accumulation. Significant negative correlations were observed between agronomic traits (pod harvest index, PHI; pod number, PdN; seed number, SdN; 100 seed weight, 100SdW; and seed per pod, Sd/Pd) and micronutrient concentration traits (SdFe and SdZn), especially between pod harvest index (PHI) and SdFe and SdZn. PHI presented a higher correlation with SdN than PdN. Seventy-nine QTLs were identified for the three populations: 14 for SdFe, 12 for SdZn, 13 for PHI, 11 for SdN, 14 for PdN, 6 for 100SdW, and 9 for Sd/Pd. Twenty-three hotspot regions were identified in which several QTLs were co-located, of which 13 hotpots displayed QTL of opposite effect for yield components and Fe/Zn accumulation. In contrast, eight QTLs for SdFe and six QTLs for SdZn were observed that segregated independently of QTL of yield components. The selection of these QTLs will enable enhanced levels of Fe/Zn and will not affect the yield performance of new cultivars focused on biofortification.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Stephen E. Beebe
- Bean Program, Crops for Health and Nutrition Area, Alliance Bioversity International – CIAT, Cali, Colombia
| |
Collapse
|
33
|
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: 51] [Impact Index Per Article: 17.0] [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
|
34
|
Iron Deficiency Caused by Intestinal Iron Loss-Novel Candidate Genes for Severe Anemia. Genes (Basel) 2021; 12:genes12121869. [PMID: 34946818 PMCID: PMC8700796 DOI: 10.3390/genes12121869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 02/06/2023] Open
Abstract
The adult human body contains about 4 g of iron. About 1-2 mg of iron is absorbed every day, and in healthy individuals, the same amount is excreted. We describe a patient who presents with severe iron deficiency anemia with hemoglobin levels below 6 g/dL and ferritin levels below 30 ng/mL. Although red blood cell concentrates and intravenous iron have been substituted every month for years, body iron stores remain depleted. Diagnostics have included several esophago-gastro-duodenoscopies, colonoscopies, MRI of the liver, repetitive bone marrow biopsies, psychological analysis, application of radioactive iron to determine intact erythropoiesis, and measurement of iron excretion in urine and feces. Typically, gastrointestinal bleeding is a major cause of iron loss. Surprisingly, intestinal iron excretion in stool in the patient was repetitively increased, without gastrointestinal bleeding. Furthermore, whole exome sequencing was performed in the patient and additional family members to identify potential causative genetic variants that may cause intestinal iron loss. Under different inheritance models, several rare mutations were identified, two of which (in CISD1 and KRI1) are likely to be functionally relevant. Intestinal iron loss in the current form has not yet been described and is, with high probability, the cause of the severe iron deficiency anemia in this patient.
Collapse
|
35
|
Epigallocatechin-3-Gallate Suppresses BMP-6-Mediated SMAD1/5/8 Transactivation of Hepcidin Gene by Inducing SMILE in Hepatocytes. Antioxidants (Basel) 2021; 10:antiox10101590. [PMID: 34679725 PMCID: PMC8533173 DOI: 10.3390/antiox10101590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/27/2021] [Accepted: 10/09/2021] [Indexed: 01/01/2023] Open
Abstract
Hepcidin, a major regulator of systemic iron homeostasis, is mainly induced in hepatocytes by activating bone morphogenetic protein 6 (BMP-6) signaling in response to changes in the iron status. Small heterodimer partner-interacting leucine zipper protein (SMILE), a polyphenol-inducible transcriptional co-repressor, regulates hepatic gluconeogenesis and lipogenesis. Here, we examine the epigallocatechin-3-gallate (EGCG) effect on BMP-6-mediated SMAD1/5/8 transactivation of the hepcidin gene. EGCG treatment significantly decreased BMP-6-induced hepcidin gene expression and secretion in hepatocytes, which, in turn, abated ferroportin degradation. SMILE overexpression significantly decreased BMP receptor-induced hepcidin promoter activity. SMILE overexpression also significantly suppressed BMP-6-mediated induction of hepcidin mRNA and its secretion in HepG2 and AML12 cells. EGCG treatment inhibited BMP-6-mediated hepcidin gene expression and secretion, which were significantly reversed by SMILE knockdown in hepatocytes. Interestingly, SMILE physically interacted with SMAD1 in the nucleus and significantly blocked DNA binding of the SMAD complex to the BMP-response element on the hepcidin gene promoter. Taken together, these findings suggest that SMILE is a novel transcriptional repressor of BMP-6-mediated hepcidin gene expression, thus contributing to the control of iron homeostasis.
Collapse
|
36
|
Chang JE, Lee HM, Kim J, Rhew K. Prevalence of Anemia in Pediatric Patients According to Asthma Control: Propensity Score Analysis. J Asthma Allergy 2021; 14:743-751. [PMID: 34234469 PMCID: PMC8254559 DOI: 10.2147/jaa.s318641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/11/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose To investigate whether the degree of asthma control is associated with anemia in pediatric patients. Patients and Methods A cross-sectional study was performed using a dataset from the Health Insurance Reviews & Assessment Service (HIRA) of South Korea in 2016, which included children and adolescent patients diagnosed with asthma. Binary logistic regression was used to assess the association between asthma control and the prevalence of anemia. Results A total of 236,429 patients under 18 years old were included in the study, including 233,975 patients with controlled and 2454 with uncontrolled asthma. Binary logistic regression after adjustment for confounding factors showed that patients with uncontrolled asthma had a 2.64-fold higher prevalence of anemia than those with well-controlled asthma (OR = 2.64, 95% CI: 2.16-3.22). While there was no effect of gender on the results, there was a statistically significant association between the prevalence of anemia and asthma control in patients under 13 years old. Conclusion These findings suggest that the prevalence of anemia is inversely correlated with asthma control in pediatric patients. Further studies are necessary to obtain pathophysiological insight into the relationship between severe inflammatory diseases and anemia.
Collapse
Affiliation(s)
- Ji-Eun Chang
- College of Pharmacy, Dongduk Women's University, Seoul, Republic of Korea
| | - Hyang-Mi Lee
- College of Pharmacy, Dongduk Women's University, Seoul, Republic of Korea
| | - Jongyoon Kim
- College of Pharmacy, Dongduk Women's University, Seoul, Republic of Korea
| | - Kiyon Rhew
- College of Pharmacy, Dongduk Women's University, Seoul, Republic of Korea
| |
Collapse
|
37
|
On Iron Metabolism and Its Regulation. Int J Mol Sci 2021; 22:ijms22094591. [PMID: 33925597 PMCID: PMC8123811 DOI: 10.3390/ijms22094591] [Citation(s) in RCA: 155] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Iron is a critical metal for several vital biological processes. Most of the body’s iron is bound to hemoglobin in erythrocytes. Iron from senescent red blood cells is recycled by macrophages in the spleen, liver and bone marrow. Dietary iron is taken up by the divalent metal transporter 1 (DMT1) in enterocytes and transported to portal blood via ferroportin (FPN), where it is bound to transferrin and taken up by hepatocytes, macrophages and bone marrow cells via transferrin receptor 1 (TfR1). While most of the physiologically active iron is bound hemoglobin, the major storage of most iron occurs in the liver in a ferritin-bound fashion. In response to an increased iron load, hepatocytes secrete the peptide hormone hepcidin, which binds to and induces internalization and degradation of the iron transporter FPN, thus controlling the amount of iron released from the cells into the blood. This review summarizes the key mechanisms and players involved in cellular and systemic iron regulation.
Collapse
|
38
|
Abstract
Despite increasing use of targeted therapies to treat cancer, anemia remains a common complication of cancer therapy. Physician concerns about the safety of intravenous (IV) iron products and erythropoiesis-stimulating agents (ESAs) have resulted in many patients with cancer receiving no or suboptimal anemia therapy. In this article, we present 4 patient cases that illustrate both common and complex clinical scenarios. We first present a review of erythropoiesis and then describe our approach to cancer-associated anemia by identifying the contributing causes before selecting specific treatments. We summarize clinical trial data affirming the safety and efficacy of currently available IV iron products used to treat cancer-associated anemia and illustrate how we use commonly available laboratory tests to assess iron status during routine patient management. We compare adverse event rates associated with IV iron vs red cell transfusion and discuss using first-line IV iron monotherapy to treat anemic patients with cancer, which decreases the need for ESAs. A possible mechanism behind ESA-induced tumor progression is discussed. Finally, we review the potential of novel therapies such as ascorbic acid, prolyl hydroxylase inhibitors, activin traps, hepcidin, and bone morphogenetic protein antagonists in treating cancer-associated anemia.
Collapse
|
39
|
Liu L, Zhang S, Zheng X, Li H, Chen Q, Qin K, Ding Y, Wei Y. Carbon dots derived from Fusobacterium nucleatum for intracellular determination of Fe 3+ and bioimaging both in vitro and in vivo. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2021; 13:1121-1131. [PMID: 33591301 DOI: 10.1039/d1ay00020a] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Intracellular Fe3+ amount is one of the critical determinants of human health. The development of simple and effective probes for the quantitative detection of Fe3+in vivo is of great significance for the early diagnosis of disease or disorder associated with iron deficiency or overload. In this study, remarkable carbon dots, which can serve as a biosensor for efficient intracellular Fe3+ detection, were synthesized by hydrothermal carbonization of Fusobacterium nucleatum, an anaerobic bacterium. The achieved F. nucleatum-carbon dots (Fn-CDs) possessed the features of strong fluorescence, high stability and excellent biocompatibility. The obtained Fn-CDs could easily internalize into both plant cells and human cells with excellent ability for cell tracking and biomedical labeling. The fluorescence of Fn-CDs could still remain for another 24 hours after penetrating into cells. Furthermore, the fluorescent Fn-CDs were very sensitive to the presence of Fe3+ ions even in cells, exhibiting great promising applications in in vivo detection of Fe3+ ions. In addition, the Fn-CDs posed no harm to the mice, being circulated and excreted within a short time, making the Fn-CDs an excellent candidate for bioimaging and biosensing in vivo.
Collapse
Affiliation(s)
- Lijuan Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China.
| | - Shengting Zhang
- Department of Education, Yunnan Minzu University, Kunming 650500, China
| | - Xiaodan Zheng
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China.
| | - Hongmei Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Qi Chen
- Department of Hematology, The Second People's Hospital of Yunnan Province, The Affiliated Hospital of Yunnan University, Kunming, 650021, China
| | - Kunhao Qin
- Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Yafang Ding
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China.
| | - Yunlin Wei
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China.
| |
Collapse
|
40
|
Traeger L, Schnittker J, Dogan DY, Oguama D, Kuhlmann T, Muckenthaler MU, Krijt J, Urzica EI, Steinbicker AU. HFE and ALK3 act in the same signaling pathway. Free Radic Biol Med 2020; 160:501-505. [PMID: 32861780 DOI: 10.1016/j.freeradbiomed.2020.08.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/21/2020] [Indexed: 12/27/2022]
Abstract
Hepcidin deficiency leads to iron overload by increased dietary iron uptake and iron release from storage cells. The most frequent mutation in Hfe leads to reduced hepcidin expression and thereby causes iron overload. Recent findings suggested that HFE activates hepcidin expression predominantly via the BMP type I receptor ALK3. Here, we investigated whether HFE exclusively utilizes ALK3 or other signaling mechanisms also. We generated mice with double deficiency of Hfe and hepatocyte-specific Alk3 and compared the iron overload phenotypes of these double knockout mice to single hepatocyte-specific Alk3 deficient or Hfe knockout mice. Double Hfe-/-/hepatic Alk3fl/fl;Alb-Cre knockouts develop a similar iron overload phenotype compared to single hepatocyte-specific Alk3 deficient mice hallmarked by serum iron levels, tissue iron content and hepcidin levels of similar grades. HFE protein levels were increased in Alk3fl/fl;Alb-Cre mice compared to Alk3fl/fl mice, which was caused by iron overload - and not by Alk3 deficiency. The data provide evidence by genetic means that 1. HFE exclusively uses the BMP type I receptor ALK3 to induce hepcidin expression and 2. HFE protein expression is induced by iron overload, which further emphasizes the iron sensing function of HFE.
Collapse
Affiliation(s)
- L Traeger
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany.
| | - J Schnittker
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany.
| | - D Y Dogan
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany.
| | - D Oguama
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany.
| | - T Kuhlmann
- Institute of Neuropathology, University Hospital Muenster, University of Muenster, Muenster, Germany.
| | - M U Muckenthaler
- Department of Pediatric Oncology, Hematology and Immunology, Molecular Medicine Partnership (MMPU), European Molecular Biology Laboratory (EMBL), University of Heidelberg, Heidelberg, Germany.
| | - J Krijt
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - E I Urzica
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany.
| | - A U Steinbicker
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany.
| |
Collapse
|
41
|
Caproni L, Raggi L, Talsma EF, Wenzl P, Negri V. European landrace diversity for common bean biofortification: a genome-wide association study. Sci Rep 2020; 10:19775. [PMID: 33188249 PMCID: PMC7666124 DOI: 10.1038/s41598-020-76417-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/21/2020] [Indexed: 12/28/2022] Open
Abstract
Mineral deficiencies represent a global challenge that needs to be urgently addressed. An adequate intake of iron and zinc results in a balanced diet that reduces chances of impairment of many metabolic processes that can lead to clinical consequences. In plants, bioavailability of such nutrients is reduced by presence of compounds such as phytic acid, that can chelate minerals and reduce their absorption. Biofortification of common bean (Phaseolus vulgaris L.) represents an important strategy to reduce mineral deficiencies, especially in areas of the world where this crop plays a key role in the diet. In this study, a panel of diversity encompassing 192 homozygous genotypes, was screened for iron, zinc and phytate seed content. Results indicate a broad variation of these traits and allowed the identification of accessions reasonably carrying favourable trait combinations. A significant association between zinc seed content and some molecular SNP markers co-located on the common bean Pv01 chromosome was detected by means of genome-wide association analysis. The gene Phvul001G233500, encoding for an E3 ubiquitin-protein ligase, is proposed to explain detected associations. This result represents a preliminary evidence that can foster future research aiming at understanding the genetic mechanisms behind zinc accumulation in beans.
Collapse
Affiliation(s)
- Leonardo Caproni
- Dipartimento di Scienze Agrarie, Alimentari e Ambientali (DSA3), Università Degli Studi Di Perugia, Borgo XX Giugno 74, 06126, Perugia, Italy
| | - Lorenzo Raggi
- Dipartimento di Scienze Agrarie, Alimentari e Ambientali (DSA3), Università Degli Studi Di Perugia, Borgo XX Giugno 74, 06126, Perugia, Italy
| | - Elise F Talsma
- Division of Human Nutrition and Health, Wageningen University and Research, PO Box 17, 6700 AA, Wageningen, The Netherlands
- HarvestPlus, International Center for Tropical Agriculture (CIAT), Km 17 Recta Cali-Palmira, Cali, Colombia
| | - Peter Wenzl
- Genetic Resources Program, International Center for Tropical Agriculture (CIAT), Km 17 Recta Cali-Palmira, Cali, Colombia
| | - Valeria Negri
- Dipartimento di Scienze Agrarie, Alimentari e Ambientali (DSA3), Università Degli Studi Di Perugia, Borgo XX Giugno 74, 06126, Perugia, Italy.
| |
Collapse
|
42
|
Abstract
Iron overload is a common clinical problem resulting from hereditary hemochromatosis or secondary hemosiderosis (mainly associated with transfusion therapy), being also associated with chronic liver diseases and metabolic disorders. Excess of iron accumulates in organs like the liver, pancreas and heart. Without treatment, patients with iron overload disorders will develop liver cirrhosis, diabetes and cardiomyopathy. Iron quantification is therefore crucial not only for diagnosis of iron overload but also to monitor iron-reducing therapies. Liver iron concentration is considered the surrogate marker of total body iron stores. Because liver biopsy is invasive and prone to high variability and sampling bias, MR imaging has emerged as a non-invasive method and gained wide acceptance, now being considered the standard of care for assessing iron overload. Nevertheless, there are different MR techniques for iron quantification and there is still no consensus about the best technique or postprocessing tool for hepatic iron quantification, with the choice of imaging technique depending mainly on the local expertise as well on the available equipment and software. Because different methods should not be used interchangeably, it is important to choose one method and use the same one when following up patients over time.
Collapse
Affiliation(s)
- Manuela França
- Radiology Department - Centro Hospitalar Universitário do Porto, Largo Prof Abel Salazar, 4099-001, Porto, Portugal.
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, I3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal.
| | - João Gomes Carvalho
- Radiology Department - Centro Hospitalar Universitário do Porto, Largo Prof Abel Salazar, 4099-001, Porto, Portugal
| |
Collapse
|
43
|
Ruetten M, Steinmetz HW, Thiersch M, Kik M, Vaughan L, Altamura S, Muckenthaler MU, Gassmann M. Iron Regulation in Elderly Asian Elephants ( Elephas maximus) Chronically Infected With Mycobacterium tuberculosis. Front Vet Sci 2020; 7:596379. [PMID: 33195633 PMCID: PMC7661576 DOI: 10.3389/fvets.2020.596379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 09/30/2020] [Indexed: 11/13/2022] Open
Abstract
Restriction of nutrients to pathogens (nutritional immunity) is a critical innate immune response mechanism that operates when pathogens such as Mycobacterium tuberculosis have the potential to evade humoral immunity. Tuberculosis is of growing concern for zoological collections worldwide and is well-illustrated by infections of Asian and African elephants, where tuberculosis is difficult to diagnose. Here, we investigated hematological parameters and iron deposition in liver, lung, and spleen of three Asian elephants (Elephas maximus) infected with Mycobacterium tuberculosis. For reference purposes, we analyzed tissue samples from control M. tuberculosis-negative elephants with and without evidence of inflammation and/or chronic disease. Molecular analyses of bacterial lesions of post mortally collected tissues confirmed M. tuberculosis infection in three elephants. DNA sequencing of the bacterial cultures demonstrated a single source of infection, most likely of human origin. In these elephants, we observed moderate microcytic anemia as well as liver (mild), lung (moderate) and spleen (severe) iron accumulation, the latter mainly occurring in macrophages. Macrophage iron sequestration in response to infection and inflammation is caused by inhibition of iron export via hepcidin-dependent and independent mechanisms. The hepatic mRNA levels of the iron-regulating hormone hepcidin were increased in only one control elephant suffering from chronic inflammation without mycobacterial infection. By contrast, all three tuberculosis-infected elephants showed low hepcidin mRNA levels in the liver and low serum hepcidin concentrations. In addition, hepatic ferroportin mRNA expression was high. This suggests that the hepcidin/ferroportin regulatory system aims to counteract iron restriction in splenic macrophages in M. tuberculosis infected elephants to provide iron for erythropoiesis and to limit iron availability for a pathogen that predominantly proliferates in macrophages. Tuberculosis infections appear to have lingered for more than 30 years in the three infected elephants, and decreased iron availability for mycobacterial proliferation may have forced the bacteria into a persistent, non-proliferative state. As a result, therapeutic iron substitution may not have been beneficial in these elephants, as this therapy may have enhanced progression of the infection.
Collapse
Affiliation(s)
- Maja Ruetten
- PathoVet AG, Pathology Diagnostic Laboratory, Lindau, Switzerland
| | | | - Markus Thiersch
- Institute of Veterinary Physiology, and Center for Clinical Studies, Vetsuisse Faculty Zurich, and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Marja Kik
- Pathology Division, Department of Biomedical Health Sciences, Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Lloyd Vaughan
- PathoVet AG, Pathology Diagnostic Laboratory, Lindau, Switzerland
| | - Sandro Altamura
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Children's Hospital, Heidelberg University Medical Center, Heidelberg, Germany
| | - Martina U Muckenthaler
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Children's Hospital, Heidelberg University Medical Center, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany.,Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany
| | - Max Gassmann
- Institute of Veterinary Physiology, and Center for Clinical Studies, Vetsuisse Faculty Zurich, and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,Universidad Peruana Cayetano Heredia (UPCH), Lima, Peru
| |
Collapse
|
44
|
Brandtner A, Tymoszuk P, Nairz M, Lehner GF, Fritsche G, Vales A, Falkner A, Schennach H, Theurl I, Joannidis M, Weiss G, Pfeifhofer-Obermair C. Linkage of alterations in systemic iron homeostasis to patients' outcome in sepsis: a prospective study. J Intensive Care 2020; 8:76. [PMID: 33014378 PMCID: PMC7528491 DOI: 10.1186/s40560-020-00495-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/22/2020] [Indexed: 12/16/2022] Open
Abstract
Background Sepsis, a dysregulated host response following infection, is associated with massive immune activation and high mortality rates. There is still a need to define further risk factors and laboratory parameters predicting the clinical course. Iron metabolism is regulated by both, the body’s iron status and the immune response. Iron itself is required for erythropoiesis but also for many cellular and metabolic functions. Moreover, iron availability is a critical determinant in infections because it is an essential nutrient for most microbes but also impacts on immune function and intravascular oxidative stress. Herein, we used a prospective study design to investigate the putative impact of serum iron parameters on the outcome of sepsis. Methods Serum markers of iron metabolism were measured in a prospective cohort of 61 patients (37 males, 24 females) with sepsis defined by Sepsis-3 criteria in a medical intensive care unit (ICU) and compared between survivors and non-survivors. Regulation of iron parameters in patients stratified by focus of infection and co-medication as well as association of the markers with sepsis severity scores and survival were investigated with linear and logistic regression corrected for sex and age effects. Results Positive correlations of increased serum iron and ferritin concentrations upon ICU admission with the severity of organ failure (SOFA score) and with mortality were observed. Moreover, high TF-Sat, elevated ferritin and serum iron levels and low transferrin concentrations were associated with reduced survival. A logistic regression model consisting of SOFA and transferrin saturation (SOFA–TF-Sat) had the best predictive power for survival in septic ICU patients. Of note, administration of blood transfusions prior to ICU admission resulted in increased TF-Sat and reduced survival of septic patients. Conclusions Our study could show an important impact of serum iron parameters on the outcome of sepsis. Furthermore, we identified transferrin saturation as a stand-alone predictor of sepsis survival and as a parameter of iron metabolism which may in a combined model improve the prediction power of the SOFA score. Trial registration The study was carried out in accordance with the recommendations of the Declaration of Helsinki on biomedical research. The study was approved by the institutional ethics review board of the Medical University Innsbruck (study AN2013-0006).
Collapse
Affiliation(s)
- Anna Brandtner
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine I, Medical University of Innsbruck, Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstr. 35, Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstr. 35, Innsbruck, Austria
| | - Georg F Lehner
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine I, Medical University of Innsbruck, Innsbruck, Austria
| | - Gernot Fritsche
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstr. 35, Innsbruck, Austria
| | - Anja Vales
- Central Institute for Blood Transfusion and Immunology, Innsbruck, Austria
| | - Andreas Falkner
- Central Institute for Blood Transfusion and Immunology, Innsbruck, Austria
| | - Harald Schennach
- Central Institute for Blood Transfusion and Immunology, Innsbruck, Austria
| | - Igor Theurl
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstr. 35, Innsbruck, Austria
| | - Michael Joannidis
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine I, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Anichstr. 35, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
45
|
Kaufner L, von Heymann C, Henkelmann A, Pace NL, Weibel S, Kranke P, Meerpohl JJ, Gill R. Erythropoietin plus iron versus control treatment including placebo or iron for preoperative anaemic adults undergoing non-cardiac surgery. Cochrane Database Syst Rev 2020; 8:CD012451. [PMID: 32790892 PMCID: PMC8095002 DOI: 10.1002/14651858.cd012451.pub2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Approximately 30% of adults undergoing non-cardiac surgery suffer from preoperative anaemia. Preoperative anaemia is a risk factor for mortality and adverse outcomes in different surgical specialties and is frequently the reason for blood transfusion. The most common causes are renal, chronic diseases, and iron deficiency. International guidelines recommend that the cause of anaemia guide preoperative anaemia treatment. Recombinant human erythropoietin (rHuEPO) with iron supplementation has frequently been used to increase preoperative haemoglobin concentrations in patients in order to avoid the need for perioperative allogeneic red blood cell (RBC) transfusion. OBJECTIVES To evaluate the efficacy of preoperative rHuEPO therapy (subcutaneous or parenteral) with iron (enteral or parenteral) in reducing the need for allogeneic RBC transfusions in preoperatively anaemic adults undergoing non-cardiac surgery. SEARCH METHODS We searched CENTRAL, Ovid MEDLINE(R), Ovid Embase, ISI Web of Science: SCI-EXPANDED and CPCI-S, and clinical trial registries WHO ICTRP and ClinicalTrials.gov on 29 August 2019. SELECTION CRITERIA We included all randomized controlled trials (RCTs) that compared preoperative rHuEPO + iron therapy to control treatment (placebo, no treatment, or standard of care with or without iron) for preoperatively anaemic adults undergoing non-cardiac surgery. We used the World Health Organization (WHO) definition of anaemia: haemoglobin concentration (g/dL) less than 13 g/dL for males, and 12 g/dL for non-pregnant females (decision of inclusion based on mean haemoglobin concentration). We defined two subgroups of rHuEPO dosage: 'low' for 150 to 300 international units (IU)/kg body weight, and 'high' for 500 to 600 IU/kg body weight. DATA COLLECTION AND ANALYSIS Two review authors collected data from the included studies. Our primary outcome was the need for RBC transfusion (no autologous transfusion, fresh frozen plasma or platelets), measured in transfused participants during surgery (intraoperative) and up to five days after surgery. Secondary outcomes of interest were: haemoglobin concentration (directly before surgery), number of RBC units (where one unit contains 250 to 450 mL) transfused per participant (intraoperative and up to five days after surgery), mortality (within 30 days after surgery), length of hospital stay, and adverse events (e.g. renal dysfunction, thromboembolism, hypertension, allergic reaction, headache, fever, constipation). MAIN RESULTS Most of the included trials were in orthopaedic, gastrointestinal, and gynaecological surgery and included participants with mild and moderate preoperative anaemia (haemoglobin from 10 to 12 g/dL). The duration of preoperative rHuEPO treatment varied across the trials, ranging from once a week to daily or a 5-to-10-day period, and in one trial preoperative rHuEPO was given on the morning of surgery and for five days postoperatively. We included 12 trials (participants = 1880) in the quantitative analysis of the need for RBC transfusion following preoperative treatment with rHuEPO + iron to correct preoperative anaemia in non-cardiac surgery; two studies were multiarmed trials with two different dose regimens. Preoperative rHuEPO + iron given to anaemic adults reduced the need RBC transfusion (risk ratio (RR) 0.55, 95% confidence interval (CI) 0.38 to 0.80; participants = 1880; studies = 12; I2 = 84%; moderate-quality evidence due to inconsistency). This analysis suggests that on average, the combined administration of rHuEPO + iron will mean 231 fewer individuals will need transfusion for every 1000 individuals compared to the control group. Preoperative high-dose rHuEPO + iron given to anaemic adults increased the haemoglobin concentration (mean difference (MD) 1.87 g/dL, 95% CI 1.26 to 2.49; participants = 852; studies = 3; I2 = 89%; low-quality evidence due to inconsistency and risk of bias) but not low-dose rHuEPO + iron (MD 0.11 g/dL, 95% CI -0.46 to 0.69; participants = 334; studies = 4; I2 = 69%; low-quality evidence due to inconsistency and risk of bias). There was probably little or no difference in the number of RBC units when rHuEPO + iron was given preoperatively (MD -0.09, 95% CI -0.23 to 0.05; participants = 1420; studies = 6; I2 = 2%; moderate-quality evidence due to imprecision). There was probably little or no difference in the risk of mortality within 30 days of surgery (RR 1.19, 95% CI 0.39 to 3.63; participants = 230; studies = 2; I2 = 0%; moderate-quality evidence due to imprecision) or of adverse events including local rash, fever, constipation, or transient hypertension (RR 0.93, 95% CI 0.68 to 1.28; participants = 1722; studies = 10; I2 = 0%; moderate-quality evidence due to imprecision). The administration of rHuEPO + iron before non-cardiac surgery did not clearly reduce the length of hospital stay of preoperative anaemic adults (MD -1.07, 95% CI -4.12 to 1.98; participants = 293; studies = 3; I2 = 87%; low-quality evidence due to inconsistency and imprecision). AUTHORS' CONCLUSIONS Moderate-quality evidence suggests that preoperative rHuEPO + iron therapy for anaemic adults prior to non-cardiac surgery reduces the need for RBC transfusion and, when given at higher doses, increases the haemoglobin concentration preoperatively. The administration of rHuEPO + iron treatment did not decrease the mean number of units of RBC transfused per patient. There were no important differences in the risk of adverse events or mortality within 30 days, nor in length of hospital stay. Further, well-designed, adequately powered RCTs are required to estimate the impact of this combined treatment more precisely.
Collapse
Affiliation(s)
- Lutz Kaufner
- Department of Anaesthesiology and Intensive Care Medicine, Charité - University Medicine Berlin, Berlin, Germany
| | - Christian von Heymann
- Department of Anaesthesiology, Intensive Care Medicine, Emergency Care Medicine and Pain Therapy, Vivantes Klinikum im Friedrichshain, Berlin, Germany
| | - Anne Henkelmann
- Department of Anaesthesiology and Intensive Care Medicine, Charité - University Medicine Berlin, Berlin, Germany
| | - Nathan L Pace
- Department of Anesthesiology, University of Utah, Salt Lake City, UT, USA
| | - Stephanie Weibel
- Department of Anaesthesia and Critical Care, University of Würzburg, Würzburg, Germany
| | - Peter Kranke
- Department of Anaesthesia and Critical Care, University of Würzburg, Würzburg, Germany
| | - Joerg J Meerpohl
- Institute for Evidence in Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Cochrane Germany, Cochrane Germany Foundation, Freiburg, Germany
| | - Ravi Gill
- Department of Anaesthetics, Southampton University Hospital NHS Trust, Southampton, UK
| |
Collapse
|
46
|
Olivier RMR, Fischer L, Steinbicker AU. Patient Blood Management. ZEITSCHRIFT FUR HERZ THORAX UND GEFASSCHIRURGIE 2020. [DOI: 10.1007/s00398-020-00383-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
47
|
Abreu R, Giri P, Quinn F. Host-Pathogen Interaction as a Novel Target for Host-Directed Therapies in Tuberculosis. Front Immunol 2020; 11:1553. [PMID: 32849525 PMCID: PMC7396704 DOI: 10.3389/fimmu.2020.01553] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 06/12/2020] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis (TB) has been a transmittable human disease for many thousands of years, and M. tuberculosis is again the number one cause of death worldwide due to a single infectious agent. The intense 6- to 10-month process of multi-drug treatment, combined with the adverse side effects that can run the spectrum from gastrointestinal disturbances to liver toxicity or peripheral neuropathy are major obstacles to patient compliance and therapy completion. The consequent increase in multidrug resistant TB (MDR-TB) and extensively drug resistant TB (XDR-TB) cases requires that we increase our arsenal of effective drugs, particularly novel therapeutic approaches. Over the millennia, host and pathogen have evolved mechanisms and relationships that greatly influence the outcome of infection. Understanding these evolutionary interactions and their impact on bacterial clearance or host pathology will lead the way toward rational development of new therapeutics that favor enhancing a host protective response. These host-directed therapies have recently demonstrated promising results against M. tuberculosis, adding to the effectiveness of currently available anti-mycobacterial drugs that directly kill the organism or slow mycobacterial replication. Here we review the host-pathogen interactions during M. tuberculosis infection, describe how M. tuberculosis bacilli modulate and evade the host immune system, and discuss the currently available host-directed therapies that target these bacterial factors. Rather than provide an exhaustive description of M. tuberculosis virulence factors, which falls outside the scope of this review, we will instead focus on the host-pathogen interactions that lead to increased bacterial growth or host immune evasion, and that can be modulated by existing host-directed therapies.
Collapse
Affiliation(s)
| | | | - Fred Quinn
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States
| |
Collapse
|
48
|
Kim YJ, Kim KS, Lim D, Yang DJ, Park JI, Kim KW, Jeong JH, Choi HS, Kim DK. Epigallocatechin-3-Gallate (EGCG)-Inducible SMILE Inhibits STAT3-Mediated Hepcidin Gene Expression. Antioxidants (Basel) 2020; 9:antiox9060514. [PMID: 32545266 PMCID: PMC7346121 DOI: 10.3390/antiox9060514] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/27/2020] [Accepted: 06/09/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatic peptide hormone hepcidin, a key regulator of iron metabolism, is induced by inflammatory cytokine interleukin-6 (IL-6) in the pathogenesis of anemia of inflammation or microbial infections. Small heterodimer partner-interacting leucine zipper protein (SMILE)/CREBZF is a transcriptional corepressor of nuclear receptors that control hepatic glucose and lipid metabolism. Here, we examined the role of SMILE in regulating iron metabolism by inflammatory signals. Overexpression of SMILE significantly decreased activation of the Janus kinase 2-signal transducer and activator of transcription 3 (STAT3)-mediated hepcidin production and secretion that is triggered by the IL-6 signal in human and mouse hepatocytes. Moreover, SMILE co-localized and physically interacted with STAT3 in the nucleus in the presence of IL-6, which significantly suppressed binding of STAT3 to the hepcidin gene promoter. Interestingly, epigallocatechin-3-gallate (EGCG), a major component of green tea, induced SMILE expression through forkhead box protein O1 (FoxO1), as demonstrated in FoxO1 knockout primary hepatocytes. In addition, EGCG inhibited IL-6-induced hepcidin expression, which was reversed by SMILE knockdown. Finally, EGCG significantly suppressed lipopolysaccharide-induced hepcidin secretion and hypoferremia through induction of SMILE expression in mice. These results reveal a previously unrecognized role of EGCG-inducible SMILE in the IL-6-dependent transcriptional regulation of iron metabolism.
Collapse
Affiliation(s)
- Yu-Ji Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Korea;
| | - Ki-Sun Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Korea; (K.-S.K.); (H.-S.C.)
| | - Daejin Lim
- Department of Microbiology, Chonnam National University Medical School, Gwangju 61468, Korea; (D.L.); (J.-H.J.)
| | - Dong Ju Yang
- Department of Oral Biology, BK21 PLUS, Yonsei University College of Dentistry, Seoul, 03722, Korea; (D.J.Y.); (K.W.K.)
| | - Jae-Il Park
- Korea Basic Science Institute, Gwangju Center at Chonnam National University, Gwangju 61186, Korea;
| | - Ki Woo Kim
- Department of Oral Biology, BK21 PLUS, Yonsei University College of Dentistry, Seoul, 03722, Korea; (D.J.Y.); (K.W.K.)
| | - Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Gwangju 61468, Korea; (D.L.); (J.-H.J.)
| | - Hueng-Sik Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Korea; (K.-S.K.); (H.-S.C.)
| | - Don-Kyu Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Korea;
- Correspondence: ; Tel.: +82-62-530-2166; Fax: +82-62-530-2160
| |
Collapse
|
49
|
Kocpinar EF, Gonul Baltaci N, Ceylan H, Kalin SN, Erdogan O, Budak H. Effect of a Prolonged Dietary Iron Intake on the Gene Expression and Activity of the Testicular Antioxidant Defense System in Rats. Biol Trace Elem Res 2020; 195:135-141. [PMID: 31309445 DOI: 10.1007/s12011-019-01817-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/04/2019] [Indexed: 02/08/2023]
Abstract
Despite the fact that iron represents a crucial element for the catalysis of many metabolic reactions, its accumulation in the cell leads to the production of reactive oxygen species (ROS), provoking pathological conditions such as cancer, cardiovascular diseases, diabetes, neurodegenerative diseases, and fertility. Thus, ROS are neutralized by the enzymatic antioxidant system for the purpose of protecting cells against any damage. Iron is a potential risk factor for male fertility. However, the mechanism of action of iron on the testicular antioxidant system at the gene and protein levels is not fully understood. Thus, the purpose of the current research was to ensure a better understanding of how the long-term iron treatment influences both gene expression and enzyme activities of the testicular antioxidant system in rat testis. The data of our study showed that a significant dose-dependent increase occurred in the iron level in rat testis. A reduction occurred in reduced glutathione (GSH) levels, which represent a marker of oxidative stress, along with long-term iron overload. The expression and activity of glucose 6-phosphate dehydrogenase (G6pd), glutathione reductase (Gr), glutathione peroxidase (Gpx), and glutathione S-transferases (Gst) were significantly affected by the presence of iron. The findings of the current research demonstrate that the long-term toxic dietary iron overload influences the gene expression and enzyme activity of the testicular antioxidant defense system, but the actual effect occurs at the protein level. This may modify the sperm function and dysfunction of the male reproductive system.
Collapse
Affiliation(s)
- Enver Fehim Kocpinar
- Science Faculty, Department of Molecular Biology and Genetics, Atatürk University, Erzurum, Turkey
- Vocational School, Department of Medical Services and Techniques, Muş Alparslan University, Mus, Turkey
| | - Nurdan Gonul Baltaci
- Science Faculty, Department of Molecular Biology and Genetics, Atatürk University, Erzurum, Turkey
| | - Hamid Ceylan
- Science Faculty, Department of Molecular Biology and Genetics, Atatürk University, Erzurum, Turkey
| | - Seyda Nur Kalin
- Science Faculty, Department of Molecular Biology and Genetics, Atatürk University, Erzurum, Turkey
| | - Orhan Erdogan
- Science Faculty, Department of Molecular Biology and Genetics, Atatürk University, Erzurum, Turkey
| | - Harun Budak
- Science Faculty, Department of Molecular Biology and Genetics, Atatürk University, Erzurum, Turkey.
| |
Collapse
|
50
|
Burton JK, Yates LC, Whyte L, Fitzsimons E, Stott DJ. New horizons in iron deficiency anaemia in older adults. Age Ageing 2020; 49:309-318. [PMID: 32103233 DOI: 10.1093/ageing/afz199] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/17/2019] [Accepted: 01/02/2020] [Indexed: 12/24/2022] Open
Abstract
Iron deficiency anaemia (IDA) is common in older adults and associated with a range of adverse outcomes. Differentiating iron deficiency from other causes of anaemia is important to ensure appropriate investigations and treatment. It is possible to make the diagnosis reliably using simple blood tests. Clinical evaluation and assessment are required to help determine the underlying cause and to initiate appropriate investigations. IDA in men and post-menopausal females is most commonly due to occult gastrointestinal blood loss until proven otherwise, although there is a spectrum of underlying causative pathologies. Investigation decisions should take account of the wishes of the patient and their competing comorbidities, individualising the approach. Management involves supplementation using oral or intravenous (IV) iron then consideration of treatment of the underlying cause of deficiency. Future research areas are outlined including the role of Hepcidin and serum soluble transferrin receptor measurement, quantitative faecal immunochemical testing, alternative dosing regimens and the potential role of IV iron preparations.
Collapse
Affiliation(s)
- Jennifer Kirsty Burton
- Academic Section of Geriatric Medicine, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Glasgow Royal Infirmary, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Luke C Yates
- Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Lindsay Whyte
- Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Edward Fitzsimons
- Department of Haematology, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - David J Stott
- Academic Section of Geriatric Medicine, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|