1
|
Wang B, Wirth R, Bergmann E, Funk L, Giehl C, Levermann I, Lueg G, Roloff T, Schnepper M, Stoev K, Zubi R, Neuendorff NR, Pourhassan M. Impact of inflammatory status on intestinal iron absorption in older hospitalized patients. Eur J Clin Nutr 2025:10.1038/s41430-025-01604-2. [PMID: 40148488 DOI: 10.1038/s41430-025-01604-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 02/19/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND AND OBJECTIVE Iron deficiency is prevalent among geriatric hospitalized patients, often coinciding with inflammation. This study aimed to determine a critical C-reactive protein (CRP) threshold for sufficient intestinal iron absorption using standardized tests. SUBJECTS/METHODS This retrospective, cross-sectional study was conducted in a geriatric acute care unit. Serum iron and CRP levels were measured before breakfast and two- and four-hours after ingestion of two iron capsules. Intestinal iron absorption was calculated by subtracting baseline values from those obtained after the test, with an increase of 100 ug/dl indicating sufficient absorption. Patients were categorized into six CRP groups: ≤0.50, 0.51-2.50, 2.51-5.0, 5.1-7.50, 7.51-10.0, and ≥10.1 mg/dl. RESULTS The study included 59 participants (73% females, age range 71-99). Iron absorption was highest in groups with lower CRP levels ≤0.50 to 2.5 mg/dl) and declined significantly as CRP increased, particularly beyond 5 mg/dl. The most significant decline was noted in patients with CRP ≥ 10.1 mg/dl. A negative correlation between inflammation, as measured by CRP, and iron absorption was found. As CRP levels escalate, there is a significant reduction in the increase of serum iron levels after 2 h. A regression analysis showed that only elevated CRP levels significantly reduced serum iron increments post-iron supplementation (P = 0.004), while other factors such as age, sex, body mass index, frailty, weight loss, hemoglobin and nutritional status had no significant impact. CONCLUSION A CRP level above 5 mg/dl is indicative of significantly impaired intestinal iron absorption in older patients, underscoring the critical influence of inflammation on iron metabolism.
Collapse
Affiliation(s)
- Baigang Wang
- Department of Geriatric Medicine, Marien Hospital Herne, Ruhr-University Bochum, Hölkeskampring 40D, 44625, Herne, Germany
| | - Rainer Wirth
- Department of Geriatric Medicine, Marien Hospital Herne, Ruhr-University Bochum, Hölkeskampring 40D, 44625, Herne, Germany
| | - Elena Bergmann
- Department of Geriatric Medicine, Marien Hospital Herne, Ruhr-University Bochum, Hölkeskampring 40D, 44625, Herne, Germany
| | - Lukas Funk
- Department of Geriatric Medicine, Marien Hospital Herne, Ruhr-University Bochum, Hölkeskampring 40D, 44625, Herne, Germany
| | - Chantal Giehl
- Department of Geriatric Medicine, Marien Hospital Herne, Ruhr-University Bochum, Hölkeskampring 40D, 44625, Herne, Germany
| | - Isabel Levermann
- Department of Geriatric Medicine, Marien Hospital Herne, Ruhr-University Bochum, Hölkeskampring 40D, 44625, Herne, Germany
| | - Gero Lueg
- Department of Geriatric Medicine, Marien Hospital Herne, Ruhr-University Bochum, Hölkeskampring 40D, 44625, Herne, Germany
| | - Tom Roloff
- Department of Geriatric Medicine, Marien Hospital Herne, Ruhr-University Bochum, Hölkeskampring 40D, 44625, Herne, Germany
| | - Maria Schnepper
- Department of Geriatric Medicine, Marien Hospital Herne, Ruhr-University Bochum, Hölkeskampring 40D, 44625, Herne, Germany
| | - Kiril Stoev
- Department of Geriatric Medicine, Marien Hospital Herne, Ruhr-University Bochum, Hölkeskampring 40D, 44625, Herne, Germany
| | - Rawi Zubi
- Department of Geriatric Medicine, Marien Hospital Herne, Ruhr-University Bochum, Hölkeskampring 40D, 44625, Herne, Germany
| | - Nina Rosa Neuendorff
- Department of Geriatric Medicine, Marien Hospital Herne, Ruhr-University Bochum, Hölkeskampring 40D, 44625, Herne, Germany
| | - Maryam Pourhassan
- Department of Geriatric Medicine, Marien Hospital Herne, Ruhr-University Bochum, Hölkeskampring 40D, 44625, Herne, Germany.
| |
Collapse
|
2
|
Kouroumalis E, Tsomidis I, Voumvouraki A. HFE-Related Hemochromatosis May Be a Primary Kupffer Cell Disease. Biomedicines 2025; 13:683. [PMID: 40149659 PMCID: PMC11940282 DOI: 10.3390/biomedicines13030683] [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: 01/14/2025] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
Iron overload can lead to increased deposition of iron and cause organ damage in the liver, the pancreas, the heart and the synovium. Iron overload disorders are due to either genetic or acquired abnormalities such as excess transfusions or chronic liver diseases. The most common genetic disease of iron deposition is classic hemochromatosis (HH) type 1, which is caused by mutations of HFE. Other rare forms of HH include type 2A with mutations at the gene hemojuvelin or type 2B with mutations in HAMP that encodes hepcidin. HH type 3, is caused by mutations of the gene that encodes transferrin receptor 2. Mutations of SLC40A1 which encodes ferroportin cause either HH type 4A or HH type 4B. In the present review, an overview of iron metabolism including absorption by enterocytes and regulation of iron by macrophages, liver sinusoidal endothelial cells (LSECs) and hepatocyte production of hepcidin is presented. Hereditary Hemochromatosis and the current pathogenetic model are analyzed. Finally, a new hypothesis based on published data was suggested. The Kupffer cell is the primary defect in HFE hemochromatosis (and possibly in types 2 and 3), while the hepcidin-relative deficiency, which is the common underlying abnormality in the three types of HH, is a secondary consequence.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Department of Gastroenterology, PAGNI University Hospital, University of Crete Medical School, 71500 Heraklion, Greece
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Ioannis Tsomidis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Greece;
| |
Collapse
|
3
|
Park MY, Agoro R, Jankauskas SS, Le Henaff C, Sitara D. Phosphorus-independent role of FGF23 in erythropoiesis and iron homeostasis. PLoS One 2024; 19:e0315228. [PMID: 39666728 PMCID: PMC11637385 DOI: 10.1371/journal.pone.0315228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024] Open
Abstract
A number of studies have reported an association between phosphorus, red blood cell (RBC) production, and iron metabolism. However, it is difficult to distinguish whether the effect of phosphorus is direct or through the actions of FGF23, and it is not clear whether phosphorus is positively or negatively associated with RBC production. In the present study, we investigated the effects of a) increased phosphorus load and b) phosphorus deficiency on erythropoiesis and iron metabolism in association with FGF23. Mice were fed either a 1.2% or 1.65% phosphorus diet and compared to mice fed a control diet containing 0.6% of phosphorus. Moreover, we used two mouse models of hypophosphatemia-induced either by dietary intervention in the form of a low phosphorus (LP) diet (0.02% of Pi) or genetically in a mouse model of X-linked hypophosphatemia (XLH)-that had opposite FGF23 levels. Phosphorus supplementation appropriately increased FGF23 levels leading to excretion of excess phosphorus and normalization of serum phosphorus levels. We also found that a phosphorus-rich diet results in inflammation-induced hypoferremia associated with reduced iron export leading to tissue iron overload. Moreover, high phosphorus intake results in ineffective erythropoiesis caused by decreased production (decreased RBCs, hemoglobin, hematocrit, and erythroid progenitors in the bone marrow) and increased destruction of RBCs, leading to anemia despite increased EPO secretion. These complications occur through the actions of elevated FGF23 in the presence of normophosphatemia. Our data also show that LP diet induces a decrease in the serum concentrations of phosphorus and FGF23, resulting in increased RBC counts, hemoglobin concentration, and hematocrit compared to mice fed normal diet. Moreover, serum iron and transferrin saturation were increased and positively correlated with serum ferritin, liver ferritin protein and mRNA expression in mice fed LP diet. However, hyp mice, the murine model of XLH, exhibit hypophosphatemia and high serum FGF23 levels, along with low number of circulating RBCs, hemoglobin, and hematocrit compared to wild-type mice. In the bone marrow, hyp mice showed reduced number of erythroid progenitors and formed significantly less BFU-E colonies compared to control mice. Serum iron levels and transferrin saturation were also decreased in hyp mice in comparison to control mice. Taken together, our data show that FGF23 acts independent of phosphorus levels to regulate erythropoiesis and iron homeostasis.
Collapse
Affiliation(s)
- Min Young Park
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States of America
| | - Rafiou Agoro
- Department of Mammalian Genetics, The Jackson Laboratory, Bar Harbor, ME, United States of America
| | | | - Carole Le Henaff
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States of America
| | - Despina Sitara
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States of America
- Department of Medicine, Holman Division of Endocrinology, Diabetes and Metabolism, New York University Grossman School of Medicine, New York, NY, United States of America
| |
Collapse
|
4
|
Mendoza E, Duque X, Reyes-Maldonado E, Hernández-Franco JI, Martínez-Andrade G, Vilchis-Gil J, Martinez H, Morán S. Serum hepcidin recalibrated values in Mexican schoolchildren by demographic characteristics, nutritional and infection/inflammation status. Ann Hematol 2024; 103:3979-3986. [PMID: 39039174 DOI: 10.1007/s00277-024-05889-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Hepcidin production is regulated by iron concentration, erythropoietic activity, and inflammation. There is no reference method for determining its levels, but results obtained through various methods strongly correlate and can be compared using recalibration equations. OBJECTIVE To describe recalibrated serum hepcidin values at different percentiles in schoolchildren, considering age, sex, inflammatory processes, H. pylori infection, and iron status. METHODS Secondary analysis of data incorporating information on inflammation, H. pylori infection, and iron status of 349 schoolchildren. Hepcidin analysis was performed using a competitive ELISA, and recalibrated hepcidin values were calculated using the inverse of the linear regression model equation obtained by van der Vorm et al. Results: Recalibrated hepcidin values were lower than non-calibrated values. In schoolchildren without infection/inflammation and without iron deficiency, recalibrated values at the 50th percentile (25th-75th) were 4.89 ng/mL (2.68-8.42). For schoolchildren without infection/inflammation but with iron deficiency, recalibrated values were 2.34 ng/mL (1.10-6.58), the lowest hepcidin values observed. The highest values were found in the group with infection/inflammation, regardless of iron deficiency status. CONCLUSIONS Recalibrated hepcidin values were lower than non-calibrated values. The highest values were observed in schoolchildren with infectious or inflammatory processes, and the lowest values were observed in schoolchildren with iron deficiency but only in the absence of infectious or inflammatory processes. Using recalibrated hepcidin values allows comparison between data obtained using different analytical methods.
Collapse
Affiliation(s)
- Eugenia Mendoza
- Infectious Diseases Research Unit, Mexican Social Security Institute, Av. Cuauhtemoc No. 330, Col. Doctores, Del. Cuauhtemoc, Mexico City, CP 06720, Mexico
| | - Ximena Duque
- Infectious Diseases Research Unit, Mexican Social Security Institute, Av. Cuauhtemoc No. 330, Col. Doctores, Del. Cuauhtemoc, Mexico City, CP 06720, Mexico.
| | - Elba Reyes-Maldonado
- Department of Hematopathology, National Polytechnic Institute, National School of Biological Sciences, Mexico City, 01135, Mexico
| | | | - Gloria Martínez-Andrade
- Academic Area of Nutrition, Institute of Health Sciences, Autonomous University of the State of Hidalgo, Pachuca Hidalgo, 42039, Mexico
| | - Jenny Vilchis-Gil
- Hospital Infantil de México "Federico Gomez", Mexico City, 06720, Mexico
| | - Homero Martinez
- Hospital Infantil de México "Federico Gomez", Mexico City, 06720, Mexico
- Global Technical Services-NTEAM, Nutrition International, Ottawa, ON, K2P 2K3, Canada
| | - Segundo Morán
- Gastroenterology Research Laboratory, Mexican Social Security Institute, Mexico City, 06720, Mexico
| |
Collapse
|
5
|
Silva-Caso W, Kym S, Merino-Luna A, Aguilar-Luis MA, Tarazona-Castro Y, Carrillo-Ng H, Bonifacio-Velez de Villa E, Aquino-Ortega R, del Valle-Mendoza J. Analysis of Ferritin, Hepcidin, Zinc, C-Reactive Protein and IL-6 Levels in COVID-19 in Patients Living at Different Altitudes in Peru. Biomedicines 2024; 12:1609. [PMID: 39062181 PMCID: PMC11275107 DOI: 10.3390/biomedicines12071609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Despite great scientific efforts, understanding the role of COVID-19 clinical biomarkers remains a challenge. METHODS A cross-sectional descriptive study in two Peruvian cities at different altitudes for comparison: Lima and Huaraz. In each place, three groups were formed, made up of 25 patients with COVID-19 in the ICU, 25 hospitalized patients with COVID-19 who did not require the ICU, and 25 healthy subjects as a control group. Five biomarkers were measured: IL-6, hepcidin, ferritin, C-reactive protein, and zinc using ELISA assays. RESULTS Ferritin, C-reactive protein, and IL-6 levels were significantly higher in the ICU and non-ICU groups at both study sites. In the case of hepcidin, the levels were significantly higher in the ICU group at both study sites compared to the non-ICU group. Among the groups within each study site, the highest altitude area presented statistically significant differences between its groups in all the markers evaluated. In the lower altitude area, differences were only observed between the groups for the zinc biomarker. CONCLUSION COVID-19 patients residing at high altitudes tend to have higher levels of zinc and IL-6 in all groups studied compared to their lower altitude counterparts.
Collapse
Affiliation(s)
- Wilmer Silva-Caso
- Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Av. San Marcos Cuadra 2, Chorrillos, Lima 15023, Peru; (M.A.A.-L.); (H.C.-N.); (R.A.-O.)
- Instituto de Investigación Nutricional, Lima 15024, Peru
| | - Sungmin Kym
- Division of Infectious Disease, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 305764, Republic of Korea
| | - Alfredo Merino-Luna
- Unidad de Cuidados Intensivos, Clinica San Pablo, Sede Huaraz, Huaraz 02002, Peru
| | - Miguel Angel Aguilar-Luis
- Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Av. San Marcos Cuadra 2, Chorrillos, Lima 15023, Peru; (M.A.A.-L.); (H.C.-N.); (R.A.-O.)
- Instituto de Investigación Nutricional, Lima 15024, Peru
| | - Yordi Tarazona-Castro
- Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Av. San Marcos Cuadra 2, Chorrillos, Lima 15023, Peru; (M.A.A.-L.); (H.C.-N.); (R.A.-O.)
| | - Hugo Carrillo-Ng
- Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Av. San Marcos Cuadra 2, Chorrillos, Lima 15023, Peru; (M.A.A.-L.); (H.C.-N.); (R.A.-O.)
| | - Eliezer Bonifacio-Velez de Villa
- Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Av. San Marcos Cuadra 2, Chorrillos, Lima 15023, Peru; (M.A.A.-L.); (H.C.-N.); (R.A.-O.)
| | - Ronald Aquino-Ortega
- Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Av. San Marcos Cuadra 2, Chorrillos, Lima 15023, Peru; (M.A.A.-L.); (H.C.-N.); (R.A.-O.)
- School of Biology, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicdas, Lima 15023, Peru
| | - Juana del Valle-Mendoza
- Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Av. San Marcos Cuadra 2, Chorrillos, Lima 15023, Peru; (M.A.A.-L.); (H.C.-N.); (R.A.-O.)
- Instituto de Investigación Nutricional, Lima 15024, Peru
| |
Collapse
|
6
|
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
|
7
|
Loveikyte R, Duijvestein M, Mujagic Z, Goetgebuer RL, Dijkstra G, van der Meulen-de Jong AE. Predicting response to iron supplementation in patients with active inflammatory bowel disease (PRIme): a randomised trial protocol. BMJ Open 2024; 14:e077511. [PMID: 38296290 PMCID: PMC10828887 DOI: 10.1136/bmjopen-2023-077511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
INTRODUCTION Iron deficiency anaemia (IDA) is the most common systemic manifestation of inflammatory bowel disease (IBD) that has detrimental effects on quality of life (QoL) and disease outcomes. Iron deficiency (ID), with or without anaemia, poses a diagnostic and therapeutic challenge in patients with IBD due to the multifactorial nature of ID(A) and its frequent recurrence. Elevated hepcidin-a systemic iron regulator that modulates systemic iron availability and intestinal iron absorption-has been associated with oral iron malabsorption in IBD. Therefore, hepcidin could assist in therapeutic decision-making. In this study, we investigate whether hepcidin can predict response to oral and intravenous iron supplementation in patients with active IBD undergoing anti-inflammatory treatment. METHODS AND ANALYSIS PRIme is an exploratory, multicentre, open-label and randomised trial. All adult patients with active IBD and ID(A) will be assessed for eligibility. The participants (n=90) will be recruited at five academic hospitals within the Netherlands and randomised into three groups (1:1:1): oral ferrous fumarate, oral ferric maltol or intravenous iron. Clinical and biochemical data will be collected at the baseline and after 6, 14 and 24 weeks. Blood samples will be collected to measure hepcidin and other biomarkers related to iron status. In addition, patient-reported outcomes regarding QoL and disease burden will be evaluated. The primary outcome is the utility of hepcidin as a predictive biomarker for response to iron therapy, which will be assessed using receiver operating curve analysis. ETHICS AND DISSEMINATION The study has been approved by the Institutional Review Board at the Leiden University Medical Center (IRB No. P21.109) and other study sites. All participants will provide written informed consent to enrol in the study. The findings will be published in a peer-reviewed journal and disseminated at scientific conferences; the dataset will be available on reasonable request. TRIAL REGISTRATION Prospectively registered in the https://clinicaltrials.gov/ and the Eudra registries. First submitted on 10 May 2022 to the ClinicalTrials.gov (ID: NCT05456932) and on 3 March 2022 to the European Union Drug Regulating Authorities Clinical Trials Database (ID: 2022-000894-16).
Collapse
Affiliation(s)
- Roberta Loveikyte
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, Groningen, The Netherlands
| | - Marjolijn Duijvestein
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Zlatan Mujagic
- Department of Gastroenterology and Hepatology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Rogier L Goetgebuer
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, Groningen, The Netherlands
| | | |
Collapse
|
8
|
Loveikyte R, Bourgonje AR, van Goor H, Dijkstra G, van der Meulen-de Jong AE. The effect of iron therapy on oxidative stress and intestinal microbiota in inflammatory bowel diseases: A review on the conundrum. Redox Biol 2023; 68:102950. [PMID: 37918126 PMCID: PMC10643537 DOI: 10.1016/j.redox.2023.102950] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 10/28/2023] [Indexed: 11/04/2023] Open
Abstract
One in five patients with Inflammatory Bowel Disease (IBD) suffers from anemia, most frequently caused by iron deficiency. Anemia and iron deficiency are associated with worse disease outcomes, reduced quality of life, decreased economic participation, and increased healthcare costs. International guidelines and consensus-based recommendations have emphasized the importance of treating anemia and iron deficiency. In this review, we draw attention to the rarely discussed effects of iron deficiency and iron therapy on the redox status, the intestinal microbiota, and the potential interplay between them, focusing on the clinical implications for patients with IBD. Current data are scarce, inconsistent, and do not provide definitive answers. Nevertheless, it is imperative to rule out infections and discern iron deficiency anemia from other types of anemia to prevent untargeted oral or intravenous iron supplementation and potential side effects, including oxidative stress. Further research is necessary to establish the clinical significance of changes in the redox status and the intestinal microbiota following iron supplementation.
Collapse
Affiliation(s)
- R Loveikyte
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands; Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - A R Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; The Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - H van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - G Dijkstra
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - A E van der Meulen-de Jong
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
9
|
Loveikyte R, Bourgonje AR, van der Reijden JJ, Bulthuis MLC, Hawinkels LJAC, Visschedijk MC, Festen EAM, van Dullemen HM, Weersma RK, van Goor H, van der Meulen-de Jong AE, Dijkstra G. Hepcidin and Iron Status in Patients With Inflammatory Bowel Disease Undergoing Induction Therapy With Vedolizumab or Infliximab. Inflamm Bowel Dis 2023:7030568. [PMID: 36748574 PMCID: PMC10393210 DOI: 10.1093/ibd/izad010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Hepcidin, the systemic iron regulator, could be critical in differentiating iron deficiency (ID) from functional iron restriction in inflammatory bowel disease (IBD). We assessed hepcidin as a diagnostic ID marker and explored the relationship between hepcidin and its regulators in patients with IBD undergoing induction therapy with infliximab (IFX) or vedolizumab (VEDO). METHODS Patients with active IBD receiving induction therapy with IFX or VEDO were included. Serum samples at baseline and after 6 weeks of induction therapy were analyzed for hepcidin, inflammation- and hypoxia-associated cytokines, and oxidative stress. Data were analyzed by stratifying based on the response at week 14. Results were compared with samples from age- and sex-matched healthy control subjects. RESULTS Patients receiving induction therapy with IFX (n = 71) or VEDO (n = 51) and healthy control subjects (n = 50) were included. At baseline, hepcidin correlated positively with ferritin and negatively with soluble transferrin receptor/log ferritin index (P < .001). ID was prevalent in 96.7% of patients who had hepcidin levels below the median. Hepcidin accurately identified ID: the area under the curve (hepcidin) was 0.89 (95% confidence interval, 0.82-0.95; P < .001). In total, 75.4% of patients responded to induction therapy; inflammation, hepcidin, and ferritin decreased significantly, while transferrin increased during induction therapy. These changes were observed only in patients who responded to the therapy. CONCLUSIONS Hepcidin levels in IBD are primarily determined by ID, even in an inflammatory state. In addition, induction therapy can decrease hepcidin levels, which might lead to better bioavailability of iron supplements. Therefore, hepcidin is a potential diagnostic ID biomarker that could assist therapeutic decision making.
Collapse
Affiliation(s)
- Roberta Loveikyte
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden University, Leiden, the Netherlands.,Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Johannes J van der Reijden
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Marian L C Bulthuis
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Lukas J A C Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
| | - Marijn C Visschedijk
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Eleonora A M Festen
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Hendrik M van Dullemen
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
10
|
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: 3.0] [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
|
11
|
Iron deficient diets modify the gut microbiome and reduce the severity of enteric infection in a mouse model of S. Typhimurium-induced enterocolitis. J Nutr Biochem 2022; 107:109065. [DOI: 10.1016/j.jnutbio.2022.109065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 01/04/2023]
|
12
|
Peng D, Gao Y, Zhang L, Liu Z, Wang H, Liu Y. The Relationship Between Hepcidin-Mediated Iron Dysmetabolism and COVID-19 Severity: A Meta-Analysis. Front Public Health 2022; 10:881412. [PMID: 35558525 PMCID: PMC9087037 DOI: 10.3389/fpubh.2022.881412] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/30/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUNDS Hepcidin has been identified as a systemic iron-regulatory hormone. Recent studies have suggested that iron metabolism disorders may be involved in the pathogenesis of acute respiratory distress syndrome and multiple organ dysfunction in coronavirus disease 2019 (COVID-19). OBJECTIVES To re-evaluate the hepcidin-related iron metabolism parameters and explore the relationship between hepcidin-mediated iron dysmetabolism and COVID-19 severity. METHODS COVID-19 is classified as mild and moderate as non-severe, severe and critical as severe. A meta-analysis was conducted. Four bibliographic databases were comprehensively searched up to December 31st 2021. RESULTS Six unique studies with data from 477 COVID-19 patients were included. Compared to non-severe cases, severe cases had higher hepcidin (standardized mean difference (SMD), -0.39; 95% Confidence Interval (CI) [-0.76, -0.03]; P = 0.03) and ferritin (SMD, -0.84; 95% CI [-1.30, -0.38]; P = 0.0004). In five out of six studies, a total of 427 patients were tested for serum iron, and there were significant differences in their levels between severe and non-severe cases (SMD, 0.22; 95% CI [0.02, 0.41]; P = 0.03). A total of 320 patients from four out of six studies were tested for transferrin saturation, and the statistical difference was not significant (SMD, 0.06; 95% CI [-0.17, 0.28]; P = 0.64). CONCLUSION Severe COVID-19 cases had higher serum levels of hepcidin and ferritin, and lower serum iron, without significant differences in transferrin saturation. Further studies are needed to verify whether targeting the hepcidin-mediated iron metabolism axis may influence the outcome and treatment of COVID-19.
Collapse
Affiliation(s)
- Denggao Peng
- Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
- Graduate Collaborative Training Base of Shenzhen Third People's Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yanzhang Gao
- Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Li Zhang
- Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Zhichao Liu
- Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Huan Wang
- Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Yingxia Liu
- Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
- Graduate Collaborative Training Base of Shenzhen Third People's Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
13
|
van der Staaij H, Donker AE, Bakkeren DL, Salemans JMJI, Mignot-Evers LAA, Bongers MY, Dieleman JP, Galesloot TE, Laarakkers CM, Klaver SM, Swinkels DW. Transferrin Saturation/Hepcidin Ratio Discriminates TMPRSS6-Related Iron Refractory Iron Deficiency Anemia from Patients with Multi-Causal Iron Deficiency Anemia. Int J Mol Sci 2022; 23:ijms23031917. [PMID: 35163840 PMCID: PMC8836508 DOI: 10.3390/ijms23031917] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/30/2022] [Accepted: 02/03/2022] [Indexed: 02/04/2023] Open
Abstract
Pathogenic TMPRSS6 variants impairing matriptase-2 function result in inappropriately high hepcidin levels relative to body iron status, leading to iron refractory iron deficiency anemia (IRIDA). As diagnosing IRIDA can be challenging due to its genotypical and phenotypical heterogeneity, we assessed the transferrin saturation (TSAT)/hepcidin ratio to distinguish IRIDA from multi-causal iron deficiency anemia (IDA). We included 20 IRIDA patients from a registry for rare inherited iron disorders and then enrolled 39 controls with IDA due to other causes. Plasma hepcidin-25 levels were measured by standardized isotope dilution mass spectrometry. IDA controls had not received iron therapy in the last 3 months and C-reactive protein levels were <10.0 mg/L. IRIDA patients had significantly lower TSAT/hepcidin ratios compared to IDA controls, median 0.6%/nM (interquartile range, IQR, 0.4-1.1%/nM) and 16.7%/nM (IQR, 12.0-24.0%/nM), respectively. The area under the curve for the TSAT/hepcidin ratio was 1.000 with 100% sensitivity and specificity (95% confidence intervals 84-100% and 91-100%, respectively) at an optimal cut-off point of 5.6%/nM. The TSAT/hepcidin ratio shows excellent performance in discriminating IRIDA from TMPRSS6-unrelated IDA early in the diagnostic work-up of IDA provided that recent iron therapy and moderate-to-severe inflammation are absent. These observations warrant further exploration in a broader IDA population.
Collapse
Affiliation(s)
- Hilde van der Staaij
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center (Radboudumc), 6525 GA Nijmegen, The Netherlands; (H.v.d.S.); (A.E.D.); (C.M.L.); (S.M.K.)
- Máxima Medical Center (MMC), Department of Pediatrics, 5504 DB Veldhoven, The Netherlands
| | - Albertine E. Donker
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center (Radboudumc), 6525 GA Nijmegen, The Netherlands; (H.v.d.S.); (A.E.D.); (C.M.L.); (S.M.K.)
- Máxima Medical Center (MMC), Department of Pediatrics, 5504 DB Veldhoven, The Netherlands
| | - Dirk L. Bakkeren
- Máxima Medical Center (MMC), Department of Clinical Chemistry, 5504 DB Veldhoven, The Netherlands;
| | - Jan M. J. I. Salemans
- Máxima Medical Center (MMC), Department of Gastroenterology, 5504 DB Veldhoven, The Netherlands;
| | | | - Marlies Y. Bongers
- Máxima Medical Center (MMC), Department of Gynecology, 5504 DB Veldhoven, The Netherlands;
- Maastricht University Medical Center, Department of Gynecology, 6229 HX Maastricht, The Netherlands
| | - Jeanne P. Dieleman
- Máxima Medical Center Academy, Máxima Medical Center (MMC), 5504 DB Veldhoven, The Netherlands;
| | - Tessel E. Galesloot
- Department for Health Evidence, Radboud Institute for Health Sciences, 6500 HB Nijmegen, The Netherlands;
| | - Coby M. Laarakkers
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center (Radboudumc), 6525 GA Nijmegen, The Netherlands; (H.v.d.S.); (A.E.D.); (C.M.L.); (S.M.K.)
- Hepcidinanalysis, Translational Metabolic Laboratory, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands
| | - Siem M. Klaver
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center (Radboudumc), 6525 GA Nijmegen, The Netherlands; (H.v.d.S.); (A.E.D.); (C.M.L.); (S.M.K.)
- Hepcidinanalysis, Translational Metabolic Laboratory, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands
| | - Dorine W. Swinkels
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center (Radboudumc), 6525 GA Nijmegen, The Netherlands; (H.v.d.S.); (A.E.D.); (C.M.L.); (S.M.K.)
- Hepcidinanalysis, Translational Metabolic Laboratory, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands
- Correspondence: ; Tel.: +31-(0)2-4361-8957
| |
Collapse
|
14
|
Investigating the Molecular Mechanisms of Renal Hepcidin Induction and Protection upon Hemoglobin-Induced Acute Kidney Injury. Int J Mol Sci 2022; 23:ijms23031352. [PMID: 35163276 PMCID: PMC8835743 DOI: 10.3390/ijms23031352] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 01/27/2023] Open
Abstract
Hemolysis is known to cause acute kidney injury (AKI). The iron regulatory hormone hepcidin, produced by renal distal tubules, is suggested to exert a renoprotective role during this pathology. We aimed to elucidate the molecular mechanisms of renal hepcidin synthesis and its protection against hemoglobin-induced AKI. In contrast to known hepatic hepcidin induction, incubation of mouse cortical collecting duct (mCCDcl1) cells with IL-6 or LPS did not induce Hamp1 mRNA expression, whereas iron (FeS) and hemin significantly induced hepcidin synthesis (p < 0.05). Moreover, iron/heme-mediated hepcidin induction in mCCDcl1 cells was caused by the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, as indicated by increased nuclear Nrf2 translocation and induced expression of Nrf2 downstream targets GCLM (p < 0.001), NQO1 (p < 0.001), and TXNRD1 (p < 0.005), which could be prevented by the known Nrf2 inhibitor trigonelline. Newly created inducible kidney-specific hepcidin KO mice demonstrated a significant reduction in renal Hamp1 mRNA expression. Phenylhydrazine (PHZ)-induced hemolysis caused renal iron loading and oxidative stress in both wildtype (Wt) and KO mice. PHZ treatment in Wt induced inflammatory markers (IL-6, TNFα) but not Hamp1. However, since PHZ treatment also significantly reduced systemic hepcidin levels in both Wt and KO mice (both p < 0.001), a dissection between the roles of systemic and renal hepcidin could not be made. Combined, the results of our study indicate that there are kidney-specific mechanisms in hepcidin regulation, as indicated by the dominant role of iron and not inflammation as an inducer of renal hepcidin, but also emphasize the complex interplay of various iron regulatory mechanisms during AKI on a local and systemic level.
Collapse
|
15
|
Anemia feriprivă – manifestare de debut al unei boli celiace oculte. ONCOLOG-HEMATOLOG.RO 2022. [DOI: 10.26416/onhe.60.3.2022.7153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
16
|
Stoffel NU, Zimmermann MB, Cepeda-Lopez AC, Cervantes-Gracia K, Llanas-Cornejo D, Zeder C, Tuntipopipat S, Moungmaithong S, Densupsoontorn N, Quack Loetscher K, Gowachirapant S, Herter-Aeberli I. Maternal iron kinetics and maternal-fetal iron transfer in normal-weight and overweight pregnancy. Am J Clin Nutr 2021; 115:1166-1179. [PMID: 34910118 PMCID: PMC8970997 DOI: 10.1093/ajcn/nqab406] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/07/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Inflammation during pregnancy may aggravate iron deficiency (ID) by increasing serum hepcidin and reducing iron absorption. This could restrict iron transfer to the fetus, increasing risk of infant ID and its adverse effects. OBJECTIVES We aimed to assess whether iron bioavailability and/or iron transfer to the fetus is impaired in overweight/obese (OW) pregnant women with adiposity-related inflammation, compared with normal-weight (NW) pregnant women. METHODS In this prospective study, we followed NW (n = 43) and OW (n = 40) pregnant women who were receiving iron supplements from the 14th week of gestation to term and followed their infants to age 6 mo. We administered 57Fe and 58Fe in test meals mid-second and mid-third trimester, and measured tracer kinetics throughout pregnancy and infancy. RESULTS In total, 38 NW and 36 OW women completed the study to pregnancy week 36, whereas 30 NW and 27 OW mother-infant pairs completed the study to 6 mo postpartum. Both groups had comparable iron status, hemoglobin, and serum hepcidin throughout pregnancy. Compared with the NW, the OW pregnant women had 1) 43% lower fractional iron absorption (FIA) in the third trimester (P = 0.033) with median [IQR] FIA of 23.9% [11.4%-35.7%] and 13.5% [10.8%-19.5%], respectively; and 2) 17% lower maternal-fetal iron transfer from the first tracer (P = 0.051) with median [IQR] maternal-fetal iron transfer of 4.8% [4.2%-5.4%] and 4.0% [3.6%-4.6%], respectively. Compared with the infants born to NW women, infants born to OW women had lower body iron stores (BIS) with median [IQR] 7.7 [6.3-8.8] and 6.6 [4.6-9.2] mg/kg body weight at age 6 mo, respectively (P = 0.024). Prepregnancy BMI was a negative predictor of maternal-fetal iron transfer (β = -0.339, SE = 0.144, P = 0.025) and infant BIS (β = -0.237, SE = 0.026, P = 0.001). CONCLUSIONS Compared with NW, OW pregnant women failed to upregulate iron absorption in late pregnancy, transferred less iron to their fetus, and their infants had lower BIS. These impairments were associated with inflammation independently of serum hepcidin.This trial was registered at clinicaltrials.gov as NCT02747316.
Collapse
Affiliation(s)
- Nicole U Stoffel
- Laboratory of Human Nutrition, Department of Health Science and Technology, ETH Zurich, Zürich, Switzerland
| | - Michael B Zimmermann
- Laboratory of Human Nutrition, Department of Health Science and Technology, ETH Zurich, Zürich, Switzerland,Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford and John Radcliffe Hospital, Oxford, United Kingdom
| | | | - Karla Cervantes-Gracia
- Department of Basic Sciences, School of Medicine, Universidad de Monterrey, Monterrey, Mexico
| | - Daniel Llanas-Cornejo
- Department of Basic Sciences, School of Medicine, Universidad de Monterrey, Monterrey, Mexico
| | - Christophe Zeder
- Laboratory of Human Nutrition, Department of Health Science and Technology, ETH Zurich, Zürich, Switzerland
| | | | | | | | | | | | - Isabelle Herter-Aeberli
- Laboratory of Human Nutrition, Department of Health Science and Technology, ETH Zurich, Zürich, Switzerland
| |
Collapse
|
17
|
The effects of reducing chronic inflammation in overweight women on serum hepcidin and iron absorption with and without supplemental ascorbic acid. Br J Nutr 2021; 126:877-884. [PMID: 33243304 DOI: 10.1017/s0007114520004730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Although hepcidin synthesis is stimulated by inflammation and inhibited by Fe deficiency, the strength of their opposing effects on serum hepcidin (SHep) in humans remains unclear. It was recently shown that an inflammatory stimulus in anaemic women did not increase SHep or decrease Fe absorption. The enhancing effect of ascorbic acid on Fe absorption may not be effective during inflammation because of increased SHep. Our study aim was to test whether reducing inflammation in Fe-depleted overweight (OW) women with low-grade inflammation would lower SHep and improve Fe absorption with and without ascorbic acid, compared with normal-weight (NW) women without inflammation. Before and after 14 d of anti-inflammatory treatment (3 × 600 mg ibuprofen daily) in OW and NW women (n 36; 19-46 years of age), we measured SHep and fractional Fe absorption (FIA) (erythrocyte Fe incorporation) from 57Fe- and 58Fe-labelled test meals with and without ascorbic acid. There were significant group effects on IL-6, C-reactive protein, serum ferritin and SHep (for all, P < 0·05). There was a significant treatment effect on SHep (P < 0·05): in OW women, treatment decreased IL-6 by approximately 30 % and SHep by approximately 45 %. However, there were no significant treatment or group effects on FIA. Body Fe stores (BIS) were a significant positive predictor of SHep before and after treatment (P < 0·001), but IL-6 was not. Reducing chronic inflammation in OW women halved SHep but did not affect Fe absorption with or without ascorbic acid, and the main predictor of Fe absorption was BIS.
Collapse
|
18
|
Papasavva M, Vikelis M, Katsarou MS, Siokas V, Dermitzakis E, Papademetriou C, Karakostis K, Lazopoulos G, Dardiotis E, Drakoulis N. Evidence That HFE H63D Variant Is a Potential Disease Modifier in Cluster Headache. J Mol Neurosci 2021; 72:393-400. [PMID: 34570359 PMCID: PMC8840935 DOI: 10.1007/s12031-021-01913-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022]
Abstract
Cluster headache (CH) is a primary headache disorder with a complex genetic background. Several studies indicate a potential link between iron homeostasis and the pathophysiology of primary headaches. The HFE gene encodes for a protein involved in iron metabolism, while genetic variants in HFE have been associated with hereditary hemochromatosis (HH), an iron overload disorder. The objective of the current study was to examine the association of the more common HFE H63D variant, with the susceptibility to develop CH and diverse clinical phenotypes in a population of Southeastern European Caucasian (SEC) origin. Genomic DNA samples from 128 CH patients and 294 neurologically healthy controls were genotyped for the HFE rs1799945 (H63D) variant. H63D genotypic and allelic frequency distribution did not differ significantly between patients and controls (p > 0.05). Subgroup analysis revealed a significantly more frequent occurrence of the variant G allele in chronic compared to episodic CH patients, indicative for a possible correlation of the HFE gene with the susceptibility for disease chronification. Although homozygosity for the less prevalent H63D variant G allele was minimal in the CH cohort, the results of the present study are in accordance with previous studies in CH and migraine patients, suggesting that HFE H63D variant modifies the disease clinical characteristics. Hence, despite the absence of a per se association with CH susceptibility in the current SEC cohort, variability in HFE gene may be potentially regarded as a disease modifier genetic factor in CH.
Collapse
Affiliation(s)
- Maria Papasavva
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771, Athens, Greece
| | | | - Martha-Spyridoula Katsarou
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771, Athens, Greece
| | - Vasileios Siokas
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | | | - Christoforos Papademetriou
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771, Athens, Greece
| | | | - George Lazopoulos
- Department of Cardiothoracic Surgery, University General Hospital of Heraklion, Medical School, University of Crete, 71003, Heraklion, Greece
| | - Efthimios Dardiotis
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Greece, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Nikolaos Drakoulis
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771, Athens, Greece.
| |
Collapse
|
19
|
Borawski B, Malyszko JS, Kwiatkowska M, Malyszko J. Current Status of Renal Anemia Pharmacotherapy-What Can We Offer Today. J Clin Med 2021; 10:jcm10184149. [PMID: 34575261 PMCID: PMC8470821 DOI: 10.3390/jcm10184149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 08/30/2021] [Accepted: 09/07/2021] [Indexed: 12/19/2022] Open
Abstract
Chronic kidney disease (CKD) is one of the fastest-growing major causes of death internationally. Better treatment of CKD and its complications is crucial to reverse this negative trend. Anemia is a frequent complication of CKD and is associated with unfavorable clinical outcomes. It is a devastating complication of progressive kidney disease, that negatively affects also the quality of life. The prevalence of anemia increases in parallel with CKD progression. The aim of this review is to summarize the current knowledge on therapy of renal anemia. Iron therapy, blood transfusions, and erythropoietin stimulating agents are still the mainstay of renal anemia treatment. There are several novel agents on the horizon that might provide therapeutic opportunities in CKD. The potential therapeutic options target the hepcidin–ferroportin axis, which is the master regulator of iron homeostasis, and the BMP-SMAD pathway, which regulates hepcidin expression in the liver. An inhibition of prolyl hydroxylase is a new therapeutic option becoming available for the treatment of anemia in CKD patients. This new class of drugs stimulates the synthesis of endogenous erythropoietin and increases iron availability. We also summarized the effects of prolyl hydroxylase inhibitors on iron parameters, including hepcidin, as their action on the hematological parameters. They could be of particular interest in the out-patient population with CKD and patients with ESA hyporesponsiveness. However, current knowledge is limited and still awaits clinical validation. One should be aware of the potential risks and benefits of novel, sophisticated therapies.
Collapse
Affiliation(s)
- Bartłomiej Borawski
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (B.B.); (M.K.)
| | - Jacek Stanislaw Malyszko
- 1st Department of Nephrology and Transplantology, Medical University of Bialystok, 15-540 Bialystok, Poland;
| | - Marlena Kwiatkowska
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (B.B.); (M.K.)
| | - Jolanta Malyszko
- Department of Nephrology, Dialysis and Internal Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland; (B.B.); (M.K.)
- Correspondence:
| |
Collapse
|
20
|
Abuga KM, Muriuki JM, Uyoga SM, Mwai K, Makale J, Mogire RM, Macharia AW, Mohammed S, Muthumbi E, Mwarumba S, Mturi N, Bejon P, Scott JAG, Nairz M, Williams TN, Atkinson SH. Hepcidin regulation in Kenyan children with severe malaria and non-typhoidal Salmonella bacteremia. Haematologica 2021; 107:1589-1598. [PMID: 34498446 PMCID: PMC9244826 DOI: 10.3324/haematol.2021.279316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Indexed: 11/09/2022] Open
Abstract
Malaria and invasive non-typhoidal Salmonella (NTS) are life-threatening infections that often co-exist in African children. The iron-regulatory hormone hepcidin is highly upregulated during malaria and controls the availability of iron, a critical nutrient for bacterial growth. We investigated the relationship between Plasmodium falciparum malaria and NTS bacteremia in all pediatric admissions aged <5 years between August 1998 and October 2019 (n=75,034). We then assayed hepcidin and measures of iron status in five groups: (1) children with concomitant severe malarial anemia (SMA) and NTS (SMA+NTS, n=16); and in matched children with (2) SMA (n=33); (3) NTS (n=33); (4) cerebral malaria (CM, n=34); and (5) community-based children. SMA and severe anemia without malaria were associated with a 2-fold or more increased risk of NTS bacteremia, while other malaria phenotypes were not associated with increased NTS risk. Children with SMA had lower hepcidin/ferritin ratios (0.10; interquartile range [IQR]: 0.03-0.19) than those with CM (0.24; IQR: 0.14-0.69; P=0.006) or asymptomatic malaria (0.19; IQR: 0.09-0.46; P=0.01) indicating suppressed hepcidin levels. Children with SMA+NTS had lower hepcidin levels (9.3 ng/mL; IQR: 4.7-49.8) and hepcidin/ferritin ratios (0.03; IQR: 0.01-0.22) than those with NTS alone (105.8 ng/mL; IQR: 17.3-233.3; P=0.02 and 0.31; IQR: 0.06-0.66; P=0.007, respectively). Since hepcidin degrades ferroportin on the Salmonella-containing vacuole, we hypothesize that reduced hepcidin in children with SMA might contribute to NTS growth by modulating iron availability for bacterial growth. Further studies are needed to understand how the hepcidin-ferroportin axis might mediate susceptibility to NTS in severely anemic children.
Collapse
Affiliation(s)
- Kelvin M. Abuga
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya,Department of Public Health, School of Human and Health Sciences, Pwani University, Kilifi, Kenya,Kelvin M. Abuga
| | - John Muthii Muriuki
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| | - Sophie M. Uyoga
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| | - Kennedy Mwai
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya,Epidemiology and Biostatistics Division, School of Public Health, University of the Witwatersrand, Johannesburg, South Africa
| | - Johnstone Makale
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| | - Reagan M. Mogire
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya,Open University, KEMRI-Wellcome Trust Research Program – Accredited Research Center, Kilifi, Kenya
| | - Alex W. Macharia
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya,Open University, KEMRI-Wellcome Trust Research Program – Accredited Research Center, Kilifi, Kenya
| | - Shebe Mohammed
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| | - Esther Muthumbi
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| | - Salim Mwarumba
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| | - Neema Mturi
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| | - Philip Bejon
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya,Center for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - J. Anthony G. Scott
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya,Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Manfred Nairz
- Department of Internal Medicine II, Medical University Innsbruck, Innsbruck, Austria
| | - Thomas N. Williams
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya,Center for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK,Department of Infectious Diseases and Institute of Global Health Innovation, Imperial College, London, UK
| | - Sarah H. Atkinson
- Kenya Medical Research Institute (KEMRI) Center for Geographic Medicine Research, KEMRI-Wellcome Trust Research Program, Kilifi, Kenya,Center for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK,Department of Pediatrics, University of Oxford, Oxford, UK,Sarah H. Atkinson
| |
Collapse
|
21
|
Speich C, Brittenham GM, Cercamondi CI, Zeder C, Nkosi-Gondwe T, Phiri KS, Moretti D, Zimmermann MB. Isotopic measurement of iron requirements in sub-Saharan African children. Am J Clin Nutr 2021; 114:986-996. [PMID: 34113969 DOI: 10.1093/ajcn/nqab161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 04/19/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Prevention of iron deficiency in African children is a public health priority. Current WHO/FAO estimations of iron requirements are derived from factorial estimates based on healthy, iron-sufficient "model" children using data derived mainly from adults. OBJECTIVES In this study, we aimed to quantify iron absorption, loss, and balance in apparently healthy 5- to 7-y-old children living in rural Africa. METHODS We directly measured long-term iron absorption and iron loss in a 2-y observational study in Malawian children (n = 48) using a novel stable iron isotope method. RESULTS Of the 36 children with height-for-age and weight-for-age z scores ≥-2, 13 (36%) were iron deficient (soluble transferrin receptor >8.3 mg/L) and 23 were iron sufficient. Iron-deficient children weighed more than iron-sufficient children [mean difference (95% CI): +2.1 (1.4, 2.7) kg; P = 0.01]. Mean iron losses did not differ significantly between iron-deficient and iron-sufficient children and were comparable to WHO/FAO median estimates of 19 µg/(d × kg). In iron-sufficient children, median (95% CI) dietary iron absorption was 32 (28, 34) µg/(d × kg), comparable to WHO/FAO-estimated median requirements of 32 µg/(d × kg). In iron-deficient children, absorption of 28 (25, 30) µg/(d × kg) was not increased to correct their iron deficit, likely because of a lack of bioavailable dietary iron. Twelve children (25%) were undernourished (underweight, stunted, or both). CONCLUSIONS Our results suggest that WHO/FAO iron requirements are adequate for healthy iron-sufficient children in this rural area of Malawi, but iron-deficient children require additional bioavailable iron to correct their iron deficit.
Collapse
Affiliation(s)
- Cornelia Speich
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, ETH Zürich, Zurich, Switzerland
| | - Gary M Brittenham
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Colin I Cercamondi
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, ETH Zürich, Zurich, Switzerland
| | - Christophe Zeder
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, ETH Zürich, Zurich, Switzerland
| | - Thandile Nkosi-Gondwe
- Training and Research Unit of Excellence, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Kamija S Phiri
- Training and Research Unit of Excellence, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Diego Moretti
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, ETH Zürich, Zurich, Switzerland.,Nutrition Group, Health Department, Swiss Distance University of Applied Sciences, Regensdorf, Switzerland
| | - Michael B Zimmermann
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
22
|
Girelli D, Marchi G, Busti F, Vianello A. Iron metabolism in infections: Focus on COVID-19. Semin Hematol 2021; 58:182-187. [PMID: 34389110 PMCID: PMC8305218 DOI: 10.1053/j.seminhematol.2021.07.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/12/2021] [Indexed: 12/15/2022]
Abstract
Iron is a micronutrient essential for a wide range of metabolic processes in virtually all living organisms. During infections, a battle for iron takes place between the human host and the invading pathogens. The liver peptide hepcidin, which is phylogenetically and structurally linked to defensins (antimicrobial peptides of the innate immunity), plays a pivotal role by subtracting iron to pathogens through its sequestration into host cells, mainly macrophages. While this phenomenon is well studied in certain bacterial infections, much less is known regarding viral infections. Iron metabolism also has implications on the functionality of cells of the immune system. Once primed by the contact with antigen presenting cells, lymphocytes need iron to sustain the metabolic burst required for mounting an effective cellular and humoral response. The COVID-19 pandemic has boosted an amount of clinical and translational research over the possible influences of nutrients on SARS-CoV-2 infection, in terms of either susceptibility or clinical course. Here we review the intersections between iron metabolism and COVID-19, belonging to the wider domain of the so-called “nutritional immunity”. A better understanding of such connections has potential broad implications, either from a mechanistic standpoint, or for the development of more effective strategies for managing COVID-19 and possible future pandemics.
Collapse
Affiliation(s)
- Domenico Girelli
- Department of Medicine, Section of Internal Medicine, University of Verona, Euro Blood Net Referral Center, Azienda Ospedaliera Universitaria Integrata Verona, Italy.
| | - Giacomo Marchi
- Department of Medicine, Section of Internal Medicine, University of Verona, Euro Blood Net Referral Center, Azienda Ospedaliera Universitaria Integrata Verona, Italy
| | - Fabiana Busti
- Department of Medicine, Section of Internal Medicine, University of Verona, Euro Blood Net Referral Center, Azienda Ospedaliera Universitaria Integrata Verona, Italy
| | - Alice Vianello
- Department of Medicine, Section of Internal Medicine, University of Verona, Euro Blood Net Referral Center, Azienda Ospedaliera Universitaria Integrata Verona, Italy
| |
Collapse
|
23
|
The critical roles of iron during the journey from fetus to adolescent: Developmental aspects of iron homeostasis. Blood Rev 2021; 50:100866. [PMID: 34284901 DOI: 10.1016/j.blre.2021.100866] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/12/2022]
Abstract
Iron is indispensable for human life. However, it is also potentially toxic, since it catalyzes the formation of harmful oxidative radicals in unbound form and may facilitate pathogen growth. Therefore, iron homeostasis needs to be tightly regulated. Rapid growth and development require large amounts of iron, while (especially young) children are vulnerable to infections with iron-dependent pathogens due to an immature immune system. Moreover, unbalanced iron status early in life may have effects on the nervous system, immune system and gut microbiota that persist into adulthood. In this narrative review, we assess the critical roles of iron for growth and development and elaborate how the body adapts to physiologically high iron demands during the journey from fetus to adolescent. As a first step towards the development of clinical guidelines for the management of iron disorders in children, we summarize the unmet needs regarding the developmental aspects of iron homeostasis.
Collapse
|
24
|
Breenfeldt Andersen A, Bonne TC, Bejder J, Jung G, Ganz T, Nemeth E, Olsen NV, Huertas JR, Nordsborg NB. Effects of altitude and recombinant human erythropoietin on iron metabolism: a randomized controlled trial. Am J Physiol Regul Integr Comp Physiol 2021; 321:R152-R161. [PMID: 34160288 DOI: 10.1152/ajpregu.00070.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Current markers of iron deficiency (ID), such as ferritin and hemoglobin, have shortcomings, and hepcidin and erythroferrone (ERFE) could be of clinical relevance in relation to early assessment of ID. Here, we evaluate whether exposure to altitude-induced hypoxia (2,320 m) alone, or in combination with recombinant human erythropoietin (rHuEPO) treatment, affects hepcidin and ERFE levels before alterations in routine ID biomarkers and stress erythropoiesis manifest. Two interventions were completed, each comprising a 4-wk baseline, a 4-wk intervention at either sea level or altitude, and a 4-wk follow-up. Participants (n = 39) were randomly assigned to 20 IU·kg body wt-1 rHuEPO or placebo injections every second day for 3 wk during the two intervention periods. Venous blood was collected weekly. Altitude increased ERFE (P ≤ 0.001) with no changes in hepcidin or routine iron biomarkers, making ERFE of clinical relevance as an early marker of moderate hypoxia. rHuEPO treatment at sea level induced a similar pattern of changes in ERFE (P < 0.05) and hepcidin levels (P < 0.05), demonstrating the impact of accelerated erythropoiesis and not of other hypoxia-induced mechanisms. Compared with altitude alone, concurrent rHuEPO treatment and altitude exposure induced additive changes in hepcidin (P < 0.05) and ERFE (P ≤ 0.001) parallel with increases in hematocrit (P < 0.001), demonstrating a relevant range of both hepcidin and ERFE. A poor but significant correlation between hepcidin and ERFE was found (R2 = 0.13, P < 0.001). The findings demonstrate that hepcidin and ERFE are more rapid biomarkers of changes in iron demands than routine iron markers. Finally, ERFE and hepcidin may be sensitive markers in an antidoping context.
Collapse
Affiliation(s)
| | - Thomas C Bonne
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jacob Bejder
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Grace Jung
- Department of Medicine and Pathology, Center for Iron Disorders, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Tomas Ganz
- Department of Medicine and Pathology, Center for Iron Disorders, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Elizabeta Nemeth
- Department of Medicine and Pathology, Center for Iron Disorders, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Niels Vidiendal Olsen
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Jesús Rodríguez Huertas
- Department of Physiology, Faculty of Sport Sciences, Institute of Nutrition and Food Technology, Biomedical Research Centre, University of Granada, Armilla, Spain
| | | |
Collapse
|
25
|
Sanyear C, Chiawtada B, Butthep P, Svasti S, Fucharoen S, Masaratana P. The hypoferremic response to acute inflammation is maintained in thalassemia mice even under parenteral iron loading. PeerJ 2021; 9:e11367. [PMID: 33987030 PMCID: PMC8092106 DOI: 10.7717/peerj.11367] [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: 12/11/2020] [Accepted: 04/07/2021] [Indexed: 11/20/2022] Open
Abstract
Background Hepcidin controls iron homeostasis by inducing the degradation of the iron efflux protein, ferroportin (FPN1), and subsequently reducing serum iron levels. Hepcidin expression is influenced by multiple factors, including iron stores, ineffective erythropoiesis, and inflammation. However, the interactions between these factors under thalassemic condition remain unclear. This study aimed to determine the hypoferremic and transcriptional responses of iron homeostasis to acute inflammatory induction by lipopolysaccharide (LPS) in thalassemic (Hbbth3/+) mice with/without parenteral iron loading with iron dextran. Methods Wild type and Hbbth3/+ mice were intramuscularly injected with 5 mg of iron dextran once daily for two consecutive days. After a 2-week equilibration, acute inflammation was induced by an intraperitoneal injection of a single dose of 1 µg/g body weight of LPS. Control groups for both iron loading and acute inflammation received equal volume(s) of saline solution. Blood and tissue samples were collected at 6 hours after LPS (or saline) injection. Iron parameters and mRNA expression of hepcidin as well as genes involved in iron transport and metabolism in wild type and Hbbth3/+ mice were analyzed and compared by Kruskal–Wallis test with pairwise Mann–Whitney U test. Results We found the inductive effects of LPS on liver IL-6 mRNA expression to be more pronounced under parenteral iron loading. Upon LPS administration, splenic erythroferrone (ERFE) mRNA levels were reduced only in iron-treated mice, whereas, liver bone morphogenetic protein 6 (BMP6) mRNA levels were decreased under both control and parenteral iron loading conditions. Despite the altered expression of the aforementioned hepcidin regulators, the stimulatory effect of LPS on hepcidin mRNA expression was blunt in iron-treated Hbbth3/+ mice. Contrary to the blunted hepcidin response, LPS treatment suppressed FPN1 mRNA expression in the liver, spleen, and duodenum, as well as reduced serum iron levels of Hbbth3/+ mice with parenteral iron loading. Conclusion Our study suggests that a hypoferremic response to LPS-induced acute inflammation is maintained in thalassemic mice with parenteral iron loading in a hepcidin-independent manner.
Collapse
Affiliation(s)
- Chanita Sanyear
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Buraporn Chiawtada
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Punnee Butthep
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Saovaros Svasti
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Suthat Fucharoen
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Patarabutr Masaratana
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
26
|
Iron deficiency: a modern primer to diagnosis and management. Curr Opin Gastroenterol 2021; 37:121-127. [PMID: 33315792 DOI: 10.1097/mog.0000000000000702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
PURPOSE OF REVIEW Iron deficiency with anemia (IDA) and without anemia remain a diagnostic and management challenge. Iron deficiency has a broad spectrum of causes, including gastrointestinal malignancy. The purpose of this review is to summarize the value and limitations of current methods to diagnose iron deficiency and underline the relevance of contemporaneous evidence to guide the pretest probability of gastrointestinal disease. RECENT FINDINGS A number of biomarkers for iron deficiency exist, and all have their caveats. Serum ferritin remains the most pragmatic means of diagnosing iron deficiency. Hepcidin holds future promise as a marker of iron status during inflammatory states. Men and postmenopausal women with IDA have the highest overall prevalence of gastrointestinal malignancy (∼11%), while premenopausal women with IDA (<1.5%) and those with iron deficiency without anemia (<0.5%) have a very low risk. Noninvasive investigation with fecal immunochemical test and fecal calprotectin hold promise to guide further investigations in lower risk groups. SUMMARY Confirmation of iron deficiency remains a challenge. Appropriate risk stratification is the key to guiding judicious gastrointestinal investigation. Use of noninvasive tests may play an important role in lower risk groups. Risk prediction tools applicable to relevant populations are required.
Collapse
|
27
|
Stover PJ, Garza C, Durga J, Field MS. Emerging Concepts in Nutrient Needs. J Nutr 2020; 150:2593S-2601S. [PMID: 33000157 PMCID: PMC7527270 DOI: 10.1093/jn/nxaa117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/12/2020] [Accepted: 04/02/2020] [Indexed: 12/14/2022] Open
Abstract
Dietary reference intakes (DRIs) are quantitative, nutrient intake-based standards used for assessing the diets and specific nutrient intakes of healthy individuals and populations and for informing national nutrition policy and nutrition programs. Because nutrition needs vary by age, sex, and physiological state, DRIs are often specified for healthy subgroups within a population. Diet is known to be the leading modifiable risk factor for chronic disease, and the prevalence of chronic disease is growing in all populations globally and across all subgroups, but especially in older adults. It is known that nutrient needs can change in some chronic disease and other clinical states. Disease states and/or disease treatment can cause whole-body or tissue-specific nutrient depletion or excess, resulting in the need for altered nutrient intakes. In other cases, disease-related biochemical dysfunction can result in a requirement for a nonessential nutrient, rendering it as conditionally essential, or result in toxicity for a food component at levels usually tolerated by healthy people, as seen in inborn errors of metabolism. Here we summarize examples from a growing body of literature of disease-altering nutrient requirements, supporting the need to give more consideration to special nutrient requirements in disease states.
Collapse
Affiliation(s)
| | - Cutberto Garza
- Division of Nutritional Sciences, Cornell University, Ithaca, NY
| | - Jane Durga
- Division of Nutritional Sciences, Cornell University, Ithaca, NY
| | - Martha S Field
- Division of Nutritional Sciences, Cornell University, Ithaca, NY
| |
Collapse
|
28
|
How Severe Anaemia Might Influence the Risk of Invasive Bacterial Infections in African Children. Int J Mol Sci 2020; 21:ijms21186976. [PMID: 32972031 PMCID: PMC7555399 DOI: 10.3390/ijms21186976] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/04/2020] [Accepted: 09/15/2020] [Indexed: 12/21/2022] Open
Abstract
Severe anaemia and invasive bacterial infections are common causes of childhood sickness and death in sub-Saharan Africa. Accumulating evidence suggests that severely anaemic African children may have a higher risk of invasive bacterial infections. However, the mechanisms underlying this association remain poorly described. Severe anaemia is characterized by increased haemolysis, erythropoietic drive, gut permeability, and disruption of immune regulatory systems. These pathways are associated with dysregulation of iron homeostasis, including the downregulation of the hepatic hormone hepcidin. Increased haemolysis and low hepcidin levels potentially increase plasma, tissue and intracellular iron levels. Pathogenic bacteria require iron and/or haem to proliferate and have evolved numerous strategies to acquire labile and protein-bound iron/haem. In this review, we discuss how severe anaemia may mediate the risk of invasive bacterial infections through dysregulation of hepcidin and/or iron homeostasis, and potential studies that could be conducted to test this hypothesis.
Collapse
|
29
|
Abdel-Razeq H, Saadeh SS, Malhis R, Yasser S, Abdulelah H, Eljaber R, Kleib A, Ismael R. Treatment of anemia in cancer patients undergoing chemotherapy with intravenous ferric carboxymaltose without erythropoiesis-stimulating agents. Ther Adv Med Oncol 2020; 12:1758835920953292. [PMID: 32952616 PMCID: PMC7485004 DOI: 10.1177/1758835920953292] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Anemia is commonly encountered in cancer patients receiving active
chemotherapy. Due to adverse events and presumed negative effects on
disease-progression and survival, erythropoiesis-stimulating agents are not
frequently used. In this study, we assess the efficacy and safety of
intravenous ferric carboxymaltose (FCM) to treat cancer-induced anemia
(CIA). Patients and Methods: We recruited adult cancer patients on active chemotherapy with a hemoglobin
(Hb) level ⩽11.0 g/dL. Based on serum ferritin (sFr) and transferrin
saturation (TSAT), patients were divided into 3 groups: group I (absolute
iron deficiency, n = 26) with sFr < 30 ng/mL and
TSAT < 20%; group II (functional iron deficiency,
n = 24) with sFr 30–800 ng/mL and TSAT < 20%; and
patients with TSAT ⩾ 20% were placed in group III as “others”
(n = 34). All patients were treated with intravenous
FCM. Serum hepcidin and C-reactive protein were used as biomarkers to
predict response. Results: A total of 84 patients with a median age (SD) of 53.8 (10.6) were recruited.
Baseline median Hb level was 10.2 (range: 8.3–11.0) gm/dL. At week 12, there
was a significant increment in Hb level for patients in groups I and II
(median increment: 2.35 and 1.5 gm/dL, respectively), with limited response
observed in group III, and most of the increment noted as early as week 3
(⩾1.0 g/dL). Responders tended to have lower levels of hepcidin. No
clinically significant adverse events were reported; however, asymptomatic
hypophosphatemia was observed in 39 (46.4%) patients. Conclusions: Intravenous FCM is a safe and effective treatment option for the management
of a subgroup of patients with CIA. The study was registered at ClinicalTrials.gov [Identifier: NCT04246021]
Collapse
Affiliation(s)
| | - Salwa S. Saadeh
- Department of Medical oncology, King Hussein
Cancer Center, Amman, Jordan
| | - Razan Malhis
- Department of Medical oncology, King Hussein
Cancer Center, Amman, Jordan
| | - Sameer Yasser
- Department of Medical oncology, King Hussein
Cancer Center, Amman, Jordan
| | - Hazem Abdulelah
- Department of Medical oncology, King Hussein
Cancer Center, Amman, Jordan
| | - Rana Eljaber
- Department of Pharmacy, King Hussein Cancer
Center, Amman, Jordan
| | - Amer Kleib
- Department of Nursing, King Hussein Cancer
Center, Amman, Jordan
| | - Rouba Ismael
- Office of Scientific Affairs and Research, King
Hussein Cancer Center, Amman, Jordan
| |
Collapse
|
30
|
Nakanishi T, Kuragano T. Potential hazards of recent trends in liberal iron use for renal anemia. Clin Kidney J 2020; 14:59-69. [PMID: 33564406 PMCID: PMC7857828 DOI: 10.1093/ckj/sfaa117] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 04/30/2020] [Indexed: 12/17/2022] Open
Abstract
A randomized controlled trial,the Proactive IV Iron Therapy in Haemodialysis Patients (PIVOTAL), has recently shown that a high-dose (‘proactive’) intravenous iron regimen was superior to a low-dose (‘reactive’) regimen for hemodialysis patient outcomes and overall safety. However, even in the low-dose group, a substantial amount of iron was administered to maintain serum ferritin >200 ng/mL. This type of comparison may have strongly affected the safety results. Iron has two opposite effects on erythropoiesis: it activates erythroid differentiation directly by supplying iron but inhibits it indirectly by stimulating hepcidin and enhancing oxidative stress. Hepcidin plays an essential role not only in iron homeostasis and the anemia of chronic kidney disease, but also in its complications such as atherosclerosis and infection. Its main stimulation by iron—and to a lesser degree by inflammation—should urge clinicians to avoid prescribing excessive amounts of iron. Furthermore, as serum ferritin is closely correlated with serum hepcidin and iron storage, it would seem preferable to rely mainly on serum ferritin to adjust iron administration, defining an upper limit for risk reduction. Based on our estimations, the optimal range of serum ferritin is ∼50–150 ng/mL, which is precisely within the boundaries of iron management in Japan. Considering the contrasting ranges of target ferritin levels between end-stage renal disease patients in Japan and the rest of the world, the optimal range proposed by us will probably be considered as unacceptable by nephrologists abroad. Only well-balanced, randomized controlled trials with both erythropoiesis-stimulating agents and iron will allow us to settle this controversy.
Collapse
Affiliation(s)
- Takeshi Nakanishi
- Department of Nephrology, Gojinkai Sumiyoshigawa Hospital, Nishinomiya, Japan
- Division of Kidney and Dialysis, Department of Cardiovascular and Renal Medicine, Nishinomiya, Japan
- Correspondence to: Takeshi Nakanishi; E-mail:
| | - Takahiro Kuragano
- Division of Kidney and Dialysis, Department of Cardiovascular and Renal Medicine, Nishinomiya, Japan
| |
Collapse
|
31
|
Viveiros A, Panzer M, Baumgartner N, Schaefer B, Finkenstedt A, Henninger B, Theurl I, Nachbaur K, Weiss G, Haubner R, Decristoforo C, Tilg H, Zoller H. Reduced iron export associated with hepcidin resistance can explain the iron overload spectrum in ferroportin disease. Liver Int 2020; 40:1941-1951. [PMID: 32450003 PMCID: PMC7496278 DOI: 10.1111/liv.14539] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/14/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Ferroportin disease (FD) and hemochromatosis type 4 (HH4) are associated with variants in the ferroportin-encoding gene SLC40A1. Both phenotypes are characterized by iron overload despite being caused by distinct variants that either mediate reduced cellular iron export in FD or resistance against hepcidin-induced inactivation of ferroportin in HH4. The aim of this study was to assess if reduced iron export also confers hepcidin resistance and causes iron overload in FD associated with the R178Q variant. METHODS The ferroportin disease variants R178Q andA77D and the HH4-variant C326Y were overexpressed in HEK-293T cells and subcellular localization was characterized by confocal microscopy and flow cytometry. Iron export and cytosolic ferritin were measured as markers of iron transport and radioligand binding studies were performed. The hepcidin-ferroportin axis was assessed by ferritin/hepcidin correlation in patients with different iron storage diseases. RESULTS In the absence of hepcidin, the R178Q and A77D variants exported less iron when compared to normal and C326Y ferroportin. In the presence of hepcidin, the R178Q and C326Y, but not the A77D-variant, exported more iron than cells expressing normal ferroportin. Regression analysis of serum hepcidin and ferritin in patients with iron overload are compatible with hepcidin deficiency in HFE hemochromatosis and hepcidin resistance in R178Q FD. CONCLUSIONS These results support a novel concept that in certain FD variants reduced iron export and hepcidin resistance could be interlinked. Evasion of mutant ferroportin from hepcidin-mediated regulation could result in uncontrolled iron absorption and iron overload despite reduced transport function.
Collapse
Affiliation(s)
- André Viveiros
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Marlene Panzer
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Nadja Baumgartner
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Benedikt Schaefer
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Armin Finkenstedt
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Benjamin Henninger
- Department of RadiologyMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Igor Theurl
- Department of Medicine IIMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Karin Nachbaur
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Günter Weiss
- Department of Medicine IIMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Roland Haubner
- Department of Nuclear MedicineMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Clemens Decristoforo
- Department of Nuclear MedicineMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Herbert Tilg
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| | - Heinz Zoller
- Department of Medicine IMedical University and University Hospital of InnsbruckInnsbruckAustria
| |
Collapse
|
32
|
The changing landscape of iron deficiency. Mol Aspects Med 2020; 75:100861. [PMID: 32418671 DOI: 10.1016/j.mam.2020.100861] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 12/25/2022]
Abstract
Iron deficiency (ID) with or without anemia is common worldwide. ID is a broad definition encompassing decreased total body iron (absolute deficiency) as well as reduced iron supply to erythropoietic and/or other organs with preserved stores (functional iron deficiency, FID), as it occurs in inflammation. Increased iron needs unbalanced by iron supply, low iron intake, reduced absorption and chronic blood loss, often in combination, are the main causes of absolute ID, easily diagnosed by low ferritin levels. In all these cases hepcidin synthesis is repressed, while in FID is augmented by inflammatory cytokines, causing iron sequestration in stores. Because of increased ferritin levels diagnosis of ID in the latter condition may be tricky: global clinical evaluation, accepted threshold of iron tests together with response to iron treatment may be of help. Search and removal of the responsible cause(s) is as important as diagnosing ID or FID. The response to oral iron treatment is suboptimal when hepcidin levels are high. Future research is needed to establish/validate markers for improved diagnosis of complex cases and to test the therapeutic value of drugs under development aimed at interfering with the altered iron trafficking.
Collapse
|
33
|
Abstract
Iron deficiency anaemia is a global health concern affecting children, women and the elderly, whilst also being a common comorbidity in multiple medical conditions. The aetiology is variable and attributed to several risk factors decreasing iron intake and absorption or increasing demand and loss, with multiple aetiologies often coexisting in an individual patient. Although presenting symptoms may be nonspecific, there is emerging evidence on the detrimental effects of iron deficiency anaemia on clinical outcomes across several medical conditions. Increased awareness about the consequences and prevalence of iron deficiency anaemia can aid early detection and management. Diagnosis can be easily made by measurement of haemoglobin and serum ferritin levels, whilst in chronic inflammatory conditions, diagnosis may be more challenging and necessitates consideration of higher serum ferritin thresholds and evaluation of transferrin saturation. Oral and intravenous formulations of iron supplementation are available, and several patient and disease-related factors need to be considered before management decisions are made. This review provides recent updates and guidance on the diagnosis and management of iron deficiency anaemia in multiple clinical settings.
Collapse
Affiliation(s)
- M D Cappellini
- Department of Clinical Sciences and Community, IRCCS Ca' Granda Foundation Maggiore Policlinico Hospital, University of Milan, Milan, Italy
| | | | - A T Taher
- Department of Internal Medicine, American University of Beirut Medical Centre, Beirut, Lebanon
| |
Collapse
|
34
|
Sangkhae V, Nemeth E. To induce or not to induce: the fight over hepcidin regulation. Haematologica 2019; 104:1093-1095. [PMID: 31152085 DOI: 10.3324/haematol.2019.216960] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Veena Sangkhae
- Center for Iron Disorders, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Elizabeta Nemeth
- Center for Iron Disorders, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
35
|
Ginzburg YZ. New diagnostic tools for delineating iron status. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:327-336. [PMID: 31808893 PMCID: PMC6913443 DOI: 10.1182/hematology.2019000035] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Recent advances in our understanding of iron metabolism regulation and crosstalk with erythropoiesis have provided insight into the pathophysiology of multiple disease conditions. For instance, the peptide hormone hepcidin is central to the regulation of iron metabolism. Its effect on cellular iron concentration involves binding ferroportin, the main iron export protein, resulting in its internalization and degradation and leading to iron sequestration within ferroportin-expressing cells. Furthermore, hepcidin regulation by erythropoiesis is attributed in large part to a bone marrow-derived hormone erythroferrone. Erythroferrone-induced hepcidin suppression in diseases of expanded hematopoiesis results in iron overload. Conversely, diseases, such as iron refractory iron deficiency anemia and anemia of chronic inflammation, are characterized by aberrantly increased hepcidin, resulting in iron sequestration and decreased circulating iron and eventually leading to iron-restricted erythropoiesis. Lastly, because iron functions in concert with erythropoietin to promote erythroid precursor survival, proliferation, and differentiation, iron deficiency anemia is a consequence not only of decreased hemoglobin synthesis in each cell but also, a decrease in erythropoietin responsiveness in the bone marrow. How to translate this new information to the clinical setting has not been fully elucidated. The purpose of this manuscript is to summarize current standard tools for identifying iron deficiency in anemic patients; explore the tools and context for evaluating novel markers, such as hepcidin, erythroferrone, and markers of the iron restriction response; and assess available evidence for how their use could increase our understanding of health outcomes in clinically challenging cases.
Collapse
|
36
|
Zhang Y, Zuo Y, Li B, Xie J, Ma Z, Thirupathi A, Yu P, Gao G, Shi M, Zhou C, Xu H, Chang Y, Shi Z. Propofol prevents oxidative stress and apoptosis by regulating iron homeostasis and targeting JAK/STAT3 signaling in SH-SY5Y cells. Brain Res Bull 2019; 153:191-201. [PMID: 31472185 DOI: 10.1016/j.brainresbull.2019.08.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/09/2019] [Accepted: 08/26/2019] [Indexed: 12/11/2022]
Abstract
The present study aimed to test the hypothesis that propofol (PRO) could exert a neuroprotective effect via inhibiting oxidative stress induced by iron accumulation. Human SH-SY5Y cells were pretreated with ferric citrate (FAC), and then were protected by PRO. Cell viability was measured by MTT method. Iron levels were assayed by ICP-MS. Cell apoptosis was examined by TUNEL and digital holographic technique. Malondialdehyde (MDA), reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) depolarization were measured by MDA, DCFH-DA and JC-1 kits, respectively. The expression of proteins or genes involved in iron metabolism such as ferritin, TfR1, DMT1, Fpn1 and hepcidin, and other apoptosis-related proteins including Bcl2, Bax, Bid, Cox2, IL-6, JAK1 and STAT3 were detected by western blot. Our results showed low concentration of PRO (5 μM) could significantly prevent FAC induced apoptosis via inhibiting oxidative stress and iron accumulation. PRO suppressed the increase of ROS and MDA and decrease of MMP induced by FAC. PRO significantly down-regulated the expression of ferritin and up-regulated the expression of TfR1and Fpn1, but had no effect of DMT1. Furthermore, this effect was not done by PRO chelating iron. Meanwhile, PRO suppressed the inflammatory response through inhibiting IL-6 and Cox2 expression and activating JAK/STAT3 signaling induced by iron overload. In conclusion, here we demonstrated a new antioxidation mechanism of PRO. PRO could protect against nerve cell injury induced by overload of iron through regulating iron metabolism and inhibiting stress oxidative and inflammation reaction pathways by targeting JAK/STAT3 signaling.
Collapse
Affiliation(s)
- Ying Zhang
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei Province, China
| | - Yong Zuo
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei Province, China
| | - Bowen Li
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei Province, China
| | - Jinhong Xie
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei Province, China
| | - Zhao Ma
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei Province, China
| | - Anand Thirupathi
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei Province, China
| | - Peng Yu
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei Province, China
| | - Guofen Gao
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei Province, China
| | - Mengtong Shi
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei Province, China
| | - Changhao Zhou
- First Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Hongmeng Xu
- Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Yanzhong Chang
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei Province, China.
| | - Zhenhua Shi
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang 050024, Hebei Province, China.
| |
Collapse
|
37
|
Cross JH, Jarjou O, Mohammed NI, Prentice AM, Cerami C. Neonatal iron distribution and infection susceptibility in full term, preterm and low birthweight babies in urban Gambia: study protocol for an observational study. Gates Open Res 2019; 3:1469. [PMID: 31588425 PMCID: PMC6757319 DOI: 10.12688/gatesopenres.12963.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Neonatal infection is the third largest cause of death in children under five worldwide. Nutritional immunity is the process by which the host innate immune system limits nutrient availability to invading organisms. Iron is an essential micronutrient for both microbial pathogens and their mammalian hosts. Changes in iron availability and distribution have significant effects on pathogen virulence and on the immune response to infection. Our previously published data shows that, during the first 24 hours of life, full-term neonates have reduced overall serum iron. Transferrin saturation decreases rapidly from 45% in cord blood to ~20% by six hours post-delivery. Methods: To study neonatal nutritional immunity and its role in neonatal susceptibility to infection, we will conduct an observational study on 300 full-term normal birth weight (FTB+NBW), 50 preterm normal birth weight (PTB+NBW), 50 preterm low birth weight (PTB+LBW) and 50 full-term low birth weight (FTB+LBW), vaginally-delivered neonates born at Kanifing General Hospital, The Gambia. We will characterize and quantify iron-related nutritional immunity during the early neonatal period and use ex vivo sentinel bacterial growth assays to assess how differences in serum iron affect bacterial growth. Blood samples will be collected from the umbilical cord (arterial and venous) and at serial time points from the neonates over the first week of life. Discussion: Currently, little is known about nutritional immunity in neonates. In this study, we will increase understanding of how nutritional immunity may protect neonates from infection during the first critical days of life by limiting the pathogenicity and virulence of neonatal sepsis causing organisms by reducing the availability of iron. Additionally, we will investigate the hypothesis that this protective mechanism may not be activated in preterm and low birth weight neonates, potentially putting these babies at an enhanced risk of neonatal infection. Trial registration: clinicaltrials.gov ( NCT03353051) 27/11/2017.
Collapse
Affiliation(s)
- James H. Cross
- MRC Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Ousman Jarjou
- MRC Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | | | - Andrew M. Prentice
- MRC Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Carla Cerami
- MRC Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| |
Collapse
|
38
|
Affiliation(s)
- Tomas Ganz
- From the Departments of Medicine and Pathology, David Geffen School of Medicine at UCLA, Los Angeles
| |
Collapse
|
39
|
Weiss G. Does iron let boys grow faster?! Haematologica 2019; 104:1503-1505. [PMID: 31366461 DOI: 10.3324/haematol.2019.222018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Günter Weiss
- Department of Internal Medicine II, Innsbruck Medical University.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Innsbruck, Austria
| |
Collapse
|
40
|
The diagnostic potential of the iron-regulatory hormone hepcidin. Hemasphere 2019; 3:HemaSphere-2019-0026. [PMID: 35309797 PMCID: PMC8925696 DOI: 10.1097/hs9.0000000000000236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/21/2019] [Accepted: 03/26/2019] [Indexed: 01/12/2023] Open
|
41
|
Cross JH, Jarjou O, Mohammed NI, Prentice AM, Cerami C. Neonatal iron distribution and infection susceptibility in full term, preterm and low birthweight babies in urban Gambia: study protocol for an observational study. Gates Open Res 2019; 3:1469. [PMID: 31588425 PMCID: PMC6757319 DOI: 10.12688/gatesopenres.12963.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2019] [Indexed: 10/15/2023] Open
Abstract
Background: Neonatal infection is the third largest cause of death in children under five worldwide. Nutritional immunity is the process by which the host innate immune system limits nutrient availability to invading organisms. Iron is an essential micronutrient for both microbial pathogens and their mammalian hosts. Changes in iron availability and distribution have significant effects on pathogen virulence and on the immune response to infection. Our previously published data shows that, during the first 24 hours of life, full-term neonates have reduced overall serum iron. Transferrin saturation decreases rapidly from 45% in cord blood to ~20% by six hours post-delivery. Methods: To study neonatal nutritional immunity and its role in neonatal susceptibility to infection, we will conduct an observational study on 300 full-term normal birth weight (FTB+NBW), 50 preterm normal birth weight (PTB+NBW), 50 preterm low birth weight (PTB+LBW) and 50 full-term low birth weight (FTB+LBW), vaginally-delivered neonates born at Kanifing General Hospital, The Gambia. We will characterize and quantify iron-related nutritional immunity during the early neonatal period and use ex vivo sentinel bacterial growth assays to assess how differences in serum iron affect bacterial growth. Blood samples will be collected from the umbilical cord (arterial and venous) and at serial time points from the neonates over the first week of life. Discussion: Currently, little is known about nutritional immunity in neonates. In this study, we will increase understanding of how nutritional immunity may protect neonates from infection during the first critical days of life by limiting the pathogenicity and virulence of neonatal sepsis causing organisms by reducing the availability of iron. Additionally, we will investigate the hypothesis that this protective mechanism may not be activated in preterm and low birth weight neonates, potentially putting these babies at an enhanced risk of neonatal infection. Trial registration: clinicaltrials.gov ( NCT03353051) 27/11/2017.
Collapse
Affiliation(s)
- James H. Cross
- MRC Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Ousman Jarjou
- MRC Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | | | - Andrew M. Prentice
- MRC Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Carla Cerami
- MRC Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| |
Collapse
|
42
|
Armitage AE, Moretti D. The Importance of Iron Status for Young Children in Low- and Middle-Income Countries: A Narrative Review. Pharmaceuticals (Basel) 2019; 12:E59. [PMID: 30995720 PMCID: PMC6631790 DOI: 10.3390/ph12020059] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/09/2019] [Accepted: 04/12/2019] [Indexed: 12/21/2022] Open
Abstract
Early childhood is characterised by high physiological iron demand to support processes including blood volume expansion, brain development and tissue growth. Iron is also required for other essential functions including the generation of effective immune responses. Adequate iron status is therefore a prerequisite for optimal child development, yet nutritional iron deficiency and inflammation-related iron restriction are widespread amongst young children in low- and middle-income countries (LMICs), meaning iron demands are frequently not met. Consequently, therapeutic iron interventions are commonly recommended. However, iron also influences infection pathogenesis: iron deficiency reduces the risk of malaria, while therapeutic iron may increase susceptibility to malaria, respiratory and gastrointestinal infections, besides reshaping the intestinal microbiome. This means caution should be employed in administering iron interventions to young children in LMIC settings with high infection burdens. In this narrative review, we first examine demand and supply of iron during early childhood, in relation to the molecular understanding of systemic iron control. We then evaluate the importance of iron for distinct aspects of physiology and development, particularly focusing on young LMIC children. We finally discuss the implications and potential for interventions aimed at improving iron status whilst minimising infection-related risks in such settings. Optimal iron intervention strategies will likely need to be individually or setting-specifically adapted according to iron deficiency, inflammation status and infection risk, while maximising iron bioavailability and considering the trade-offs between benefits and risks for different aspects of physiology. The effectiveness of alternative approaches not centred around nutritional iron interventions for children should also be thoroughly evaluated: these include direct targeting of common causes of infection/inflammation, and maternal iron administration during pregnancy.
Collapse
Affiliation(s)
- Andrew E Armitage
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK.
| | - Diego Moretti
- Laboratory of Human Nutrition, Institute of Food Nutrition and Health, Department of Health Sciences and Technology, ETH Zürich, CH-8092 Zürich, Switzerland.
- Nutrition Group, Health Department, Swiss Distance University of Applied Sciences, CH-8105 Regensdorf, Switzerland.
| |
Collapse
|