1
|
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
|
2
|
Du Pont-Thibodeau G, Li SYH, Ducharme-Crevier L, Jutras C, Pantopoulos K, Farrell C, Roumeliotis N, Harrington K, Thibault C, Roy N, Shah A, Lacroix J, Stanworth SJ. Iron Deficiency in Anemic Children Surviving Critical Illness: Post Hoc Analysis of a Single-Center Prospective Cohort in Canada, 2019-2022. Pediatr Crit Care Med 2024; 25:344-353. [PMID: 38358779 DOI: 10.1097/pcc.0000000000003442] [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] [Indexed: 02/16/2024]
Abstract
OBJECTIVES Many children leave the PICU with anemia. The mechanisms of post-PICU anemia are poorly investigated, and treatment of anemia, other than blood, is rarely started during PICU. We aimed to characterize the contributions of iron depletion (ID) and/or inflammation in the development of post-PICU anemia and to explore the utility of hepcidin (a novel iron marker) at detecting ID during inflammation. DESIGN Post hoc analysis of a single-center prospective study (November 2019 to September 2022). SETTING PICU, quaternary center, Canada. PATIENTS Children admitted to PICU with greater than or equal to 48 hours of invasive or greater than or equal to 96 hours of noninvasive ventilation. We excluded patients with preexisting conditions causing anemia or those admitted after cardiac surgery. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Hematological and iron profiles were performed at PICU discharge on 56 participants of which 37 (37/56) were diagnosed with anemia. Thirty-three children (33/56; 59%) were younger than 2 years. Median Pediatric Logistic Organ Dysfunction score was 11 (interquartile range, 6-16). Twenty-four of the 37 anemic patients had repeat bloodwork 2 months post-PICU. Of those, four (4/24; 16%) remained anemic. Hematologic profiles were categorized as: anemia of inflammation (AI), iron deficiency anemia (IDA), IDA with inflammation, and ID (low iron stores without anemia). Seven (7/47; 15%) had AI at discharge, and one had persistent AI post-PICU. Three patients (3/47; 6%) had IDA at discharge; of which one was lost to follow-up and the other two were no longer anemic but had ID post-PICU. Eleven additional patients developed ID post-PICU. In the exploratory analysis, we identified a diagnostic cutoff value for ID during inflammation from the receiver operating characteristic curve for hepcidin of 31.9 pg/mL. This cutoff would increase the detection of ID at discharge from 6% to 34%. CONCLUSIONS The burden of ID in children post-PICU is high and better management strategies are required. Hepcidin may increase the diagnostic yield of ID in patients with inflammation.
Collapse
Affiliation(s)
| | - Shu Yin Han Li
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | | | - Camille Jutras
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montréal, QC, Canada
| | - Catherine Farrell
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Nadia Roumeliotis
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Karen Harrington
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Céline Thibault
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Noémi Roy
- Department of Hematology, John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
| | - Akshay Shah
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, United Kingdom
| | - Jacques Lacroix
- Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Simon J Stanworth
- Department of Hematology, John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
| |
Collapse
|
3
|
Mwangi MN, Mzembe G, Ngwira CC, Vokhiwa M, Kapulula MD, Larson LM, Braat S, Harding R, McLean ARD, Hamadani JD, Biggs BA, Ataíde R, Phiri KS, Pasricha SR. Protocol for a randomised, multicentre, four-arm, double-blinded, placebo-controlled trial to assess the benefits and safety of iron supplementation with malaria chemoprevention to children in Malawi: IRMA trial. BMJ Open 2023; 13:e069011. [PMID: 37832986 PMCID: PMC10583080 DOI: 10.1136/bmjopen-2022-069011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 09/19/2023] [Indexed: 10/15/2023] Open
Abstract
INTRODUCTION Approximately 40% of children aged 6-59 months worldwide are anaemic. Iron-containing multiple micronutrient powders (MNPs) and iron supplements (syrup/drops) are used to combat anaemia in children in different parts of the world. However, evidence for functional benefits of iron supplementation in children is scarce, and potential risks remain poorly defined, particularly concerning diarrhoea and malaria. This trial aims to determine if: (1) the efficacy of iron supplements or MNPs (containing iron) given with malaria chemoprevention is superior to malaria chemoprevention alone, or (2) if the efficacy of malaria chemoprevention alone is superior to placebo on child cognitive development. METHODS AND ANALYSIS IRMA is a four-arm, parallel-group, double-blinded, placebo-controlled, triple-dummy, randomised trial in Southern Malawi. The study recruits 2168 infants aged 6 months, with an intervention period of 6 months and a post-intervention period of a further 6 months. Children are randomised into four arms: (1) No intervention (placebo); (2) malaria chemoprevention only; (3) MNPs and malaria chemoprevention; and (4) iron syrup and malaria chemoprevention. The primary outcome, cognitive development (Cognitive Composite Score (CogCS)), is measured at the end of the 6 months intervention. Secondary outcomes include CogCS at a further 6 months post-intervention, motor, language and behavioural development, physical growth and prevalence of anaemia and iron deficiency. Safety outcomes include incidence of malaria and other infections, and prevalence of malaria parasitaemia during and post-intervention period. ETHICS AND DISSEMINATION The trial is approved by the National Health Sciences Research Committee (#19/01/2213) (Malawi) and the Human Research Ethics Committee (WEHI: 19/012) (Australia). Written informed consent in the local language is obtained from each participant before conducting any study-related procedure. Results will be shared with the local community and internationally with academic and policy stakeholders. TRIAL REGISTRATION NUMBER ACTRN12620000386932.
Collapse
Affiliation(s)
- Martin N Mwangi
- Training and Research Unit of Excellence (TRUE), Blantyre, Malawi
- The Micronutrient Forum, Healthy Mothers Healthy Babies Consortium, Washington DC, Washington, USA
| | - Glory Mzembe
- Training and Research Unit of Excellence (TRUE), Blantyre, Malawi
- Department of Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | | | - Maclean Vokhiwa
- Training and Research Unit of Excellence (TRUE), Blantyre, Malawi
- Department of Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | | | - Leila M Larson
- Department of Health Promotion, Education, and Behaviour, University of South Carolina Arnold School of Public Health, Columbia, South Carolina, USA
| | - Sabine Braat
- Department of Infectious Diseases at the Peter Doherty Institute of Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Epidemiology and Biostatistics, University of Melbourne School of Population and Global Health, Carlton, Victoria, Australia
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Rebecca Harding
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Alistair R D McLean
- Centre for Epidemiology and Biostatistics, University of Melbourne School of Population and Global Health, Carlton, Victoria, Australia
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Jena D Hamadani
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Beverley-Ann Biggs
- Department of Infectious Diseases at the Peter Doherty Institute of Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
- Diagnostic Haematology, The Royal Melbourne Hospital; and Clinical Haematology, Melbourne, Victoria, Australia
| | - Ricardo Ataíde
- Department of Infectious Diseases at the Peter Doherty Institute of Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Kamija S Phiri
- Training and Research Unit of Excellence (TRUE), Blantyre, Malawi
- Department of Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Sant-Rayn Pasricha
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Diagnostic Haematology, The Royal Melbourne Hospital; and Clinical Haematology, Melbourne, Victoria, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Mohammed NI, Wason J, Mendy T, Nass SA, Ofordile O, Camara F, Baldeh B, Sanyang C, Jallow AT, Hossain I, Faria N, Powell JJ, Prentice AM, Pereira DI. A novel nano-iron supplement versus standard treatment for iron deficiency anaemia in children 6-35 months (IHAT-GUT trial): a double-blind, randomised, placebo-controlled non-inferiority phase II trial in The Gambia. EClinicalMedicine 2023; 56:101853. [PMID: 36880049 PMCID: PMC9985047 DOI: 10.1016/j.eclinm.2023.101853] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Iron deficiency anaemia (IDA) is the leading cause of years lost to disability in most sub-Saharan African countries and is especially common in young children. The IHAT-GUT trial assessed the efficacy and safety of a novel nano iron supplement, which is a dietary ferritin analogue termed iron hydroxide adipate tartrate (IHAT), for the treatment of IDA in children under 3 years of age. METHODS In this single-country, randomised, double-blind, parallel, placebo-controlled, non-inferiority Phase II study in The Gambia, children 6-35 months with IDA (7≤Hb < 11 g/dL and ferritin<30 μg/L) were randomly assigned (1:1:1) to receive either IHAT, ferrous sulphate (FeSO4) or placebo daily for 3 months (85 days). The daily iron dose was 12.5 mg Fe equivalent for FeSO4 and the estimated dose with comparable iron-bioavailability for IHAT (20 mg Fe). The primary efficacy endpoint was the composite of haemoglobin response at day 85 and correction of iron deficiency. The non-inferiority margin was 0.1 absolute difference in response probability. The primary safety endpoint was moderate-severe diarrhoea analysed as incidence density and prevalence over the 3 months intervention. Secondary endpoints reported herein include hospitalisation, acute respiratory infection, malaria, treatment failures, iron handling markers, inflammatory markers, longitudinal prevalence of diarrhoea and incidence density of bloody diarrhoea. Main analyses were per-protocol (PP) and intention-to-treat (ITT) analyses. This trial is registered with clinicaltrials.gov (NCT02941081). FINDINGS Between Nov 2017 and Nov 2018, 642 children were randomised into the study (214 per group) and included in the ITT analysis, the PP population included 582 children. A total of 50/177 (28.2%) children in the IHAT group achieved the primary efficacy endpoint, as compared with 42/190 (22.1%) in the FeSO4 group (OR 1.39, 80% CI 1.01-1.91, PP population) and with 2/186 (1.1%) in the placebo group. Diarrhoea prevalence was similar between groups, with 40/189 (21.2%) children in the IHAT group developing at least one episode of moderate-severe diarrhoea over the 85 days intervention, compared with 47/198 (23.7%) in the FeSO4 group (OR 1.18, 80% CI 0.86-1.62) and 40/195 (20.5%) in the placebo group (OR 0.96, 80% CI 0.7-1.33, PP population). Incidence density of moderate-severe diarrhoea was 2.66 in the IHAT group and 3.42 in the FeSO4 group (RR 0.76, 80% CI 0.59-0.99, CC-ITT population).There were 143/211 (67.8%) children with adverse events (AEs) in the IHAT group, 146/212 (68.9%) in the FeSO4 group and 143/214 (66.8%) in the placebo group. There were overall 213 diarrhoea-related AEs; 35 (28.5%) cases reported in the IHAT group compared with 51 (41.5%) cases in the FeSO4 group and 37 (30.1%) cases in the placebo group. INTERPRETATION In this first Phase II study conducted in young children with IDA, IHAT showed sufficient non-inferiority compared to standard-of-care FeSO4, in terms of ID correction and haemoglobin response, to warrant a definitive Phase III trial. In addition, IHAT had lower incidence of moderate-severe diarrhoea than FeSO4, with no increased adverse events in comparison with placebo. FUNDING The Bill & Melinda Gates Foundation (OPP1140952).
Collapse
Affiliation(s)
- Nuredin I. Mohammed
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, Gambia
| | - James Wason
- MRC Biostatistics Unit, Institute of Public Health, University of Cambridge, Cambridge, CB2 0SR, UK
- Population Health Sciences Institute, Newcastle University, Newcastle, NE2 4BN, UK
| | - Thomas Mendy
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, Gambia
| | - Stefan A. Nass
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, Gambia
- Medical Humanities, Amsterdam-UMC - VUmc Location, Vrije Universiteit, Amsterdam, the Netherlands
| | - Ogochukwu Ofordile
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, Gambia
| | - Famalang Camara
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, Gambia
| | - Bakary Baldeh
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, Gambia
| | - Chilel Sanyang
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, Gambia
| | - Amadou T. Jallow
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, Gambia
| | - Ilias Hossain
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, Gambia
| | - Nuno Faria
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Jonathan J. Powell
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Andrew M. Prentice
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, Gambia
- Corresponding author. MRC Unit The Gambia at London School of Hygiene & Tropical Medicine, Atlantic Boulevard, Fajara, PO Box 273, Banjul, Gambia.
| | - Dora I.A. Pereira
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, Gambia
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| |
Collapse
|
5
|
Vinke JSJ, Altulea DHA, Eisenga MF, Jagersma RL, Niekolaas TM, van Baarle D, Heiden MVD, Steenhuis M, Rispens T, Abdulahad WH, Sanders JSF, De Borst MH. Ferric carboxymaltose and SARS-CoV-2 vaccination-induced immunogenicity in kidney transplant recipients with iron deficiency: The COVAC-EFFECT randomized controlled trial. Front Immunol 2023; 13:1017178. [PMID: 36618359 PMCID: PMC9822258 DOI: 10.3389/fimmu.2022.1017178] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/05/2022] [Indexed: 12/25/2022] Open
Abstract
Background Kidney transplant recipients (KTRs) have an impaired immune response after vaccination against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Iron deficiency (ID) may adversely affect immunity and vaccine efficacy. We aimed to investigate whether ferric carboxymaltose (FCM) treatment improves humoral and cellular responses after SARS-CoV-2 vaccination in iron-deficient KTRs. Methods We randomly assigned 48 iron-deficient KTRs to intravenous FCM (1-4 doses of 500mg with six-week intervals) or placebo. Co-primary endpoints were SARS-CoV-2-specific anti-Receptor Binding Domain (RBD) Immunoglobulin G (IgG) titers and T-lymphocyte reactivity against SARS-CoV-2 at four weeks after the second vaccination with mRNA-1273 or mRNA-BNT162b2. Results At four weeks after the second vaccination, patients receiving FCM had higher plasma ferritin and transferrin saturation (P<0.001 vs. placebo) and iron (P=0.02). However, SARS-CoV-2-specific anti-RBD IgG titers (FCM: 66.51 [12.02-517.59] BAU/mL; placebo: 115.97 [68.86-974.67] BAU/mL, P=0.07) and SARS-CoV-2-specific T-lymphocyte activation (FCM: 93.3 [0.85-342.5] IFN-ɣ spots per 106 peripheral blood mononuclear cells (PBMCs), placebo: 138.3 [0.0-391.7] IFN-ɣ spots per 106 PBMCs, P=0.83) were not significantly different among both arms. After the third vaccination, SARS-CoV-2-specific anti-RBD IgG titers remained similar between treatment groups (P=0.99). Conclusions Intravenous iron supplementation efficiently restored iron status but did not improve the humoral or cellular immune response against SARS-CoV-2 after three vaccinations.
Collapse
Affiliation(s)
| | - Dania H. A. Altulea
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Michele F. Eisenga
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Renate L. Jagersma
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Tessa M. Niekolaas
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands
| | - Debbie van Baarle
- Department of Immunology, University Medical Center Groningen, Groningen, Netherlands
| | | | - Maurice Steenhuis
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands
| | - Wayel H. Abdulahad
- Department of Immunology, University Medical Center Groningen, Groningen, Netherlands
| | | | - Martin H. De Borst
- Department of Nephrology, University Medical Center Groningen, Groningen, Netherlands,*Correspondence: Martin H. De Borst,
| |
Collapse
|
6
|
Wegmüller R, Bah A, Kendall L, Goheen MM, Sanyang S, Danso E, Sise EA, Jallow A, Verhoef H, Jallow MW, Wathuo M, Armitage AE, Drakesmith H, Pasricha SR, Cross JH, Cerami C, Prentice AM. Hepcidin-guided screen-and-treat interventions for young children with iron-deficiency anaemia in The Gambia: an individually randomised, three-arm, double-blind, controlled, proof-of-concept, non-inferiority trial. Lancet Glob Health 2023; 11:e105-e116. [PMID: 36521942 PMCID: PMC9764454 DOI: 10.1016/s2214-109x(22)00449-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 09/22/2022] [Accepted: 10/04/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Iron deficiency is the most prevalent nutritional disorder worldwide. Iron supplementation has modest efficacy, causes gastrointestinal side-effects that limit compliance, and has been associated with serious adverse outcomes in children across low-income settings. We aimed to compare two hepcidin-guided screen-and-treat regimens designed to reduce overall iron dosage by targeting its administration to periods when children were safe and ready to receive iron supplementation, with WHO's recommendation of universal iron supplementation. METHODS We conducted an individually randomised, three-arm, double-blind, controlled, proof-of-concept, non-inferiority trial in 12 rural communities across The Gambia. Eligible participants were children aged 6-23 months with anaemia. Participants were randomly assigned (1:1:1) to either the WHO recommended regimen of one sachet of multiple micronutrient powder (MMP) daily containing 12·0 mg iron as encapsulated ferrous fumarate (control group); to MMP with 12·0 mg per day iron for the next 7 days if plasma hepcidin concentration was less than 5·5 μg/L, or to MMP without iron for the next 7 days if plasma hepcidin concentration was at least 5·5 μg/L (12 mg screen-and-treat group); or to MMP with 6·0 mg per day iron for the next 7 days if plasma hepcidin concentration was less than 5·5 μg/L, or to MMP without iron for the next 7 days if plasma hepcidin concentration was at least 5·5 μg/L (6 mg screen-and-treat group). Randomisation was done by use of a permuted block design (block size of 9), with stratification by haemoglobin and age, using computer-generated numbers. Participants and the research team (except for the data manager) were masked to group allocation. The primary outcome was haemoglobin concentration, with a non-inferiority margin of -5 g/L. A per-protocol analysis, including only children who had consumed at least 90% of the supplements (ie, supplement intake on ≥75 days during the study), was done to assess non-inferiority of the primary outcome at day 84 using a one-sided t test adjusted for multiple comparisons. Safety was assessed by use of ex-vivo growth tests of Plasmodium falciparum in erythrocytes and three species of sentinel bacteria in plasma samples from participants. This trial is registered with the ISRCTN registry, ISRCTN07210906. FINDINGS Between April 23, 2014, and Aug 7, 2015, we prescreened 783 children, of whom 407 were enrolled into the study: 135 were randomly assigned to the control group, 136 to the 12 mg screen-and-treat group, and 136 to the 6 mg screen-and-treat group. 345 (85%) children were included in the per-protocol population: 115 in the control group, 116 in the 12 mg screen-and-treat group, and 114 in the 6 mg screen-and-treat group. Directly observed adherence was high across all groups (control group 94·8%, 12 mg screen-and-treat group 95·3%, and 6 mg screen-and-treat group 95·0%). 82 days of iron supplementation increased mean haemoglobin concentration by 7·7 g/L (95% CI 3·2 to 12·2) in the control group. Both screen-and-treat regimens were significantly less efficacious at improving haemoglobin (-5·6 g/L [98·3% CI -9·9 to -1·3] in the 12 mg screen-and-treat group and -7·8 g/L [98·3% CI -12·2 to -3·5] in the 6 mg screen-and-treat group) and neither regimen met the preset non-inferiority margin of -5 g/L. The 12 mg screen-and-treat regimen reduced iron dosage to 6·1 mg per day and the 6 mg screen-and-treat regimen reduced dosage to 3·0 mg per day. 580 adverse events were observed in 316 participants, of which eight were serious adverse events requiring hospitalisation mainly due to diarrhoeal disease (one [1%] participant in the control group, three [2%] in the 12 mg screen-and-treat group, and four [3%] in the 6 mg screen-and-treat group). The most common causes of non-serious adverse events (n=572) were diarrhoea (145 events [25%]), upper respiratory tract infections (194 [34%]), lower respiratory tract infections (62 [11%]), and skin infections (122 [21%]). No adverse events were deemed to be related to the study interventions. INTERPRETATION The hepcidin-guided screen-and-treat strategy to target iron administration succeeded in reducing overall iron dosage, but was considerably less efficacious at increasing haemoglobin and combating iron deficiency and anaemia than was WHO's standard of care, and showed no differences in morbidity or safety outcomes. FUNDING Bill & Melinda Gates Foundation and UK Medical Research Council.
Collapse
Affiliation(s)
- Rita Wegmüller
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia; GroundWork, Fläsch, Switzerland
| | - Amat Bah
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia; National Nutrition Agency, Bakau, The Gambia
| | - Lindsay Kendall
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Morgan M Goheen
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia; Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Saikou Sanyang
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Ebrima Danso
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Ebrima A Sise
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Amadou Jallow
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Hans Verhoef
- Wageningen University & Research, Wageningen, Netherlands
| | - Momodou W Jallow
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia; Regeneron Genetics Center, Tarrytown, NY, USA
| | - Miriam Wathuo
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia; One Acre Fund, Kigali, Rwanda
| | - Andrew E Armitage
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Hal Drakesmith
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Sant-Rayn Pasricha
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - James H Cross
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Carla Cerami
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Andrew M Prentice
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia.
| |
Collapse
|
7
|
González-Fernández D, Nemeth E, Pons EDC, Sinisterra OT, Rueda D, Starr L, Sangkhae V, Murillo E, Scott ME, Koski KG. Multiple Indicators of Undernutrition, Infection, and Inflammation in Lactating Women Are Associated with Maternal Iron Status and Infant Anthropometry in Panama: The MINDI Cohort. Nutrients 2022; 14:nu14173497. [PMID: 36079755 PMCID: PMC9460351 DOI: 10.3390/nu14173497] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 02/06/2023] Open
Abstract
Maternal infections, nutrient deficiencies, and inflammation (MINDI) co-exist in lactating indigenous women in Panama, but their impact on maternal iron status and infant growth is unknown. For this secondary analysis of cross-sectional data of lactating mothers from our MINDI cohort, we investigated associations of MINDI variables with maternal anemia, elevated serum transferrin receptor (sTfR), low serum iron, hepcidin, ferritin, and infant weight-for-age (WAZ), length-for-age (LAZ), and head-circumference-for-age (HCAZ) Z-scores in 99 mother-infant dyads. A bootstrapping resampling procedure preselected covariates for inclusion in multivariable regressions models from chronic maternal infections and nutritional status [folate, vitamins A, D, retinol-binding protein (RBP), insulin-growth factor-1 (IGF-1)] and inflammation [C-reactive protein (CRP), cytokines, platelet indices] indicators. Anemia was prevalent (53.5%) but underestimated due to widespread low plasma volume (<2.2 L, 79.9%) and was associated with indicators of malnutrition [lower IGF-1, body mass index (BMI), vitamin D, and intake of green/leafy vegetables], but not inflammation. Higher CRP was associated with lower serum iron, and higher hepcidin and ferritin, whereas maternal platelets were associated with lower HCAZ (β = −0.22), WAZ (β = −0.17), and LAZ (β = −0.17). Higher LAZ was also associated with maternal serum vitamin D (β = 0.23), whereas maternal iron supplementation lowered LAZ (β = −0.22). Assessment of iron status in this MINDI cohort is complex and supplementation strategies must consider consequences for both the mother and the infant.
Collapse
Affiliation(s)
- Doris González-Fernández
- School of Human Nutrition, McGill University, (Macdonald Campus), Ste-Anne de Bellevue, QC H9X 3V9, Canada
| | - Elizabeta Nemeth
- Center for Iron Disorders, David Geffen School of Medicine, University of California, Los Angeles, CA 90089, USA
| | | | | | - Delfina Rueda
- “Comarca Ngäbe-Buglé” Health Region, Panamanian Ministry of Health, Panama City, Panama
| | - Lisa Starr
- Department of Biochemistry, University of Panama, Panama City, Panama
| | - Veena Sangkhae
- Center for Iron Disorders, David Geffen School of Medicine, University of California, Los Angeles, CA 90089, USA
| | - Enrique Murillo
- Department of Biochemistry, University of Panama, Panama City, Panama
| | - Marilyn E. Scott
- Institute of Parasitology, McGill University, (Macdonald Campus), Ste-Anne de Bellevue, QC H9X 3V9, Canada
| | - Kristine G. Koski
- School of Human Nutrition, McGill University, (Macdonald Campus), Ste-Anne de Bellevue, QC H9X 3V9, Canada
- Correspondence: ; Tel.: +1-514-398-7845
| |
Collapse
|
8
|
Clinical interpretation of serum hepcidin-25 in inflammation and renal dysfunction. J Mass Spectrom Adv Clin Lab 2022; 24:43-49. [PMID: 35403094 PMCID: PMC8983384 DOI: 10.1016/j.jmsacl.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/16/2022] [Accepted: 03/27/2022] [Indexed: 11/22/2022] Open
Abstract
log[hepcidin]:log[ferritin] ratio may serve as a biomarker for iron deficiency in complex cases. Hepcidin testing is not warranted in patients with CRP > 10 mg/l and/or eGFR < 30 ml/min/1.73 m2. Inflammation is not a determinant of serum hepcidin-25 in the setting of renal dysfunction. eGFR is not a major determinant of serum hepcidin-25 concentration in patient with eGFR ≥ 30 ml/min/1.73 m2.
Introduction Hepcidin is a hormone that regulates systemic iron homeostasis. Serum hepcidin levels are under the influence of various stimuli, particularly inflammation and renal dysfunction. The measurement of hepcidin in circulation is a potentially useful clinical tool in the diagnosis, monitoring and treatment of iron metabolism disorder, although clinical interpretation of hepcidin level remains difficult. We evaluated he diagnostic potential and limitations of hepcidin-25 by investigating its relationship with iron and hematological indices, inflammation, and renal dysfunction. Methods This retrospective study included 220 adult patients not requiring dialysis. Variations of biologically active hepcidin-25 were examined using a mass spectrometry-based assay in various inflammatory and renal states. The log[hepcidin]:log[ferritin] ratio was calculated as an hepcidin index. Results In 220 adult patients not requiring dialysis, variation in hepcidin-25 level was significantly larger once CRP exceeded 10 mg/l (p < 0.001). Inflammation was not a determinant of hepcidin-25 in the setting of renal dysfunction. Hepcidin-25 median (7.37 nM) and variance were significantly higher (p < 0.001), once estimated glomerular filtration rate (eGFR) dropped below 30 ml/min/1.73 m2. The log[hepcidin]:log[ferritin] index normalized hepcidin levels. Patients with iron deficiency have a notably lower index when compared to controls (-0.66 vs 0.3). Conclusion Severe renal dysfunction (eGFR < 30) affected hepcidin-25 expression and clearance to variable degree between individuals. Although, hepcidin-25 testing is not warranted in patients with infection, inflammatory autoimmune conditions (CRP > 10 mg/l) and/or severe renal dysfunction (eGFR < 30), the hepcidin index may serve as a potential biomarker for iron deficiency in complex cases.
Collapse
|
9
|
Rana S, Prabhakar N. Iron disorders and hepcidin. Clin Chim Acta 2021; 523:454-468. [PMID: 34755647 DOI: 10.1016/j.cca.2021.10.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022]
Abstract
Iron is an essential element due to its role in a wide variety of physiological processes. Iron homeostasis is crucial to prevent iron overload disorders as well as iron deficiency anemia. The liver synthesized peptide hormone hepcidin is a master regulator of systemic iron metabolism. Given its role in overall health, measurement of hepcidin can be used as a predictive marker in disease states. In addition, hepcidin-targeting drugs appear beneficial as therapeutic agents. This review emphasizes recent development on analytical techniques (immunochemical, mass spectrometry and biosensors) and therapeutic approaches (hepcidin agonists, stimulators and antagonists). These insights highlight hepcidin as a potential biomarker as well as an aid in the development of new drugs for iron disorders.
Collapse
Affiliation(s)
- Shilpa Rana
- Department of Biochemistry, Sector-25, Panjab University, Chandigarh 160014, India
| | - Nirmal Prabhakar
- Department of Biochemistry, Sector-25, Panjab University, Chandigarh 160014, India.
| |
Collapse
|
10
|
Association of vitamin A with anemia and serum hepcidin levels in children aged 6 to 59 mo. Nutrition 2021; 91-92:111463. [PMID: 34628277 DOI: 10.1016/j.nut.2021.111463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/10/2021] [Accepted: 08/19/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVE This study evaluates the association of serum retinol, hepcidin levels, and anemia in children. METHODS This cross-sectional study included 312 children, ages 6 to 59 mo, from Rio de Janeiro, Brazil. The association between hepcidin and retinol levels, hematologic parameters, and body mass index (BMI) was analyzed using a generalized linear model with and without adjustment for C-reactive protein (CRP) level. Logistic regression analysis was used to test anemia as an outcome and serum retinol level as a predictive variable using the odds ratio (OR) function. RESULTS Anemia was present in 14.6% of the children, 5.8% presented iron deficiency anemia, and 9.6% had vitamin A deficiency. The increase in serum retinol levels reduced the chances of anemia (OR = 0.13; confidence interval = 0.29-0.59). When CRP level was not adjusted for in the multiple regression analyses, retinol, ferritin levels, and BMI/age were predictors of serum hepcidin levels (β = -3.36, 0.14, 1.02, respectively; P = 0.032). Accordingly, serum retinol levels were inversely associated with CRP levels (β = -0.025 and P < 0.001). CONCLUSIONS The association between serum retinol and hepcidin levels in children ages 6 to 59 mo seems to be dependent on inflammation. Taken together, the results reinforce the need for the development of further studies to better understand the relationship between vitamin A and anemia of inflammation.
Collapse
|
11
|
Wessells KR, Arnold CD, Stewart CP, Prado EL, Abbeddou S, Adu-Afarwuah S, Arnold BF, Ashorn P, Ashorn U, Becquey E, Brown KH, Byrd KA, Campbell RK, Christian P, Fernald L, Fan YM, Galasso E, Hess SY, Huybregts L, Jorgensen JM, Kiprotich M, Kortekangas E, Lartey A, Le Port A, Leroy JL, Lin A, Maleta K, Matias SL, Mbuya M, Mridha MK, Mutasa K, Naser AM, Paul RR, Okronipa H, Ouédraogo JB, Pickering AJ, Rahman M, Schulze K, Smith LE, Weber AM, Zongrone A, Dewey KG. Characteristics that modify the effect of small-quantity lipid-based nutrient supplementation on child anemia and micronutrient status: an individual participant data meta-analysis of randomized controlled trials. Am J Clin Nutr 2021; 114:68S-94S. [PMID: 34590114 PMCID: PMC8560313 DOI: 10.1093/ajcn/nqab276] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 08/04/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Small-quantity lipid-based nutrient supplements (SQ-LNSs) have been shown to reduce the prevalence of child anemia and iron deficiency, but effects on other micronutrients are less well known. Identifying subgroups who benefit most from SQ-LNSs could support improved program design. OBJECTIVES We aimed to identify study-level and individual-level modifiers of the effect of SQ-LNSs on child hemoglobin (Hb), anemia, and inflammation-adjusted micronutrient status outcomes. METHODS We conducted a 2-stage meta-analysis of individual participant data from 13 randomized controlled trials of SQ-LNSs provided to children 6-24 mo of age (n = 15,946). We generated study-specific and subgroup estimates of SQ-LNSs compared with control, and pooled the estimates using fixed-effects models. We used random-effects meta-regression to examine potential study-level effect modifiers. RESULTS SQ-LNS provision decreased the prevalence of anemia (Hb < 110 g/L) by 16% (relative reduction), iron deficiency (plasma ferritin < 12 µg/L) by 56%, and iron deficiency anemia (IDA; Hb < 110 g/L and plasma ferritin <12 µg/L) by 64%. We observed positive effects of SQ-LNSs on hematological and iron status outcomes within all subgroups of the study- and individual-level effect modifiers, but effects were larger in certain subgroups. For example, effects of SQ-LNSs on anemia and iron status were greater in trials that provided SQ-LNSs for >12 mo and provided 9 (as opposed to <9) mg Fe/d, and among later-born (than among first-born) children. There was no effect of SQ-LNSs on plasma zinc or retinol, but there was a 7% increase in plasma retinol-binding protein (RBP) and a 56% reduction in vitamin A deficiency (RBP < 0.70 µmol/L), with little evidence of effect modification by individual-level characteristics. CONCLUSIONS SQ-LNSs can substantially reduce the prevalence of anemia, iron deficiency, and IDA among children across a range of individual, population, and study design characteristics. Policy-makers and program planners should consider SQ-LNSs within intervention packages to prevent anemia and iron deficiency.This trial was registered at www.crd.york.ac.uk/PROSPERO as CRD42020156663.
Collapse
Affiliation(s)
| | - Charles D Arnold
- Institute for Global Nutrition and Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Christine P Stewart
- Institute for Global Nutrition and Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Elizabeth L Prado
- Institute for Global Nutrition and Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Souheila Abbeddou
- Public Health Nutrition, Department of Public Health and Primary Care, University of Ghent, Ghent, Belgium
| | - Seth Adu-Afarwuah
- Department of Nutrition and Food Science, University of Ghana, Legon, Accra, Ghana
| | - Benjamin F Arnold
- Francis I Proctor Foundation, University of California, San Francisco, San Francisco, CA, USA
| | - Per Ashorn
- Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland,Department of Paediatrics, Tampere University Hospital, Tampere, Finland
| | - Ulla Ashorn
- Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Elodie Becquey
- Poverty, Health, and Nutrition Division, International Food Policy Research Institute, Washington, DC, USA
| | - Kenneth H Brown
- Institute for Global Nutrition and Department of Nutrition, University of California, Davis, Davis, CA, USA,Helen Keller International, New York, NY, USA
| | | | - Rebecca K Campbell
- Division of Epidemiology and Biostatistics, University of Illinois at Chicago School of Public Health, Chicago, IL, USA
| | - Parul Christian
- Program in Human Nutrition, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Lia C H Fernald
- School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Yue-Mei Fan
- Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Sonja Y Hess
- Institute for Global Nutrition and Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Lieven Huybregts
- Poverty, Health, and Nutrition Division, International Food Policy Research Institute, Washington, DC, USA
| | - Josh M Jorgensen
- Nutrition Program, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR, USA
| | | | - Emma Kortekangas
- Center for Child Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Anna Lartey
- Department of Nutrition and Food Science, University of Ghana, Legon, Accra, Ghana
| | | | - Jef L Leroy
- Poverty, Health, and Nutrition Division, International Food Policy Research Institute, Washington, DC, USA
| | - Audrie Lin
- School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Kenneth Maleta
- Department of Public Health, School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Susana L Matias
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Mduduzi N N Mbuya
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe,Global Alliance for Improved Nutrition, Washington, DC, USA
| | - Malay K Mridha
- Center for Non-communicable Diseases and Nutrition, BRAC James P Grant School of Public Health, Dhaka, Bangladesh
| | - Kuda Mutasa
- Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Abu M Naser
- International Center for Diarrheal Diseases Research (icddr,b), Dhaka, Bangladesh,Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Rina R Paul
- Center for Non-communicable Diseases and Nutrition, BRAC James P Grant School of Public Health, Dhaka, Bangladesh
| | - Harriet Okronipa
- Department of Population Medicine and Diagnostic Sciences, Cornell University, Ithaca, NY, USA
| | | | | | - Mahbubur Rahman
- International Center for Diarrheal Diseases Research (icddr,b), Dhaka, Bangladesh
| | - Kerry Schulze
- Program in Human Nutrition, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Laura E Smith
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, The State University of New York at Buffalo, Buffalo, NY, USA
| | - Ann M Weber
- Division of Epidemiology, School of Community Health Sciences, University of Nevada, Reno, Reno, NV, USA
| | | | - Kathryn G Dewey
- Institute for Global Nutrition and Department of Nutrition, University of California, Davis, Davis, CA, USA
| |
Collapse
|
12
|
Svenson N, Bailey J, Durairaj S, Dempsey-Hibbert N. A simplified diagnostic pathway for the differential diagnosis of iron deficiency anaemia and anaemia of chronic disease. Int J Lab Hematol 2021; 43:1644-1652. [PMID: 34288431 DOI: 10.1111/ijlh.13666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/08/2021] [Accepted: 07/06/2021] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Iron deficiency anaemia (IDA) and anaemia of chronic disease (ACD) are common causes of anaemia with similar clinical and laboratory features. IDA is caused by low iron stores while ACD is due to iron-restricted erythropoiesis occurring in inflammatory states. Differential diagnosis requires analysis of multiple biochemical and haematological parameters. IDA can occur simultaneously to ACD (mixed aetiology). It is essential that true iron deficiency is identified, as these patients will require iron therapy. This preliminary study investigated whether hepcidin, the master regulator of iron homeostasis, in conjunction with reticulocyte haemoglobin equivalent (RetHe) has the potential to differentiate IDA from ACD, and to exclude IDA in patients with mixed aetiology. METHODS Hepcidin concentration (measured using a commercially available ELISA method), RetHe, and iron parameters along with C-reactive protein (CRP) were analysed in 77 Gastroenterology patients with anaemia in a secondary care setting. RESULTS Receiver operator characteristic (ROC) analysis showed that hepcidin at an optimal cut-off concentration of <6ng/ml could identify IDA with a sensitivity and specificity of 88.9% and 90.6% respectively and could distinguish ACD from IDA with both a sensitivity and specificity of 100% at a cut-off of >46ng/ml. Identifying true IDA in mixed aetiology patients could be achieved by RetHe analysis and applying an optimal cut-off of <30pg. CONCLUSION Hepcidin, in conjunction with RetHe, offers a new simplified diagnostic pathway for differential diagnosis of IDA and ACD, thereby reducing the diagnostic turnaround time and allowing appropriate treatment of patients with a true iron deficiency.
Collapse
Affiliation(s)
- Nicola Svenson
- Department of Haematology, Hull and East Yorkshire Hospitals NHS Trust, Hull, UK
| | - James Bailey
- Queen's Centre for Oncology and Haematology, Castle Hill Hospital, Cottingham, UK
| | | | | |
Collapse
|
13
|
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: 17] [Impact Index Per Article: 5.7] [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
|
14
|
Benson CS, Shah A, Frise MC, Frise CJ. Iron deficiency anaemia in pregnancy: A contemporary review. Obstet Med 2021; 14:67-76. [PMID: 34394714 PMCID: PMC8358243 DOI: 10.1177/1753495x20932426] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/13/2020] [Indexed: 01/01/2023] Open
Abstract
Iron deficiency anaemia is a global health problem, which particularly affects pregnant women. Iron deficiency anaemia during pregnancy is associated with increased maternal and perinatal morbidity and mortality. Maternal iron deficiency may also be associated with neurocognitive deficits in infants. Iron requirements increase during pregnancy and are influenced by hepcidin, the master regulator of iron homeostasis. The enduring global burden of maternal anaemia suggests that currently employed iron supplementation strategies are suboptimal. Recent developments in our understanding of systemic and placental iron homeostasis may improve therapeutic effectiveness by altering the dose and frequency of oral iron. Intravenous iron appears to be a safe treatment to correct maternal anaemia rapidly but research on patient-centred outcomes and cost-effectiveness is needed. Future trials should be adequately powered to assess outcomes relevant to pregnant women.
Collapse
Affiliation(s)
- Charlotte S Benson
- Department of Obstetrics, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Akshay Shah
- Department of Medicine, University of Oxford, Oxford, UK
- Nuffield Department of Anaesthesia, John Radcliffe Hospital, Oxford, UK
| | - Matthew C Frise
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Intensive Care Unit, Royal Berkshire Hospital, NHS Foundation Trust, Reading, UK
| | - Charlotte J Frise
- Fetal Maternal Medicine Unit, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Queen Charlotte’s and Chelsea Hospital, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
15
|
Xu Y, Alfaro-Magallanes VM, Babitt JL. Physiological and pathophysiological mechanisms of hepcidin regulation: clinical implications for iron disorders. Br J Haematol 2021; 193:882-893. [PMID: 33316086 PMCID: PMC8164969 DOI: 10.1111/bjh.17252] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023]
Abstract
The discovery of hepcidin has provided a solid foundation for understanding the mechanisms of systemic iron homeostasis and the aetiologies of iron disorders. Hepcidin assures the balance of circulating and stored iron levels for multiple physiological processes including oxygen transport and erythropoiesis, while limiting the toxicity of excess iron. The liver is the major site where regulatory signals from iron, erythropoietic drive and inflammation are integrated to control hepcidin production. Pathologically, hepcidin dysregulation by genetic inactivation, ineffective erythropoiesis, or inflammation leads to diseases of iron deficiency or overload such as iron-refractory iron-deficiency anaemia, anaemia of inflammation, iron-loading anaemias and hereditary haemochromatosis. In the present review, we discuss recent insights into the molecular mechanisms governing hepcidin regulation, how these pathways are disrupted in iron disorders, and how this knowledge is being used to develop novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Yang Xu
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Víctor M. Alfaro-Magallanes
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- LFE Research Group, Department of Health and Human Performance, Faculty of Physical Activity and Sport Sciences, Universidad Politécnica de Madrid (UPM), Madrid, Spain
| | - Jodie L. Babitt
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Muriuki JM, Mentzer AJ, Mitchell R, Webb EL, Etyang AO, Kyobutungi C, Morovat A, Kimita W, Ndungu FM, Macharia AW, Ngetsa CJ, Makale J, Lule SA, Musani SK, Raffield LM, Cutland CL, Sirima SB, Diarra A, Tiono AB, Fried M, Gwamaka M, Adu-Afarwuah S, Wirth JP, Wegmüller R, Madhi SA, Snow RW, Hill AVS, Rockett KA, Sandhu MS, Kwiatkowski DP, Prentice AM, Byrd KA, Ndjebayi A, Stewart CP, Engle-Stone R, Green TJ, Karakochuk CD, Suchdev PS, Bejon P, Duffy PE, Davey Smith G, Elliott AM, Williams TN, Atkinson SH. Malaria is a cause of iron deficiency in African children. Nat Med 2021; 27:653-658. [PMID: 33619371 PMCID: PMC7610676 DOI: 10.1038/s41591-021-01238-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022]
Abstract
Malaria and iron deficiency (ID) are common and interrelated public health problems in African children. Observational data suggest that interrupting malaria transmission reduces the prevalence of ID1. To test the hypothesis that malaria might cause ID, we used sickle cell trait (HbAS, rs334 ), a genetic variant that confers specific protection against malaria2, as an instrumental variable in Mendelian randomization analyses. HbAS was associated with a 30% reduction in ID among children living in malaria-endemic countries in Africa (n = 7,453), but not among individuals living in malaria-free areas (n = 3,818). Genetically predicted malaria risk was associated with an odds ratio of 2.65 for ID per unit increase in the log incidence rate of malaria. This suggests that an intervention that halves the risk of malaria episodes would reduce the prevalence of ID in African children by 49%.
Collapse
Affiliation(s)
- John Muthii Muriuki
- Kenya Medical Research Institute (KEMRI), Centre for Geographic Medicine Research, Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya.
- Open University, KEMRI-Wellcome Trust Research Programme, Accredited Research Centre, Kilifi, Kenya.
| | - Alexander J Mentzer
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Ruth Mitchell
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Emily L Webb
- MRC Tropical Epidemiology Group, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Anthony O Etyang
- Kenya Medical Research Institute (KEMRI), Centre for Geographic Medicine Research, Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Alireza Morovat
- Department of Clinical Biochemistry, Oxford University Hospitals, Oxford, UK
| | - Wandia Kimita
- Kenya Medical Research Institute (KEMRI), Centre for Geographic Medicine Research, Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Francis M Ndungu
- Kenya Medical Research Institute (KEMRI), Centre for Geographic Medicine Research, Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Alex W Macharia
- Kenya Medical Research Institute (KEMRI), Centre for Geographic Medicine Research, Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Caroline J Ngetsa
- Kenya Medical Research Institute (KEMRI), Centre for Geographic Medicine Research, Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Johnstone Makale
- Kenya Medical Research Institute (KEMRI), Centre for Geographic Medicine Research, Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Swaib A Lule
- MRC/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Solomon K Musani
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Clare L Cutland
- South African Medical Research Council: Vaccines and Infectious Diseases Analytical Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sodiomon B Sirima
- Groupe de Recherche Action en Sante (GRAS), 06 BP 10248, Ouagadougou, Burkina Faso
| | - Amidou Diarra
- Groupe de Recherche Action en Sante (GRAS), 06 BP 10248, Ouagadougou, Burkina Faso
| | - Alfred B Tiono
- Groupe de Recherche Action en Sante (GRAS), 06 BP 10248, Ouagadougou, Burkina Faso
| | - Michal Fried
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Moses Gwamaka
- Mother Offspring Malaria Studies (MOMS) Project, Seattle Biomedical Research Institute, Seattle, WA, USA
- Muheza Designated District Hospital, Muheza, Tanzania
- University of Dar es Salaam, Mbeya College of Health and Allied Sciences, Mbeya, Tanzania
| | - Seth Adu-Afarwuah
- Department of Nutrition and Food Science, University of Ghana, Legon, Ghana
| | | | | | - Shabir A Madhi
- South African Medical Research Council: Vaccines and Infectious Diseases Analytical Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Robert W Snow
- Kenya Medical Research Institute (KEMRI), Centre for Geographic Medicine Research, Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Adrian V S Hill
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Centre for Clinical Vaccinology and Tropical Medicine and the Jenner Institute Laboratories, University of Oxford, Oxford, UK
| | - Kirk A Rockett
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Sanger Institute, Hinxton, UK
| | | | - Dominic P Kwiatkowski
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Wellcome Sanger Institute, Hinxton, UK
| | - Andrew M Prentice
- MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | | | | | | | - Reina Engle-Stone
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Tim J Green
- SAHMRi Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Crystal D Karakochuk
- Food, Nutrition, and Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Parminder S Suchdev
- Department of Pediatrics, Emory University and Emory Global Health Institute, Atlanta, GA, USA
| | - Philip Bejon
- Kenya Medical Research Institute (KEMRI), Centre for Geographic Medicine Research, Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - George Davey Smith
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Alison M Elliott
- MRC/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, UK
| | - Thomas N Williams
- Kenya Medical Research Institute (KEMRI), Centre for Geographic Medicine Research, Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of 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), Centre for Geographic Medicine Research, Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Department of Paediatrics, University of Oxford, Oxford, UK.
| |
Collapse
|
17
|
Pasricha SR, Gilbertson M, Indran T, Bennett A, van Dam M, Coughlin E, Dev A, Chunilal S, Opat S. Safety of rapid injection of undiluted ferric carboxymaltose to patients with iron-deficiency anaemia: a Phase II single-arm study. Intern Med J 2021; 51:1304-1311. [PMID: 33462917 DOI: 10.1111/imj.15195] [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: 10/18/2019] [Revised: 07/13/2020] [Accepted: 12/30/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Ferric carboxymaltose is increasingly utilised to treat iron deficiency and is usually diluted in saline and administered as an intravenous infusion over 15 min. Although this is highly convenient compared with older formulations, we hypothesised the drug could be administered, safely given as a rapid bolus injection. AIMS To define the risk of serious adverse events following administration of an undiluted, rapid, high-dose ferric carboxymaltose injection. Secondary aims included all other adverse events, as well as longitudinal effects on haemoglobin, iron stores, phosphate and hepcidin. METHODS In a single-arm, Phase II study in 121 patients with iron-deficiency anaemia, we administered up to 1000 mg of ferric carboxymaltose as a rapid undiluted bolus injection, and recorded adverse events and collected blood samples over the first hour, and again at 2 and 4 weeks post-treatment. RESULTS No patient experienced a serious adverse event. Flushing during the injection was common, as was a transient headache in the subsequent weeks. One patient experienced Grade 3 chest tightness, necessitating emergency department assessment but not admission or treatment. Treatment produced an average 12.3 g/L improvement in haemoglobin within 2 weeks, but commonly caused reductions in serum phosphate (although none of these was clinically symptomatic). Parenteral iron caused elevations in hepcidin sustained to 4 weeks post-injection. Patients stated they would be prepared to receive the treatment again. CONCLUSION Rapid injection of undiluted ferric carboxymaltose is well tolerated and could provide an approach to treat patients in the ambulatory setting.
Collapse
Affiliation(s)
- Sant-Rayn Pasricha
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia.,Diagnostic and Clinical Haematology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Medical Biology, and Medicine, The University of Melbourne, Melbourne, Victoria, Australia
| | - Michael Gilbertson
- Department of Haematology, Monash Health, Melbourne, Victoria, Australia
| | - Tishya Indran
- Department of Haematology, Monash Health, Melbourne, Victoria, Australia
| | - Ashwini Bennett
- Department of Haematology, Monash Health, Melbourne, Victoria, Australia
| | - Matthew van Dam
- Department of Haematology, Monash Health, Melbourne, Victoria, Australia
| | - Elizabeth Coughlin
- Department of Haematology, Monash Health, Melbourne, Victoria, Australia
| | - Anouk Dev
- Department of Gastroenterology, Monash Health, Melbourne, Victoria, Australia.,Department of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Sanjeev Chunilal
- Department of Haematology, Monash Health, Melbourne, Victoria, Australia
| | - Stephen Opat
- Department of Haematology, Monash Health, Melbourne, Victoria, Australia
| |
Collapse
|
18
|
Pasricha SR, Tye-Din J, Muckenthaler MU, Swinkels DW. Iron deficiency. Lancet 2021; 397:233-248. [PMID: 33285139 DOI: 10.1016/s0140-6736(20)32594-0] [Citation(s) in RCA: 400] [Impact Index Per Article: 133.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023]
Abstract
Iron deficiency is one of the leading contributors to the global burden of disease, and particularly affects children, premenopausal women, and people in low-income and middle-income countries. Anaemia is one of many consequences of iron deficiency, and clinical and functional impairments can occur in the absence of anaemia. Iron deprivation from erythroblasts and other tissues occurs when total body stores of iron are low or when inflammation causes withholding of iron from the plasma, particularly through the action of hepcidin, the main regulator of systemic iron homoeostasis. Oral iron therapy is the first line of treatment in most cases. Hepcidin upregulation by oral iron supplementation limits the absorption efficiency of high-dose oral iron supplementation, and of oral iron during inflammation. Modern parenteral iron formulations have substantially altered iron treatment and enable rapid, safe total-dose iron replacement. An underlying cause should be sought in all patients presenting with iron deficiency: screening for coeliac disease should be considered routinely, and endoscopic investigation to exclude bleeding gastrointestinal lesions is warranted in men and postmenopausal women presenting with iron deficiency anaemia. Iron supplementation programmes in low-income countries comprise part of the solution to meeting WHO Global Nutrition Targets.
Collapse
Affiliation(s)
- Sant-Rayn Pasricha
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Diagnostic Haematology, The Royal Melbourne Hospital, Parkville, VIC, Australia; Department of Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| | - Jason Tye-Din
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Gastroenterology, The Royal Melbourne Hospital, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Martina U Muckenthaler
- Department of Pediatric Oncology, Hematology, and Immunology and Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany; Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany; Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany; German Centre for Cardiovascular Research, Partner Site Heidelberg, Mannheim, Germany
| | - Dorine W Swinkels
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
19
|
Inflammatory Markers and Hepcidin are Elevated but Serum Iron is Lower in Obese Women of Reproductive Age. Nutrients 2021; 13:nu13010217. [PMID: 33466578 PMCID: PMC7828682 DOI: 10.3390/nu13010217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 11/17/2022] Open
Abstract
Limited evidence suggests that serum iron and hepcidin concentrations are dysregulated in obesity and inflammation. The objective of the present study was to compare C-reactive protein, interleukin-6, circulating levels of hepcidin, serum lipids, and iron status in obese vs. normal-weight women of childbearing age. Healthy women aged 18–30 years were recruited for the study (n = 47: 25 obese and 22 normal weight). Fasting blood samples were obtained to measure serum lipids (total cholesterol, HDL, LDL cholesterol, triglycerides, non-HDL cholesterol), complete blood count, serum iron, total iron-binding capacity, transferrin saturation, serum ferritin, hepcidin, C-reactive protein, and interleukin-6. Obese women had significantly higher mean serum C-reactive protein (p < 0.001), interleukin-6 (p < 0.001), hepcidin (p = 0.024), triglycerides (p < 0.001) and total cholesterol/HDL ratio (p < 0.001) but lower HDL (p = 0.001) and serum iron/hepcidin ratio (p = 0.011) compared with normal-weight women. BMI correlated positively with inflammatory markers, triglycerides, LDL and total cholesterol/HDL ratio, and negatively with HDL and serum iron/hepcidin ratio. Serum iron correlated negatively with ferritin in the obese group (p = 0.030) but positively in normal weight women (p = 0.002). BMI and ferritin were the only predictors of serum iron/hepcidin ratio accounting for 23% of the variation among subjects. Studies are needed to examine anti-inflammatory dietary approaches that can improve iron biomarkers in obese women.
Collapse
|
20
|
Prevention of iron deficiency anemia in infants and toddlers. Pediatr Res 2021; 89:63-73. [PMID: 32330927 DOI: 10.1038/s41390-020-0907-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 11/08/2022]
Abstract
Anemia, defined as a low blood hemoglobin concentration, is a major global public health problem. Identification of anemia is crucial to public health interventions. It is estimated globally that 273 million children under 5 years of age were anemic in 2011, and about ~50% of those cases were attributable to iron deficiency (Lancet Global Health 1:e16-e25, 2013). Iron-deficiency anemia (IDA) in infants adversely impacts short-term hematological indices and long-term neuro-cognitive functions of learning and memory that result in both fatigue and low economic productivity. IDA contributes to death and disability and is an important risk factor for maternal and perinatal mortality, including the risks for stillbirths, prematurity, and low birth weight (Comparative Quantification of Health Risks: Global and Regional Burden of Disease Attributable to Selected Major Risk Factors. Ch. 3 (World Health Organization, Geneva, 2004)). Reduction in early infantile anemia and newborn mortality rates is possible with easily implemented, low- to no-cost intervention such as delayed cord clamping (DCC). DCC until 1-3 min after birth facilitates placental transfusion and iron-rich blood flow to the newborn. DCC, an effective anemia prevention strategy, requires cooperation among health providers involved in childbirth, and a participatory culture change in public health. Public intervention strategies must consider multiple factors associated with anemia listed in this review before designing intervention studies that aim to reduce anemia prevalence in infants and toddlers. IMPACT: Anemia, defined as a low blood hemoglobin concentration, is a major global public health problem and identification of anemia is crucial to public health interventions. Delayed cord clamping (DCC) until 1-3 min after birth facilitates placental transfusion and iron-rich blood flow to the newborn. Reduction in early infantile anemia and newborn mortality rates is possible with easily implemented, low- to no-cost intervention such as DCC.
Collapse
|
21
|
Gutschow P, Han H, Olbina G, Westerman K, Nemeth E, Ganz T, Copeland K, Westerman M, Ostland V. Clinical Immunoassay for Human Hepcidin Predicts Iron Deficiency in First-Time Blood Donors. J Appl Lab Med 2020; 5:943-953. [PMID: 32674118 PMCID: PMC7497288 DOI: 10.1093/jalm/jfaa038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/03/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Serum markers currently used as indicators of iron status have clinical limitations. Hepcidin, a key regulator of iron homeostasis, is reduced in iron deficiency (ID) and increased in iron overload. We describe the first CLIA-validated immunoassay with excellent accuracy and precision to quantify human serum hepcidin. Its diagnostic utility for detecting ID in first-time blood donors was demonstrated. METHODS A monoclonal competitive ELISA (C-ELISA) was developed for the quantitation of human hepcidin and validated according to CLIA guidelines. Sera from nonanemic first-time blood donors (n = 292) were analyzed for hepcidin, ferritin, transferrin, and serum iron. Logistic regression served to determine the utility of hepcidin as a predictor of ID. RESULTS The C-ELISA was specific for human hepcidin and had a low limit of quantitation (4.0 ng/mL). The hepcidin concentration measured with the monoclonal C-ELISA was strongly correlated with a previously established, extensively tested polyclonal C-ELISA (Blood 2008;112:4292-7) (r = 0.95, P < 0.001). The area under the receiver operating characteristic curve for hepcidin as a predictor of ID, defined by 3 ferritin concentration thresholds, was >0.9. For predicting ID defined by ferritin <15 ng/mL, hepcidin <10 ng/mL yielded sensitivity of 93.1% and specificity of 85.5%, whereas the same hepcidin cutoff for ferritin <30 ng/mL yielded sensitivity of 67.6% and specificity of 91.7%. CONCLUSION The clinical measurement of serum hepcidin concentrations was shown to be a potentially useful tool for diagnosing ID.
Collapse
Affiliation(s)
| | | | | | | | - Elizabeta Nemeth
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | | | | | | |
Collapse
|
22
|
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: 7.5] [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
|
23
|
Donker AE, Galesloot TE, Laarakkers CM, Klaver SM, Bakkeren DL, Swinkels DW. Standardized serum hepcidin values in Dutch children: Set point relative to body iron changes during childhood. Pediatr Blood Cancer 2020; 67:e28038. [PMID: 31724793 DOI: 10.1002/pbc.28038] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/13/2019] [Accepted: 09/23/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Use of serum hepcidin measurements in pediatrics would benefit from standardized age- and sex-specific reference ranges in children, in order to enable the establishment of clinical decision limits that are universally applicable. PROCEDURE We measured serum hepcidin-25 levels in 266 healthy Dutch children aged 0.3-17 years, using an isotope dilution mass spectrometry assay, standardized with our commutable secondary reference material (RM), assigned by a candidate primary RM. RESULTS We constructed age- and sex-specific values for serum hepcidin and its ratio with ferritin and transferrin saturation (TSAT). Serum hepcidin levels and hepcidin/ferritin and TSAT/hepcidin ratios were similar for both sexes. Serum hepcidin and hepcidin/ferritin ratio substantially declined after the age of 12 years and TSAT/hepcidin ratio gradually increased with increasing age. Serum hepcidin values for Dutch children <12 years (n = 170) and >12 years (n = 96) were 1.9 nmol/L (median); 0.1-13.1 nmol/L (p2.5-p97.5) and 0.9 nmol/L; 0.0-9.1 nmol/L, respectively. Serum ferritin was the most significant correlate of serum hepcidin in our study population, explaining 15.1% and 7.9% of variance in males and females, respectively. Multivariable linear regression analysis including age, blood sampling time, iron parameters, ALT, CRP, and body mass index as independent variables showed a statistically significant negative association between age as a dichotomous variable (≤12 vs >12 years) and log-transformed serum hepcidin levels in both sexes. CONCLUSIONS We demonstrate that serum hepcidin relative to indicators of body iron is age dependent in children, suggesting that the set point of serum hepcidin relative to stored and circulating iron changes during childhood.
Collapse
Affiliation(s)
- Albertine E Donker
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Pediatrics, Máxima Medical Center, Veldhoven, The Netherlands
| | - Tessel E Galesloot
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Coby M Laarakkers
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Translational Metabolic Laboratory (TML, 830), Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Siem M Klaver
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Translational Metabolic Laboratory (TML, 830), Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dirk L Bakkeren
- Department of Clinical Chemistry, Máxima Medical Center , Veldhoven, The Netherlands
| | - Dorine W Swinkels
- Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Translational Metabolic Laboratory (TML, 830), Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
24
|
A possible role for hepcidin in the detection of iron deficiency in severely anaemic HIV-infected patients in Malawi. PLoS One 2020; 15:e0218694. [PMID: 32107492 PMCID: PMC7046342 DOI: 10.1371/journal.pone.0218694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 01/22/2020] [Indexed: 11/19/2022] Open
Abstract
Introduction Iron deficiency is a treatable cause of severe anaemia in low-and-middle-income-countries (LMIC). Diagnosing it remains challenging as peripheral blood markers poorly reflect bone-marrow iron deficiency (BM-ID), especially in the context of HIV-infection. Methods Severely anaemic (haemoglobin ≤70g/l) HIV-infected adults were recruited at Queen Elizabeth Central Hospital, Blantyre, Malawi. BM-ID was evaluated. Accuracy of blood markers (including hepcidin, mean corpuscular volume, mean cellular haemoglobin concentration, serum iron, serum ferritin, soluble transferrin receptor (sTfR), sTfR index, sTfR–ratio) to detect BM-ID was evaluated by ROC area under the curve (AUCROC). Results Seventy-three patients were enrolled and 35 (48.0%) had BM-ID. Although hepcidin and MCV performed best (AUCROC of 0.593 and 0.545 respectively) all markers performed poorly in identifying BM-ID (ROC<0.6). The AUCROC of hepcidin in males was 0.767 (sensitivity 80%, specificity 78%) and in women 0.490 (sensitivity 60%, specificity 61%). Conclusion BM-ID deficiency was common in severely anaemic HIV-infected patients. It is an important and potential treatable contributor to severe anaemia but lack of definitive biomarkers makes it difficult to accurately assess iron status in these patients. Further investigation of the potential of hepcidin is needed, including exploration of the differences in hepcidin results between males and females.
Collapse
|
25
|
Abdullah N, Ahmed A. Growth-differentiation factor-15 expression in anemia of chronic disease and iron-deficiency anemia. IRAQI JOURNAL OF HEMATOLOGY 2020. [DOI: 10.4103/ijh.ijh_7_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
26
|
Moxon CA, Gibbins MP, McGuinness D, Milner DA, Marti M. New Insights into Malaria Pathogenesis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2019; 15:315-343. [PMID: 31648610 DOI: 10.1146/annurev-pathmechdis-012419-032640] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Malaria remains a major public health threat in tropical and subtropical regions across the world. Even though less than 1% of malaria infections are fatal, this leads to about 430,000 deaths per year, predominantly in young children in sub-Saharan Africa. Therefore, it is imperative to understand why a subset of infected individuals develop severe syndromes and some of them die and what differentiates these cases from the majority that recovers. Here, we discuss progress made during the past decade in our understanding of malaria pathogenesis, focusing on the major human parasite Plasmodium falciparum.
Collapse
Affiliation(s)
- Christopher A Moxon
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, United Kingdom; ,
| | - Matthew P Gibbins
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, United Kingdom; ,
| | - Dagmara McGuinness
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, United Kingdom; ,
| | - Danny A Milner
- American Society for Clinical Pathology, Chicago, Illinois 60603, USA.,Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, United Kingdom; , .,Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| |
Collapse
|
27
|
Soluble extracts from carioca beans (Phaseolus vulgaris L.) affect the gut microbiota and iron related brush border membrane protein expression in vivo (Gallus gallus). Food Res Int 2019; 123:172-180. [DOI: 10.1016/j.foodres.2019.04.060] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/16/2019] [Accepted: 04/25/2019] [Indexed: 01/10/2023]
|
28
|
Modelling pathogen load dynamics to elucidate mechanistic determinants of host-Plasmodium falciparum interactions. Nat Microbiol 2019; 4:1592-1602. [PMID: 31209307 PMCID: PMC6708439 DOI: 10.1038/s41564-019-0474-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/30/2019] [Indexed: 12/21/2022]
Abstract
During infection, increasing pathogen load stimulates both protective and
harmful aspects of the host response. The dynamics of this interaction are hard
to quantify in humans, but doing so could improve understanding of mechanisms of
disease and protection. We sought to model the contributions of parasite
multiplication rate and host response to observed parasite load in individual
subjects with Plasmodium falciparum malaria, using only data
obtained at the time of clinical presentation, and then to identify their
mechanistic correlates. We predicted higher parasite multiplication rates and
lower host responsiveness in severe malaria cases, with severe anemia being more
insidious than cerebral malaria. We predicted that parasite growth-inhibition
was associated with platelet consumption, lower expression of
CXCL10 and type-1 interferon-associated genes, but
increased cathepsin G and matrix metallopeptidase 9 expression. We found that
cathepsin G and matrix metallopeptidase 9 directly inhibit parasite invasion
into erythrocytes. Parasite multiplication rate was associated with host iron
availability and higher complement factor H levels, lower expression of
gametocyte-associated genes but higher expression of translation-associated
genes in the parasite. Our findings demonstrate the potential of using explicit
modelling of pathogen load dynamics to deepen understanding of host-pathogen
interactions and identify mechanistic correlates of protection.
Collapse
|
29
|
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: 0.4] [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
|
30
|
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: 6.4] [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
|
31
|
Armitage AE, Agbla SC, Betts M, Sise EA, Jallow MW, Sambou E, Darboe B, Worwui A, Weinstock GM, Antonio M, Pasricha SR, Prentice AM, Drakesmith H, Darboe MK, Kwambana-Adams BA. Rapid growth is a dominant predictor of hepcidin suppression and declining ferritin in Gambian infants. Haematologica 2019; 104:1542-1553. [PMID: 30733275 PMCID: PMC6669141 DOI: 10.3324/haematol.2018.210146] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/31/2019] [Indexed: 12/18/2022] Open
Abstract
Iron deficiency and iron deficiency anemia are highly prevalent in low-income countries, especially among young children. Hepcidin is the major regulator of systemic iron homeostasis. It controls dietary iron absorption, dictates whether absorbed iron is made available in circulation for erythropoiesis and other iron-demanding processes, and predicts response to oral iron supplementation. Understanding how hepcidin is itself regulated is therefore important, especially in young children. We investigated how changes in iron-related parameters, inflammation and infection status, seasonality, and growth influenced plasma hepcidin and ferritin concentrations during infancy using longitudinal data from two birth cohorts of infants in rural Gambia (n=114 and n=193). This setting is characterized by extreme seasonality, prevalent childhood anemia, undernutrition, and frequent infection. Plasma was collected from infants at birth and at regular intervals, up to 12 months of age. Hepcidin, ferritin and plasma iron concentrations declined markedly during infancy, with reciprocal increases in soluble transferrin receptor and transferrin concentrations, indicating declining iron stores and increasing tissue iron demand. In cross-sectional analyses at 5 and 12 months of age, we identified expected relationships of hepcidin with iron and inflammatory markers, but also observed significant negative associations between hepcidin and antecedent weight gain. Correspondingly, longitudinal fixed effects modeling demonstrated weight gain to be the most notable dynamic predictor of decreasing hepcidin and ferritin through infancy across both cohorts. Infants who grow rapidly in this setting are at particular risk of depletion of iron stores, but since hepcidin concentrations decrease with weight gain, they may also be the most responsive to oral iron interventions.
Collapse
Affiliation(s)
- Andrew E Armitage
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Schadrac C Agbla
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Modupeh Betts
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Ebrima A Sise
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Momodou W Jallow
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Ellen Sambou
- WHO Collaborating Center for New Vaccines Surveillance, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Bakary Darboe
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Archibald Worwui
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | | | - Martin Antonio
- WHO Collaborating Center for New Vaccines Surveillance, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Sant-Rayn Pasricha
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.,Walter and Eliza Hall Institute for Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, The University of Melbourne, VIC, Melbourne, Australia
| | - Andrew M Prentice
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.,Haematology Theme, Oxford Biomedical Research Centre, Oxford, UK
| | - Momodou K Darboe
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa
| | - Brenda Anna Kwambana-Adams
- WHO Collaborating Center for New Vaccines Surveillance, MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia, Africa .,NIHR Global Health Research Unit on Mucosal Pathogens, Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
32
|
Shah A, Wray K, James T, Shine B, Morovat R, Stanworth S, McKechnie S, Kirkbride R, Griffith DM, Walsh TS, Drakesmith H, Roy N. Serum hepcidin potentially identifies iron deficiency in survivors of critical illness at the time of hospital discharge. Br J Haematol 2019; 184:279-281. [PMID: 29363744 DOI: 10.1111/bjh.15067] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Akshay Shah
- Nuffield Department of Anaesthetics, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Katherine Wray
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Timothy James
- Department of Clinical Biochemistry, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Brian Shine
- Department of Clinical Biochemistry, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Reza Morovat
- Department of Clinical Biochemistry, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Simon Stanworth
- NHS Blood and Transplant, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Stuart McKechnie
- Nuffield Department of Anaesthetics, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Rachael Kirkbride
- Department of Anaesthesia, Critical Care and Pain Medicine, University of Edinburgh, Edinburgh, UK
| | - David M Griffith
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Anaesthesia, Critical Care and Pain Medicine, University of Edinburgh, Edinburgh, UK
| | - Timothy S Walsh
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Anaesthesia, Critical Care and Pain Medicine, University of Edinburgh, Edinburgh, UK
| | - Hal Drakesmith
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Biomedical Research Centre Blood Theme, NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Noémi Roy
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
33
|
Stewart CP, Dewey KG, Lin A, Pickering AJ, Byrd KA, Jannat K, Ali S, Rao G, Dentz HN, Kiprotich M, Arnold CD, Arnold BF, Allen LH, Shahab-Ferdows S, Ercumen A, Grembi JA, Naser AM, Rahman M, Unicomb L, Colford JM, Luby SP, Null C. Effects of lipid-based nutrient supplements and infant and young child feeding counseling with or without improved water, sanitation, and hygiene (WASH) on anemia and micronutrient status: results from 2 cluster-randomized trials in Kenya and Bangladesh. Am J Clin Nutr 2019; 109:148-164. [PMID: 30624600 PMCID: PMC6358037 DOI: 10.1093/ajcn/nqy239] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 08/14/2018] [Indexed: 12/24/2022] Open
Abstract
Background Anemia in young children is a global health problem. Risk factors include poor nutrient intake and poor water quality, sanitation, or hygiene. Objective We evaluated the effects of water quality, sanitation, handwashing, and nutrition interventions on micronutrient status and anemia among children in rural Kenya and Bangladesh. Design We nested substudies within 2 cluster-randomized controlled trials enrolling pregnant women and following their children for 2 y. These substudies included 4 groups: water, sanitation, and handwashing (WSH); nutrition (N), including lipid-based nutrient supplements (LNSs; ages 6-24 mo) and infant and young child feeding (IYCF) counseling; WSH+N; and control. Hemoglobin and micronutrient biomarkers were measured after 2 y of intervention and compared between groups using generalized linear models with robust SEs. Results In Kenya, 699 children were assessed at a mean ± SD age of 22.1 ± 1.8 mo, and in Bangladesh 1470 participants were measured at a mean ± SD age of 28.0 ± 1.9 mo. The control group anemia prevalences were 48.8% in Kenya and 17.4% in Bangladesh. There was a lower prevalence of anemia in the 2 N intervention groups in both Kenya [N: 36.2%; prevalence ratio (PR): 0.74; 95% CI: 0.58, 0.94; WSH+N: 27.3%; PR: 0.56; 95% CI: 0.42, 0.75] and Bangladesh (N: 8.7%; PR: 0.50; 95% CI: 0.32, 0.78; WSH+N: 7.9%, PR: 0.46; 95% CI: 0.29, 0.73). In both trials, the 2 N groups also had significantly lower prevalences of iron deficiency, iron deficiency anemia, and low vitamin B-12 and, in Kenya, a lower prevalence of folate and vitamin A deficiencies. In Bangladesh, the WSH group had a lower prevalence of anemia (12.8%; PR: 0.74; 95% CI: 0.54, 1.00) than the control group, whereas in Kenya, the WSH+N group had a lower prevalence of anemia than did the N group (PR: 0.75; 95% CI: 0.53, 1.07), but this was not significant (P = 0.102). Conclusions IYCF counseling with LNSs reduced the risks of anemia, iron deficiency, and low vitamin B-12. Effects on folate and vitamin A varied between studies. Improvements in WSH also reduced the risk of anemia in Bangladesh but did not provide added benefit over the nutrition-specific intervention. These trials were registered at clinicaltrials.gov as NCT01590095 (Bangladesh) and NCT01704105 (Kenya).
Collapse
Affiliation(s)
- Christine P Stewart
- Department of Nutrition, University of California, Davis, Davis, CA,Address correspondence to CPS (e-mail: )
| | - Kathryn G Dewey
- Department of Nutrition, University of California, Davis, Davis, CA
| | - Audrie Lin
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA
| | - Amy J Pickering
- Department of Civil and Environmental Engineering,Present address for AJP: Department of Civil and Environmental Engineering, Tufts University, 200 College Avenue, Medford, MA 02155
| | - Kendra A Byrd
- Department of Nutrition, University of California, Davis, Davis, CA
| | - Kaniz Jannat
- International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Shahjahan Ali
- International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Gouthami Rao
- Innovations for Poverty Action, Nairobi, Kenya,Present address for GR: Waterborne Disease Prevention Branch, Division of Foodborne, Waterborne, and Environmental Diseases, CDC, 1600 Clifton Road, Atlanta, GA 30329
| | - Holly N Dentz
- Department of Nutrition, University of California, Davis, Davis, CA,Innovations for Poverty Action, Nairobi, Kenya
| | - Marion Kiprotich
- Innovations for Poverty Action, Nairobi, Kenya,Present address for MK: One Acre Fund, Nairobi, Kenya
| | - Charles D Arnold
- Department of Nutrition, University of California, Davis, Davis, CA
| | - Benjamin F Arnold
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA
| | - Lindsay H Allen
- USDA, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA
| | | | - Ayse Ercumen
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA
| | | | - Abu Mohd Naser
- International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Mahbubur Rahman
- International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Leanne Unicomb
- International Center for Diarrheal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - John M Colford
- Division of Epidemiology and Biostatistics, University of California, Berkeley, Berkeley, CA
| | - Stephen P Luby
- Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, CA
| | - Clair Null
- Innovations for Poverty Action, Nairobi, Kenya,Present address for CN: Mathmatica Policy Research, 1100 First Street, NE, 12th Floor, Washington, DC 20002
| |
Collapse
|
34
|
Vitamin D Supplementation Modestly Reduces Serum Iron Indices of Healthy Arab Adolescents. Nutrients 2018; 10:nu10121870. [PMID: 30513812 PMCID: PMC6315440 DOI: 10.3390/nu10121870] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/21/2018] [Accepted: 11/27/2018] [Indexed: 01/17/2023] Open
Abstract
Vitamin D deficiency has been shown to affect iron status via decreased calcitriol production, translating to decreased erythropoiesis. The present study aimed to determine for the first time whether vitamin D supplementation can affect iron levels among Arab adolescents. A total of 125 out of the initial 200 Saudi adolescents with vitamin D deficiency (serum 25(OH)D < 50 nmol/L) were selected from the Vitamin D-School Project of King Saud University in Riyadh, Saudi Arabia. Cluster randomization was done in schools, and students received either vitamin D tablets (1000 IU/day) (N = 53, mean age 14.1 ± 1.0 years) or vitamin D-fortified milk (40IU/200mL) (N = 72, mean age 14.8 ± 1.4 years). Both groups received nutritional counseling. Anthropometrics, glucose, lipids, iron indices, and 25(OH)D were measured at baseline and after six months. Within group analysis showed that post-intervention, serum 25(OH)D significantly increased by as much as 50%, and a parallel decrease of −42% (p-values <0.001 and 0.002, respectively) was observed in serum iron in the tablet group. These changes were not observed in the control group. Between-group analysis showed a clinically significant increase in serum 25(OH)D (p = 0.001) and decrease in iron (p < 0.001) in the tablet group. The present findings suggest a possible inhibitory role of vitamin D supplementation in the iron indices of healthy adolescents whose 25(OH)D levels are sub-optimal but not severely deficient, implying that the causal relationship between both micronutrients may be dependent on the severity of deficiency, type of iron disorder, and other vascular conditions that are known to affect hematologic indices. Well-designed, randomized trials are needed to confirm the present findings.
Collapse
|
35
|
Affiliation(s)
- Chaim Hershko
- Professor Emeritus, Department of Hematology, Shaare Zedek Medical Center, Jerusalem, Israel
| |
Collapse
|
36
|
Pereira DI, Mohammed NI, Ofordile O, Camara F, Baldeh B, Mendy T, Sanyang C, Jallow AT, Hossain I, Wason J, Prentice AM. A novel nano-iron supplement to safely combat iron deficiency and anaemia in young children: The IHAT-GUT double-blind, randomised, placebo-controlled trial protocol. Gates Open Res 2018; 2:48. [PMID: 30569038 PMCID: PMC6266659 DOI: 10.12688/gatesopenres.12866.2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2018] [Indexed: 12/11/2022] Open
Abstract
Background: Iron deficiency and its associated anaemia (IDA) are the leading forms of micronutrient malnutrition worldwide. Here we describe the rationale and design of the first clinical trial evaluating the efficacy and safety of an innovative nano iron supplement, iron hydroxide adipate tartrate (IHAT), for the treatment of IDA in young children (IHAT-GUT trial). Oral iron is often ineffective due to poor absorption and/or gastrointestinal adverse effects. IHAT is novel since it is effectively absorbed whilst remaining nanoparticulate in the gut, therefore should enable supplementation with fewer symptoms. Methods: IHAT-GUT is a three-arm, double-blind, randomised, placebo-controlled phase II trial conducted in Gambian children 6-35 months of age. The intervention consists of a 12-week supplementation with either IHAT, ferrous sulphate (both at doses bioequivalent to 12.5 mg Fe/day) or placebo. The trial aims to include 705 children with IDA who will be randomly assigned (1:1:1) to each arm. The primary objectives are to test non-inferiority of IHAT in relation to ferrous sulphate at treating IDA, and to test superiority of IHAT in relation to ferrous sulphate and non-inferiority in relation to placebo in terms of diarrhoea incidence and prevalence. Secondary objectives are mechanistic assessments, to test whether IHAT reduces the burden of enteric pathogens, morbidity, and intestinal inflammation, and that it does not cause detrimental changes to the gut microbiome, particularly in relation to Lactobacillaceae, Bifidobacteriaceae and Enterobacteriaceae. Discussion: This trial will test the hypothesis that supplementation with IHAT eliminates iron deficiency and improves haemoglobin levels without inducing gastrointestinal adverse effects. If shown to be the case, this would open the possibility for further testing and use of IHAT as a novel iron source for micronutrient intervention strategies in resource-poor countries, with the ultimate aim to help reduce the IDA global burden. Registration: This trial is registered at clinicaltrials.gov ( NCT02941081).
Collapse
Affiliation(s)
- Dora I.A. Pereira
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Nuredin I. Mohammed
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Ogochukwu Ofordile
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Famalang Camara
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Bakary Baldeh
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Thomas Mendy
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Chilel Sanyang
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Amadou T. Jallow
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Ilias Hossain
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - James Wason
- MRC Biostatistics Unit, Institute of Public Health, University of Cambridge, Cambridge, CB2 0SR, UK
- Institute of Health and Society, Newcastle University, Newcastle, NE2 4BN, UK
| | - Andrew M. Prentice
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| |
Collapse
|
37
|
Pereira DIA, Mohammed NI, Ofordile O, Camara F, Baldeh B, Mendy T, Sanyang C, Jallow AT, Hossain I, Wason J, Prentice AM. A novel nano-iron supplement to safely combat iron deficiency and anaemia in young children: The IHAT-GUT double-blind, randomised, placebo-controlled trial protocol. Gates Open Res 2018. [PMID: 30569038 DOI: 10.12688/gatesopenres.12866.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Iron deficiency and its associated anaemia (IDA) are the leading forms of micronutrient malnutrition worldwide. Here we describe the rationale and design of the first clinical trial evaluating the efficacy and safety of an innovative nano iron supplement, iron hydroxide adipate tartrate (IHAT), for the treatment of IDA in young children (IHAT-GUT trial). Oral iron is often ineffective due to poor absorption and/or gastrointestinal adverse effects. IHAT is novel since it is effectively absorbed whilst remaining nanoparticulate in the gut, therefore should enable supplementation with fewer symptoms. Methods: IHAT-GUT is a three-arm, double-blind, randomised, placebo-controlled phase II trial conducted in Gambian children 6-35 months of age. The intervention consists of a 12-week supplementation with either IHAT, ferrous sulphate (both at doses bioequivalent to 12.5 mg Fe/day) or placebo. The trial aims to include 705 children with IDA who will be randomly assigned (1:1:1) to each arm. The primary objectives are to test non-inferiority of IHAT in relation to ferrous sulphate at treating IDA, and to test superiority of IHAT in relation to ferrous sulphate and non-inferiority in relation to placebo in terms of diarrhoea incidence and prevalence. Secondary objectives are mechanistic assessments, to test whether IHAT reduces the burden of enteric pathogens, morbidity, and intestinal inflammation, and that it does not cause detrimental changes to the gut microbiome, particularly in relation to Lactobacillaceae, Bifidobacteriaceae and Enterobacteriaceae. Discussion: This trial will test the hypothesis that supplementation with IHAT eliminates iron deficiency and improves haemoglobin levels without inducing gastrointestinal adverse effects. If shown to be the case, this would open the possibility for further testing and use of IHAT as a novel iron source for micronutrient intervention strategies in resource-poor countries, with the ultimate aim to help reduce the IDA global burden. Registration: This trial is registered at clinicaltrials.gov ( NCT02941081).
Collapse
Affiliation(s)
- Dora I A Pereira
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK.,Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Nuredin I Mohammed
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Ogochukwu Ofordile
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Famalang Camara
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Bakary Baldeh
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Thomas Mendy
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Chilel Sanyang
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Amadou T Jallow
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Ilias Hossain
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - James Wason
- MRC Biostatistics Unit, Institute of Public Health, University of Cambridge, Cambridge, CB2 0SR, UK.,Institute of Health and Society, Newcastle University, Newcastle, NE2 4BN, UK
| | - Andrew M Prentice
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| |
Collapse
|
38
|
Muriuki JM, Atkinson SH. How Eliminating Malaria May Also Prevent Iron Deficiency in African Children. Pharmaceuticals (Basel) 2018; 11:ph11040096. [PMID: 30275421 PMCID: PMC6315967 DOI: 10.3390/ph11040096] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/21/2018] [Accepted: 09/21/2018] [Indexed: 02/07/2023] Open
Abstract
Malaria and iron deficiency are common among children living in sub-Saharan Africa. Several studies have linked a child’s iron status to their future risk of malaria infection; however, few have examined whether malaria might be a cause of iron deficiency. Approximately a quarter of African children at any one time are infected by malaria and malaria increases hepcidin and tumor necrosis factor-α concentrations leading to poor iron absorption and recycling. In support of a hypothetical link between malaria and iron deficiency, studies indicate that the prevalence of iron deficiency in children increases over a malaria season and decreases when malaria transmission is interrupted. The link between malaria and iron deficiency can be tested through the use of observational studies, randomized controlled trials and genetic epidemiology studies, each of which has its own strengths and limitations. Confirming the existence of a causal link between malaria infection and iron deficiency would readjust priorities for programs to prevent and treat iron deficiency and would demonstrate a further benefit of malaria control.
Collapse
Affiliation(s)
| | - Sarah H Atkinson
- KEMRI-Wellcome Trust Research Programme, 80108 Kilifi, Kenya.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7FZ, UK.
- Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK.
| |
Collapse
|
39
|
Ghatpande NS, Apte PP, Naik SS, Kulkarni PP. Fruit and Vegetable Consumption and Their Association With the Indicators of Iron and Inflammation Status Among Adolescent Girls. J Am Coll Nutr 2018; 38:218-226. [PMID: 30130470 DOI: 10.1080/07315724.2018.1492470] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND The objective of this study was to identify an association among dietary components, iron, and inflammatory status among adolescent girls. METHOD Dietary information for 85 adolescent girls was collected through food frequency questionnaires. Biomarkers of iron and inflammatory status were analyzed. RESULTS We found that 28.2% of adolescent girls had anemia and 65.9% girls were iron-deficient. Girls who did not consume guava had 3.8-fold (95% confidence interval =1.1-9.4; p = 0.020) increased the risk of having low serum iron levels. Girls who consumed amaranth had significantly (p = 0.024) higher serum hepcidin levels (n = 44; 129.7 ± 81.40 pg/mL vs n = 41; 94.6 ± 55.8 pg/mL) as well as ferritin levels (n = 44; 19.7 ± 16.4 µg/L vs n = 41; 14.0 ± 10.2 µg/L). Overall consumption of fruits and green leafy vegetables among girls significantly affects their iron status. CONCLUSIONS Regular consumption of vitamin C-rich fruits and green leafy vegetable intake are imperative for improvement of iron status among adolescent girls.
Collapse
Affiliation(s)
- Niraj S Ghatpande
- a Bioprospecting Group, Agharkar Research Institute , Pune , India.,b Savitribai Phule Pune University, Ganeshkhind , Pune , India
| | - Priti P Apte
- b Savitribai Phule Pune University, Ganeshkhind , Pune , India
| | | | | |
Collapse
|
40
|
Kanuri G, Chichula D, Sawhney R, Kuriakose K, De'Souza S, Pais F, Arumugam K, Shet AS. Optimizing diagnostic biomarkers of iron deficiency anemia in community-dwelling Indian women and preschool children. Haematologica 2018; 103:1991-1996. [PMID: 30093400 PMCID: PMC6269316 DOI: 10.3324/haematol.2018.193243] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 08/06/2018] [Indexed: 12/31/2022] Open
Abstract
The detection of iron deficiency anemia is challenged by the paucity of diagnostic tests demonstrating high sensitivity and specificity. Using two biomarkers, zinc-protoporphyrin/heme and hepcidin, we established the diagnostic cut-off values for iron deficiency anemia in preschool children and women. We randomly selected non-anemic individuals (n=190; women=90, children=100) and individuals with iron deficiency anemia (n=200; women=100, children=100) from a preexisting cohort of healthy preschool children and their mothers. The diagnostic performance of these biomarkers was estimated by analyzing receiver operating characteristic curves. Diagnostic cut-offs with a high predictive value for iron deficiency anemia were selected. Median zinc-protoporphyrin/heme and hepcidin values in non-anemic children were 49 μmol/mol heme and 42 ng/mL, respectively, and in non-anemic women these values were 66 μmol/mol heme and 17.7ng/mL, respectively. Children and women with iron deficiency anemia had higher zinc-protoporphyrin/heme ratios (children=151 μmol/mol heme and women=155 μmol/mol heme) and lower hepcidin levels (children=1.2ng/mL and women=0.6ng/mL). A zinc-protoporphyrin/heme ratio cut-off >90 μmole/mole heme in children and >107 μmole/mole heme in women was associated with a high diagnostic likelihood for iron deficiency anemia (children, likelihood ratio=20.2: women, likelihood ratio=10.8). Hepcidin cut-off values of ≤6.8ng/mL in children and ≤4.5ng/mL in women were associated with a high diagnostic likelihood for iron deficiency anemia (children, likelihood ratio=14.3: women, likelihood ratio=16.2). The reference ranges and cut-off values identified in this study provide clinicians with guidance for applying these tests to detect iron deficiency anemia. Erythrocyte zinc-protoporphyrin/heme ratio is a valid point-of-care biomarker to diagnose iron deficiency anemia.
Collapse
Affiliation(s)
- Giridhar Kanuri
- Wellcome Trust-DBT Hematology Research Unit, St. Johns Research Institute, Bangalore, Karnataka, India.,Department of Biotechnology, KLEF, Greenfields, Vaddeswaram, Andhra Pradesh, India
| | - Deepti Chichula
- Wellcome Trust-DBT Hematology Research Unit, St. Johns Research Institute, Bangalore, Karnataka, India
| | - Ritica Sawhney
- Wellcome Trust-DBT Hematology Research Unit, St. Johns Research Institute, Bangalore, Karnataka, India
| | - Kevin Kuriakose
- Wellcome Trust-DBT Hematology Research Unit, St. Johns Research Institute, Bangalore, Karnataka, India
| | - Sherwin De'Souza
- Wellcome Trust-DBT Hematology Research Unit, St. Johns Research Institute, Bangalore, Karnataka, India
| | - Faye Pais
- Wellcome Trust-DBT Hematology Research Unit, St. Johns Research Institute, Bangalore, Karnataka, India
| | - Karthika Arumugam
- Wellcome Trust-DBT Hematology Research Unit, St. Johns Research Institute, Bangalore, Karnataka, India
| | - Arun S Shet
- Wellcome Trust-DBT Hematology Research Unit, St. Johns Research Institute, Bangalore, Karnataka, India .,National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
41
|
Lee N, Makani J, Tluway F, Makubi A, Armitage AE, Pasricha SR, Drakesmith H, Prentice AM, Cox SE. Decreased Hepcidin Levels Are Associated with Low Steady-state Hemoglobin in Children With Sickle Cell Disease in Tanzania. EBioMedicine 2018; 34:158-164. [PMID: 30056060 PMCID: PMC6116423 DOI: 10.1016/j.ebiom.2018.07.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 07/17/2018] [Accepted: 07/17/2018] [Indexed: 01/17/2023] Open
Abstract
Background The contribution of hepcidin as a regulator of iron metabolism & erythropoiesis on the severity of anemia in sickle cell disease (SCD) remains poorly characterized, especially in Sub-Saharan African populations. The aims of the study were to determine if hepcidin is associated with severity of steady-state anemia in SCD and to investigate factors associated with hepcidin and anemia in SCD. Methods Archived samples from 199 Tanzanian children, 56% boys aged 3–18 with laboratory-confirmed SCD were analysed based on recorded averaged steady-state hemoglobin (ASSH) quartiles (lowest vs. highest). Univariable and multivariable logistic regression was used to assess associations with ASSH quartiles. Findings In univariable analysis, hepcidin <5·5 ng/mL was associated with increased odds of being in the lowest ASSH quartile (OR 2·20; 95%CI 1·2–3·93) but which was limited to girls (OR 4·85, 95%CI 1·79–13·09, p = .046 for interaction). In multivariable analyses including either reticulocyte percentage or erythropoietin, lower hepcidin remained significantly associated with lowest ASSH quartile, although the hepcidin-sex interaction no longer reached statistical significance. No associations with ASSH quartile were observed for markers of inflammation, hemolysis or potential iron markers except for microcytosis, associated with higher ASSH, but which was confounded by reticulocyte percentage and alpha-thalassaemia status. Interpretation Hepcidin is lower in more severely anaemic children with SCD independent of inflammation or markers of erythropoiesis. Funding Funding sources include The Wellcome Trust (080025, 095009, 094780 & 070114), MRC-UK (MC-A760-5QX00), NIHR Oxford Biomedical Research Centre, and the Bill and Melinda Gates Foundation (“Hepcidin and Iron in Global Health”, OPP1055865). Low hepcidin levels are associated with severe anemia independent of markers of erythropoiesis and inflammation. Low serum ferritin is present in some children but is not associated with severe anemia. Although iron markers remain difficult to interpret in sickle cell disease, hepcidin levels were mostly below levels that would be expected to inhibit iron absorption and recycling.
Collapse
Affiliation(s)
- Nathaniel Lee
- School of Tropical Medicine & Global Health, Nagasaki University, Nagasaki, Japan
| | - Julie Makani
- Sickle Cell Programme, Muhimbili University of Health & Allied Sciences, Dar-es-Salaam, Tanzania; Department of Haematology & Blood Transfusion, Muhimbili University of Health and Allied Sciences, Dar-es-Salaam, Tanzania
| | - Furahini Tluway
- Sickle Cell Programme, Muhimbili University of Health & Allied Sciences, Dar-es-Salaam, Tanzania
| | - Abel Makubi
- Department of Haematology & Blood Transfusion, Muhimbili University of Health and Allied Sciences, Dar-es-Salaam, Tanzania
| | - Andrew E Armitage
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, UK
| | - Sant-Rayn Pasricha
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, UK
| | - Hal Drakesmith
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, UK
| | | | - Sharon E Cox
- School of Tropical Medicine & Global Health, Nagasaki University, Nagasaki, Japan; London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
42
|
Abstract
Iron deficiency remains the largest nutritional deficiency worldwide and the main cause of anaemia. Severe iron deficiency leads to anaemia known as iron deficiency anaemia (IDA), which affects a total of 1·24 billion people, the majority of whom are children and women from resource-poor countries. In sub-Saharan Africa, iron deficiency is frequently exacerbated by concomitant parasitic and bacterial infections and contributes to over 120 000 maternal deaths a year, while it irreparably limits the cognitive development of children and leads to poor outcomes in pregnancy.Currently available iron compounds are cheap and readily available, but constitute a non-physiological approach to providing iron that leads to significant side effects. Consequently, iron deficiency and IDA remain without an effective treatment, particularly in populations with high burden of infectious diseases. So far, despite considerable investment in the past 25 years in nutrition interventions with iron supplementation and fortification, we have been unable to significantly decrease the burden of this disease in resource-poor countries.If we are to eliminate this condition in the future, it is imperative to look beyond the strategies used until now and we should make an effort to combine community engagement and social science approaches to optimise supplementation and fortification programmes.
Collapse
|
43
|
Camaschella C, Pagani A. Advances in understanding iron metabolism and its crosstalk with erythropoiesis. Br J Haematol 2018; 182:481-494. [PMID: 29938779 DOI: 10.1111/bjh.15403] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent years have witnessed impressive advances in our understanding of iron metabolism. A number of studies of iron disorders and of their animal models have provided landmark insights into the mechanisms of iron trafficking, distribution and homeostatic regulation, the latter essential to prevent both iron deficiency and iron excess. Our perception of iron metabolism has been completely changed by an improved definition of cellular and systemic iron homeostasis, of the molecular pathogenesis of iron disorders, the fine tuning of the iron hormone hepcidin by activators and inhibitors and the dissection of the components of the hepcidin regulatory pathway. Important for haematology, the crosstalk of erythropoiesis, the most important iron consumer, and the hepcidin pathway has been at least partially clarified. Novel potential biomarkers are available and novel therapeutic targets for iron-related disorders have been tested in murine models. These preclinical studies provided proofs of principle and are laying the ground for clinical trials. Understanding iron control in tissues other than erythropoiesis remains a challenge for the future.
Collapse
Affiliation(s)
- Clara Camaschella
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Vita Salute University, Milano, Italy
| | - Alessia Pagani
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Vita Salute University, Milano, Italy
| |
Collapse
|
44
|
Lynch S, Pfeiffer CM, Georgieff MK, Brittenham G, Fairweather-Tait S, Hurrell RF, McArdle HJ, Raiten DJ. Biomarkers of Nutrition for Development (BOND)-Iron Review. J Nutr 2018; 148:1001S-1067S. [PMID: 29878148 PMCID: PMC6297556 DOI: 10.1093/jn/nxx036] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/27/2017] [Accepted: 11/07/2017] [Indexed: 12/20/2022] Open
Abstract
This is the fifth in the series of reviews developed as part of the Biomarkers of Nutrition for Development (BOND) program. The BOND Iron Expert Panel (I-EP) reviewed the extant knowledge regarding iron biology, public health implications, and the relative usefulness of currently available biomarkers of iron status from deficiency to overload. Approaches to assessing intake, including bioavailability, are also covered. The report also covers technical and laboratory considerations for the use of available biomarkers of iron status, and concludes with a description of research priorities along with a brief discussion of new biomarkers with potential for use across the spectrum of activities related to the study of iron in human health.The I-EP concluded that current iron biomarkers are reliable for accurately assessing many aspects of iron nutrition. However, a clear distinction is made between the relative strengths of biomarkers to assess hematological consequences of iron deficiency versus other putative functional outcomes, particularly the relationship between maternal and fetal iron status during pregnancy, birth outcomes, and infant cognitive, motor and emotional development. The I-EP also highlighted the importance of considering the confounding effects of inflammation and infection on the interpretation of iron biomarker results, as well as the impact of life stage. Finally, alternative approaches to the evaluation of the risk for nutritional iron overload at the population level are presented, because the currently designated upper limits for the biomarker generally employed (serum ferritin) may not differentiate between true iron overload and the effects of subclinical inflammation.
Collapse
Affiliation(s)
| | - Christine M Pfeiffer
- National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA
| | - Michael K Georgieff
- Division of Neonatology, Department of Pediatrics, University of Minnesota School of Medicine, Minneapolis, MN
| | - Gary Brittenham
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, NY
| | - Susan Fairweather-Tait
- Department of Nutrition, Norwich Medical School, Norwich Research Park, University of East Anglia, Norwich NR4 7JT, UK
| | - Richard F Hurrell
- Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Harry J McArdle
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen AB21 9SB, UK
| | - Daniel J Raiten
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH)
| |
Collapse
|
45
|
Abstract
Measurement of serum ferritin (SF) is currently the laboratory test recommended for diagnosing iron deficiency. In the absence of an associated disease, a low SF value is an early and highly specific indicator of iron deficiency. The WHO criteria proposed to define depleted storage iron are 12μg/L for children under 5 years and 15μg/L for those over 5 years. A higher threshold of 30μg/L is used in the presence of infection or inflammation. Iron deficiency anemia, with typical low mean corpuscular volume and mean corpuscular hemoglobin, is only present at the end stage of iron deficiency. Other diagnostic tests for iron deficiency including iron parameters (low serum iron, increased total iron-binding capacity, low transferrin saturation) and erythrocyte traits (low mean corpuscular volume, increased zinc protoporphyrin) provide little additional diagnostic value over SF. In children, serum soluble transferrin receptor (sTfR) has been reported to be a sensitive indicator of iron deficiency and is relatively unaffected by inflammation. On the other hand, sTfR is directly related to extent of erythroid activity and not commonly used in clinical practice. In population surveys, approaches based on combinations of markers have been explored to improve the specificity and sensitivity of diagnostic. In addition to Hb value determination, a combination of parameters (among transferrin saturation, zinc protoporphyrin, mean corpuscular volume or serum ferritin) was generally used to assess iron deficiency. More recently sTfR/ ferritin index were evaluated, sTfR in conjunction with SF allowing to better distinguishing iron deficiency from inflammatory anemia. Also, hepcidin measurements appeared an interesting marker for diagnosing iron deficiency and identifying individuals in need of iron supplementation in populations where inflammatory or infectious diseases are frequently encountered. Reticulocyte Hb content (CHr) determination is an early parameter of iron deficiency erythropoiesis. CHr can be measured with several automated hematology analyzers and so, used for individual's iron status assessment. In addition to Hb concentration determination, individual's iron status is commonly assessed in the pediatric clinical practice by the SF measurement accompanied by the determination of C-reactive protein for detection of a simultaneous acute infection and/or inflammation.
Collapse
Affiliation(s)
- I Thuret
- Service d'onco-hématologie pédíatrique, CHU Timone Enfants, centre de référence des thalassémies, 264, rue Saint-Pierre, 13005 Marseille, France.
| |
Collapse
|
46
|
Abioye AI, Park S, Ripp K, McDonald EA, Kurtis JD, Wu H, Pond-Tor S, Sharma S, Ernerudh J, Baltazar P, Acosta LP, Olveda RM, Tallo V, Friedman JF. Anemia of Inflammation during Human Pregnancy Does Not Affect Newborn Iron Endowment. J Nutr 2018; 148:427-436. [PMID: 29546300 PMCID: PMC6454452 DOI: 10.1093/jn/nxx052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/27/2017] [Indexed: 12/20/2022] Open
Abstract
Background To our knowledge, no studies have addressed whether maternal anemia of inflammation (AI) affects newborn iron status, and few have addressed risk factors for specific etiologies of maternal anemia. Objectives The study aims were to evaluate 1) the contribution of AI and iron deficiency anemia (IDA) to newborn iron endowment, 2) hepcidin as a biomarker to distinguish AI from IDA among pregnant women, and 3) risk factors for specific etiologies of maternal anemia. Methods We measured hematologic biomarkers in maternal blood at 12 and 32 wk of gestation and in cord blood from a randomized trial of praziquantel in 358 pregnant women with Schistosoma japonicum in The Philippines. IDA was defined as anemia with serum ferritin <30 ng/mL and non-IDA (NIDA), largely due to AI, as anemia with ferritin ≥30 ng/mL. We identified cutoffs for biomarkers to distinguish IDA from NIDA by using area under the curve (AUC) analyses and examined the impact of different causes of anemia on newborn iron status (primary outcome) by using multivariate regression modeling. Results Of the 358 mothers, 38% (n = 136) had IDA and 9% (n = 32) had NIDA at 32 wk of gestation. At 32 wk of gestation, serum hepcidin performed better than soluble transferrin receptor (sTfR) in identifying women with NIDA compared with the rest of the cohort (AUCs: 0.75 and 0.70, respectively) and in identifying women with NIDA among women with anemia (0.73 and 0.72, respectively). The cutoff that optimally distinguished women with NIDA from women with IDA in our cohort was 6.1 µg/L. Maternal IDA, but not NIDA, was associated with significantly lower newborn ferritin (114.4 ng/mL compared with 148.4 µg/L; P = 0.042). Conclusions Hepcidin performed better than sTfR in identifying pregnant women with NIDA, but its cost may limit its use. Maternal IDA, but not NIDA, is associated with decreased newborn iron stores, emphasizing the need to identify this cause and provide iron therapy. This trial was registered at www.clinicaltrials.gov as NCT00486863.
Collapse
Affiliation(s)
- Ajibola I Abioye
- The Warren Alpert Medical School of Brown University, Providence, RI,Department of Pediatrics, Center for International Health Research, and Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, RI,Department of Center for International Health Research, and Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, RI
| | - Sangshin Park
- The Warren Alpert Medical School of Brown University, Providence, RI,Department of Pediatrics, Center for International Health Research, and Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, RI,Department of Center for International Health Research, and Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, RI,Address correspondence to SP (e-mail: or )
| | - Kelsey Ripp
- The Warren Alpert Medical School of Brown University, Providence, RI
| | - Emily A McDonald
- The Warren Alpert Medical School of Brown University, Providence, RI,Department of Pediatrics, Center for International Health Research, and Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, RI,Department of Center for International Health Research, and Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, RI
| | - Jonathan D Kurtis
- The Warren Alpert Medical School of Brown University, Providence, RI,Department of Center for International Health Research, and Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, RI,Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, RI
| | - Hannah Wu
- The Warren Alpert Medical School of Brown University, Providence, RI,Department of Pediatrics, Center for International Health Research, and Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, RI,Department of Center for International Health Research, and Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, RI
| | - Sunthorn Pond-Tor
- Department of Center for International Health Research, and Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, RI
| | - Surendra Sharma
- The Warren Alpert Medical School of Brown University, Providence, RI,Department of Pediatrics, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Jan Ernerudh
- Departments of Clinical Immunology and Transfusion Medicine and Clinical and Experimental Medicine, Linkoping University, Linkoping, Sweden,Departments of Clinical and Experimental Medicine, Linkoping University, Linkoping, Sweden
| | - Palmera Baltazar
- Research Institute for Tropical Medicine, Manila, Philippines,Remedios Trinidad Romualdez Hospital, Tacloban City, Leyte, Philippines
| | - Luz P Acosta
- Research Institute for Tropical Medicine, Manila, Philippines
| | | | - Veronica Tallo
- Research Institute for Tropical Medicine, Manila, Philippines
| | - Jennifer F Friedman
- The Warren Alpert Medical School of Brown University, Providence, RI,Department of Pediatrics, Center for International Health Research, and Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, RI,Department of Center for International Health Research, and Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, RI
| |
Collapse
|
47
|
Approaches for Reducing the Risk of Early-Life Iron Deficiency-Induced Brain Dysfunction in Children. Nutrients 2018; 10:nu10020227. [PMID: 29462970 PMCID: PMC5852803 DOI: 10.3390/nu10020227] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/12/2018] [Accepted: 02/14/2018] [Indexed: 12/23/2022] Open
Abstract
Iron deficiency is the most common micronutrient deficiency in the world. Women of reproductive age and young children are particularly vulnerable. Iron deficiency in late prenatal and early postnatal periods can lead to long-term neurobehavioral deficits, despite iron treatment. This may occur because screening and treatment of iron deficiency in children is currently focused on detection of anemia and not neurodevelopment. Anemia is the end-stage state of iron deficiency. The brain becomes iron deficient before the onset of anemia due to prioritization of the available iron to the red blood cells (RBCs) over other organs. Brain iron deficiency, independent of anemia, is responsible for the adverse neurological effects. Early diagnosis and treatment of impending brain dysfunction in the pre-anemic stage is necessary to prevent neurological deficits. The currently available hematological indices are not sensitive biomarkers of brain iron deficiency and dysfunction. Studies in non-human primate models suggest that serum proteomic and metabolomic analyses may be superior for this purpose. Maternal iron supplementation, delayed clamping or milking of the umbilical cord, and early iron supplementation improve the iron status of at-risk infants. Whether these strategies prevent iron deficiency-induced brain dysfunction has yet to be determined. The potential for oxidant stress, altered gastrointestinal microbiome and other adverse effects associated with iron supplementation cautions against indiscriminate iron supplementation of children in malaria-endemic regions and iron-sufficient populations.
Collapse
|
48
|
Daru J, Colman K, Stanworth SJ, De La Salle B, Wood EM, Pasricha SR. Serum ferritin as an indicator of iron status: what do we need to know? Am J Clin Nutr 2017; 106:1634S-1639S. [PMID: 29070560 PMCID: PMC5701723 DOI: 10.3945/ajcn.117.155960] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Determination of iron status in pregnancy and in young children is essential for both clinical and public health practice. Clinical diagnosis of iron deficiency (ID) through sampling of bone marrow to identify the absence of body iron stores is impractical in most cases. Serum ferritin (SF) concentrations are the most commonly deployed indicator for determining ID, and low SF concentrations reflect a state of iron depletion. However, there is considerable variation in SF cutoffs recommended by different expert groups to diagnose ID. Moreover, the cutoffs used in different clinical laboratories are heterogeneous. There are few studies of diagnostic test accuracy to establish the sensitivity and specificity of SF compared with key gold standards (such as absent bone marrow iron stores, increased intestinal iron absorption, and hemoglobin response to SF) among noninflamed, outpatient populations. The limited data available suggest the commonly recommended SF cutoff of <15 μg/L is a specific but not sensitive cutoff, although evidence is limited. Data from women during pregnancy or from young children are especially uncommon. Most data are from studies conducted >30 y ago, do not reflect ethnic or geographic diversity, and were performed in an era for which laboratory methods no longer reflect present practice. Future studies to define the appropriate SF cutoffs are urgently needed and would also provide an opportunity to compare this indicator with other established and emerging iron indexes. In addition, future work would benefit from a focus on elucidating cutoffs and indexes relevant to iron adequacy.
Collapse
Affiliation(s)
- Jahnavi Daru
- Women's Health Research Unit, Barts and the London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Katherine Colman
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - Simon J Stanworth
- NHS Blood and Transplant/Oxford University Hospitals NHS Trust, Oxford, United Kingdom
| | - Barbara De La Salle
- United Kingdom National External Quality Assessment Service, General Haematology, Watford, United Kingdom; and
| | - Erica M Wood
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - Sant-Rayn Pasricha
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, United Kingdom
| |
Collapse
|
49
|
Allen A, Allen S, Rodrigo R, Perera L, Shao W, Li C, Wang D, Olivieri N, Weatherall DJ, Premawardhena A. Iron status and anaemia in Sri Lankan secondary school children: A cross-sectional survey. PLoS One 2017; 12:e0188110. [PMID: 29155855 PMCID: PMC5695819 DOI: 10.1371/journal.pone.0188110] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/01/2017] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Iron deficiency, the most common micronutrient disorder and cause of anaemia globally, impairs growth, cognition, behaviour and resistance to infection. METHODS/RESULTS As part of a national survey of inherited haemoglobin variants in 7526 students from 72 secondary schools purposefully selected from the 25 districts of Sri Lanka, we studied 5912 students with a normal haemoglobin genotype. Median age was 16.0 (IQR 15.0-17.0) years and 3189 (53.9%) students were males. Most students were Sinhalese (65.7%), with fewer Tamils (23.1%) and Muslims (11.2%). Anaemia occurred in 470 students and was more common in females (11.1%) than males (5.6%). Haemoglobin, serum ferritin, transferrin receptor and iron were determined in 1196 students with low red cell indices and a structured sample of those with normal red cell indices (n = 513). The findings were weighted to estimate the frequencies of iron deficiency and iron deficiency anaemia classified according to WHO criteria. Iron depletion (serum ferritin <15ug/ml) occurred in 19.2% and cellular iron deficiency (low serum ferritin and transferrin receptor >28.1 nmol/l) in 11.6% students. Iron deficiency anaemia (cellular iron deficiency with low haemoglobin) occurred in only 130/2794 (4.6%) females and 28/2789 (1.0%) males. Iron biomarkers were normal in 83/470 (14.6%) students with anaemia. In multiple regression analysis, the odds for iron depletion and cellular iron deficiency were about one-third in males compared with females, and the odds for iron deficiency anaemia were about one fifth in males compared to females. Tamil ethnicity and age <16 years increased the risk of all three stages of iron deficiency and living at high altitude significantly reduced the risk of iron depletion. CONCLUSIONS Low iron status and anaemia remain common problems in Sri Lankan secondary school students especially females, younger students and the socioeconomically disadvantaged Tamil population. More research is needed to identify factors other than low iron status that contribute to anaemia in adolescents.
Collapse
Affiliation(s)
- Angela Allen
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- * E-mail:
| | - Stephen Allen
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Rexan Rodrigo
- Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Lakshman Perera
- Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Wei Shao
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Second Military Medical University, Shanghai, China
| | - Chao Li
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Duolao Wang
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Nancy Olivieri
- Department of Medicine and Public Health Sciences, University of Toronto, Toronto, Canada
| | - David J. Weatherall
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
50
|
Ghatpande NS, Apte PP, Naik SS, Joshi BN, Gokhale MK, Kulkarni PP. Association of B12 deficiency and anemia synergistically increases the risk of high TNF-α levels among adolescent girls. Metallomics 2017; 8:734-8. [PMID: 27346169 DOI: 10.1039/c6mt00129g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We investigated the association between iron status, B12, and inflammatory markers among 101 adolescent girls. We found that B12 showed significant negative association with tumor necrosis factor-alpha (TNF-α) (rs = -0.232, P = 0.020) and positive association with serum ferritin (SF) (rs = 0.209, P = 0.036) among girls. Our results showed that hepcidin discriminates anemic and non-anemic population under normal B12 conditions. The logistic regression analysis revealed that the risk of having higher TNF-α levels was 13.2 times higher in low B12 girls in the presence of anemia compared to the girls having normal hemoglobin and B12 levels.
Collapse
Affiliation(s)
- N S Ghatpande
- Bioprospecting Group, Agharkar Research Institute, Pune, 411004, India.
| | - P P Apte
- Bioprospecting Group, Agharkar Research Institute, Pune, 411004, India.
| | - S S Naik
- K. E. M. Hospital and Research Center, Pune, 411011, India
| | - B N Joshi
- Bioprospecting Group, Agharkar Research Institute, Pune, 411004, India.
| | - M K Gokhale
- Bioprospecting Group, Agharkar Research Institute, Pune, 411004, India.
| | - P P Kulkarni
- Bioprospecting Group, Agharkar Research Institute, Pune, 411004, India.
| |
Collapse
|