1
|
Larson LM, Baldi A, Hasan MI, Jones N, Owens Pickle E, Ataide R, Bennett C, Bhuiyan MSA, Tipu SMU, Tofail F, Biggs BA, Hamadani J, Pasricha SR, Braat S. Role of hepcidin on benefits and risks of supplementation with iron syrup and multiple micronutrient powders in Bangladeshi children: a substudy to a randomized controlled trial. Am J Clin Nutr 2025; 121:1090-1098. [PMID: 39983916 DOI: 10.1016/j.ajcnut.2025.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 02/07/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Hepcidin can determine individuals' responses to iron supplementation, but limited evidence exists from pediatric trials. OBJECTIVES We aimed to examine the influence of hepcidin on the effects of supplementation with iron syrup and multiple micronutrient powders (MNPs) on hemoglobin and ferritin concentrations and the incidence of diarrhea in young children. METHODS Participants included a subsample of 1281 8-mo-old children enrolled in a 3-arm, double-blind, double-dummy, individually randomized controlled trial examining the efficacy of 3 mo of universal supplementation with daily iron syrup (12.5 mg iron), MNPs (containing 12.5 mg iron), or placebo in children living in Bangladesh. In all participants at baseline, immediately postintervention (month 3), and after a further 9 mo of follow-up (month 12), serum hepcidin concentrations were measured by enzyme-linked immunosorbent assay, venous hemoglobin by HemoCue 301, and incidence of diarrhea by caregiver report. We used a likelihood-based longitudinal data analysis model to examine effect modification from baseline hepcidin on the effects of iron syrup or MNPs on hemoglobin and ferritin concentrations and a log-binomial model on the incidence of diarrhea at months 3 and 12. RESULTS Hepcidin modified the effect of MNPs, but not iron syrup, compared with placebo on hemoglobin and ferritin concentrations immediately postintervention. The treatment effect of MNPs compared with placebo in the change from baseline to month 3 was larger among children with low compared with not-low baseline hepcidin [hemoglobin: mean difference 11.6 g/L (7.2, 15.9) compared with 4.3 (3.09, 5.7), P-interaction = 0.002; ferritin: geometric mean ratio 2.4 (1.6, 3.6) compared with 1.5 (1.3, 1.7), P-interaction = 0.024]. This effect modification was not sustained at month 12. Hepcidin did not modify the effects of either intervention on the incidence of diarrhea. CONCLUSIONS Immediate effects of MNPs on child hemoglobin and iron status are larger among those with low compared with not-low hepcidin, indicating that preintervention screening could help identify those who would benefit most from MNPs. This trial was registered at the Australian New Zealand Clinical Trials Registry as ACTRN12617000660381.
Collapse
Affiliation(s)
- Leila M Larson
- Department of Health Promotion, Education, and Behavior, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States.
| | - Andrew Baldi
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Mohammed Imrul Hasan
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Naomi Jones
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Emily Owens Pickle
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States
| | - Ricardo Ataide
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; Department of Infectious Diseases at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Cavan Bennett
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | | | - Sm Mulk Uddin Tipu
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Fahmida Tofail
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Beverley-Ann Biggs
- Department of Infectious Diseases at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Jena Hamadani
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Sant-Rayn Pasricha
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
| | - Sabine Braat
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia; Department of Infectious Diseases at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
2
|
Mutumba R, Mbabazi J, Pesu H, Lewis JI, Mølgaard C, Ritz C, Olsen MF, Briend A, Nabukeera-Barungi N, Wells JC, Friis H, Grenov B, Mupere E. Effect of lipid-based nutrient supplements on morbidity among children with stunting: secondary analysis of a randomized trial in Uganda. Eur J Clin Nutr 2025:10.1038/s41430-025-01611-3. [PMID: 40164777 DOI: 10.1038/s41430-025-01611-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/06/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Children with stunting are at risk of infections. We assessed the effect of lipid-based nutrient supplement (LNS) on morbidity in children with stunting. METHODS This was a secondary analysis of a randomized, 2×2 factorial trial among 12-59 months-old, stunted children in Uganda. Children were randomized to LNS containing milk or soy protein and whey permeate or maltodextrin, or no supplementation, for 12 weeks. The outcomes were caregiver-reported morbidity after 2, 4, 8 and 12 weeks, serum C-reactive protein (S-CRP), α1-acid glycoprotein (S-AGP), and phase-angle (PhA) by bioimpedance. RESULTS Of 750 children, mean (SD) age was 32.0 (11.7) months, 55% (n = 412) were male. LNS increased diarrhoea prevalence (18.1% vs 7.3%, P = 0.001) during the first two weeks, but not thereafter. There was no effect of LNS on cough or fever. LNS resulted in greater decline in S-AGP (-0.10 g/L, 95% CI: -0.17, -0.03, P = 0.003) but not S-CRP (25%, 95% CI: -11, 74, P = 0.193), and greater increase in PhA (0.10 degrees, 95% CI: 0.01, 0.18, P = 0.030), explained by greater fat-free mass. Milk compared to soy protein in LNS resulted in higher PhA (0.10 degrees, 95% CI: 0.02, 0.17, P = 0.013), not explained by fat-free mass. CONCLUSION LNS supplementation in children with stunting had no effect on morbidity but resulted in a small reduction in sub-acute systemic inflammation. The possible effect of LNS supplementation on inflammation in stunted children requires further evaluation. ( www.isrctn.com : ISRCTN13093195).
Collapse
Affiliation(s)
- Rolland Mutumba
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda.
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.
| | - Joseph Mbabazi
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Hannah Pesu
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jack I Lewis
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Christian Mølgaard
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Christian Ritz
- The National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Mette F Olsen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Andre Briend
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
- Tampere Centre for Child Health Research, Tampere University, Tampere, Finland
| | - Nicolette Nabukeera-Barungi
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Jonathan C Wells
- Childhood Nutrition Research Centre, Population Policy and Practice Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Henrik Friis
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Benedikte Grenov
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Ezekiel Mupere
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
3
|
Lei W, Liu Z, Lai HP, Fu R. Gut microbiota and risk of iron deficiency anemia: A two-sample Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41617. [PMID: 39993092 PMCID: PMC11856941 DOI: 10.1097/md.0000000000041617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
Previous studies have suggested a link between gut microbiota and iron-deficiency anemia (IDA). However, interpreting these findings is difficult due to various factors that influence microbiome composition and the limitations of observational studies, such as confounding variables and reverse causation. This study aims to explore the causal relationship between gut microbiota and IDA using Mendelian randomization (MR) to overcome these limitations. We conducted a 2-sample MR analysis using data from genome-wide association studies from the MiBioGen Consortium and the UK Biobank. The gut microbiome data included 211 genus-level microbes linked to single-nucleotide polymorphisms from 18,340 participants in the MiBioGen Consortium. The outcome data for IDA were obtained from 484,598 participants in the UK Biobank, with 2941 cases and 481,657 controls. We assessed causal relationships using various MR techniques, primarily inverse variance weighting, and performed sensitivity analyses to confirm the robustness of our results. Nine genus-level gut microbes were significantly associated with IDA (P < .05). Protective factors included Clostridia, Actinomycetaceae, Pasteurellaceae, Oscillospira, Prevotella, and Roseburia, while risk factors included Ruminococcus gnavus group, Hungatella, and Parasutterella. Sensitivity analyses showed the reliability of these findings without significant variability. This study provides evidence for a causal relationship between specific gut bacteria and IDA risk, identifying potential targets for therapies aimed at improving outcomes for those with IDA. Further research is needed to clarify the bacteria involved.
Collapse
Affiliation(s)
- Wenhui Lei
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, PR China
- Tianjin Institute of Hematology, Tianjin, China
- Department of Internal Medicine, Lishui Municipal Central Hospital, Lishui, Zhejiang Province, PR China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
| | - Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, PR China
- Tianjin Institute of Hematology, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
| | - Hai-Ping Lai
- Department of Medicine, Ganzhou Tumor Hospital, Ganzhou, Jiangxi Province, PR China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, PR China
- Tianjin Institute of Hematology, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
| |
Collapse
|
4
|
Helmyati S, Lusmilasari L, Sandhi A, Hardiyanti M, Rosilia G, Rachmawati YN, Aristyarini M. Systematic review on supplementation, fortification, and food-based interventions for preventing iron deficiency anemia in low- and middle-income countries. Asia Pac J Clin Nutr 2025; 34:10-35. [PMID: 39828255 PMCID: PMC11742596 DOI: 10.6133/apjcn.202502_34(1).0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/14/2024] [Accepted: 07/09/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND AND OBJECTIVES Prioritizing key preventive and therapeutic interventions is one of the actions to accelerate the reduction of anemia. This study aimed to examine interventions designed to prevent anemia. METHODS AND STUDY DESIGN A systematic search was conducted in PubMed, Web of Science, Scopus, and Cochrane Library. Analysis of publication bias was done using The Joanna-Briggs Institute critical appraisal tool. Data collected from articles included author, year of publication, setting and location of the study, study type, participant of the study, intervention and control given, main outcome, main findings, and risk of bias. RESULTS Three nutrition-specific interventions aimed at preventing iron deficiency anemia in low- and middle-income countries used various types and dosages of iron. While most studies showed success, some indicated a worsening trend in anemia, even with standard dosages and the same form of iron. Determining effective interventions requires consideration of factors such as other micronutrient composition, compliance rate, availability of educational intervention, and dietary backgrounds in those countries. CONCLUSIONS Supplementation, fortification, and food-based interventions generally lead to higher hemoglobin levels and a lower prevalence of anemia. However, it is important to consider several factors before deciding on an approach.
Collapse
Affiliation(s)
- Siti Helmyati
- Department of Nutrition and Health, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia. ;
- Center for Health and Human Nutrition, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Lely Lusmilasari
- Center for Health and Human Nutrition, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Department of Pediatrics and Maternity Nursing, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Ayyu Sandhi
- Department of Pediatrics and Maternity Nursing, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Marina Hardiyanti
- Department of Nutrition and Health, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Gifani Rosilia
- Center for Health and Human Nutrition, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Yuliana Novita Rachmawati
- Center for Health and Human Nutrition, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Mitha Aristyarini
- Center for Health and Human Nutrition, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
5
|
Wunderlich J, Kotov V, Votborg-Novél L, Ntalla C, Geffken M, Peine S, Portugal S, Strauss J. Iron transport pathways in the human malaria parasite Plasmodium falciparum revealed by RNA-sequencing. Front Cell Infect Microbiol 2024; 14:1480076. [PMID: 39575308 PMCID: PMC11578967 DOI: 10.3389/fcimb.2024.1480076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/14/2024] [Indexed: 11/24/2024] Open
Abstract
Host iron deficiency is protective against severe malaria as the human malaria parasite Plasmodium falciparum depends on bioavailable iron from its host to proliferate. The essential pathways of iron acquisition, storage, export, and detoxification in the parasite differ from those in humans, as orthologs of the mammalian transferrin receptor, ferritin, or ferroportin, and a functional heme oxygenase are absent in P. falciparum. Thus, the proteins involved in these processes may be excellent targets for therapeutic development, yet remain largely unknown. Here, we show that parasites cultured in erythrocytes from an iron-deficient donor displayed significantly reduced growth rates compared to those grown in red blood cells from healthy controls. Sequencing of parasite RNA revealed diminished expression of genes involved in overall metabolism, hemoglobin digestion, and metabolite transport under low-iron versus control conditions. Supplementation with hepcidin, a specific ferroportin inhibitor, resulted in increased labile iron levels in erythrocytes, enhanced parasite replication, and transcriptional upregulation of genes responsible for merozoite motility and host cell invasion. Through endogenous GFP tagging of differentially expressed putative transporter genes followed by confocal live-cell imaging, proliferation assays with knockout and knockdown lines, and protein structure predictions, we identified six proteins that are likely required for ferrous iron transport in P. falciparum. Of these, we localized PfVIT and PfZIPCO to cytoplasmic vesicles, PfMRS3 to the mitochondrion, and the novel putative iron transporter PfE140 to the plasma membrane for the first time in P. falciparum. PfNRAMP/PfDMT1 and PfCRT were previously reported to efflux Fe2+ from the digestive vacuole. Our data support a new model for parasite iron homeostasis, in which PfE140 is involved in iron uptake across the plasma membrane, PfMRS3 ensures non-redundant Fe2+ supply to the mitochondrion as the main site of iron utilization, PfVIT transports excess iron into cytoplasmic vesicles, and PfZIPCO exports Fe2+ from these organelles in case of iron scarcity. These results provide new insights into the parasite's response to differential iron availability in its environment and into the mechanisms of iron transport in P. falciparum as promising candidate targets for future antimalarial drugs.
Collapse
Affiliation(s)
- Juliane Wunderlich
- Malaria Parasite Biology Group, Max Planck Institute for Infection Biology (MPIIB), Berlin, Germany
- Membrane Protein Structural Biology Group, Center for Structural Systems Biology (CSSB), Hamburg, Germany
| | - Vadim Kotov
- Membrane Protein Structural Biology Group, Center for Structural Systems Biology (CSSB), Hamburg, Germany
| | - Lasse Votborg-Novél
- Malaria Parasite Biology Group, Max Planck Institute for Infection Biology (MPIIB), Berlin, Germany
| | - Christina Ntalla
- Malaria Parasite Biology Group, Max Planck Institute for Infection Biology (MPIIB), Berlin, Germany
| | - Maria Geffken
- Institute of Transfusion Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Sven Peine
- Institute of Transfusion Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Silvia Portugal
- Malaria Parasite Biology Group, Max Planck Institute for Infection Biology (MPIIB), Berlin, Germany
| | - Jan Strauss
- Membrane Protein Structural Biology Group, Center for Structural Systems Biology (CSSB), Hamburg, Germany
| |
Collapse
|
6
|
Yang C, Xiao J, Xu Z, Wang Z. Gut Microbiota Changes and Its Potential Relations with Thyroid Disorders: From Composition to Therapeutic Targets. Int J Gen Med 2024; 17:3719-3731. [PMID: 39219667 PMCID: PMC11363920 DOI: 10.2147/ijgm.s481183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Composed of over 1200 species of anaerobes and aerobes bacteria along with bacteriophages, viruses, and fungal species, the human gut microbiota (GM) is vital to health, including digestive equilibrium, immunologic, hormonal, and metabolic homeostasis. Micronutrients, usually refer to trace elements (copper, iodine, iron, selenium, zinc) and vitamins (A, C, D, E), interact with the GM to influence host immune metabolism. So far, microbiome studies have revealed an association between disturbances in the microbiota and various pathological disorders, such as anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis, anxiety, depression, early-onset cancers, type 1 diabetes (T1D) and type 2 diabetes (T2D). As common conditions, thyroid diseases, encompassing Graves' disease (GD), Graves' orbitopathy (GO), Hashimoto's thyroiditis (HT), benign nodules, and papillary thyroid cancer (TC), have negative impacts on the health of all populations. Following recent studies, GM might play an integral role in triggering diseases of the thyroid gland. Not only do environmental triggers and genetic predisposing background lead to auto-aggressive damage, involving cellular and humoral networks of the immune system, but the intestinal microbiota interacts with distant organs by signals that may be part of the bacteria themselves or their metabolites. The review aims to describe the current knowledge about the GM in the metabolism of thyroid hormones and the pathogenesis of thyroid diseases and its involvement in the appearance of benign nodules and papillary TC. We further focused on the reciprocal interaction between GM composition and the most used treatment drugs for thyroid disorders. However, the exact etiology has not yet been known. To elucidate more precisely the mechanism for GM involvement in the development of thyroid diseases, future work is needed.
Collapse
Affiliation(s)
- Cai Yang
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| | - Jiafeng Xiao
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| | - Zibei Xu
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| | - Zehong Wang
- Department of Laboratory Medicine, Medical Center Hospital of Qionglai City, Chengdu, Sichuan, 611530, People’s Republic of China
| |
Collapse
|
7
|
Wu Y, Li G, Tang H. Antibiotics Trigger Host Innate Immune Response via Microbiota-Brain Communication in C. elegans. Int J Mol Sci 2024; 25:8866. [PMID: 39201552 PMCID: PMC11354627 DOI: 10.3390/ijms25168866] [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: 07/08/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Besides their direct bactericidal effect, antibiotics have also been suggested to stimulate the host immune response to defend against pathogens. However, it remains unclear whether any antibiotics may stimulate the host immune response by affecting bacterial activity. In this study, reasoning that genetic mutations inhibit bacterial activities and, thereby, may mimic the effects of antibiotics, we performed genome-wide screening and identified 77 E. coli genes whose inactivation induces C. elegans cyp-14A4, representing an innate immune and detoxification response. Further analyses reveal that this host immune response can clearly be induced through either inactivating the E. coli respiratory chain via the bacterial cyoB mutation or using the antibiotic Q203, which is able to enhance host survival when encountering the pathogen Pseudomonas aeruginosa. Mechanistically, the innate immune response triggered by both the cyoB mutation and Q203 is found to depend on the host brain response, as evidenced by their reliance on the host neural gene unc-13, which is required for neurotransmitter release in head neurons. Therefore, our findings elucidate the critical involvement of the microbiota-brain axis in modulating the host immune response, providing mechanistic insights into the role of antibiotics in triggering the host immune response and, thus, facilitating host defense against pathogens.
Collapse
Affiliation(s)
- Yangyang Wu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China;
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Guanqun Li
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Hongyun Tang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China
| |
Collapse
|
8
|
Palmer AC, Bedsaul-Fryer JR, Stephensen CB. Interactions of Nutrition and Infection: The Role of Micronutrient Deficiencies in the Immune Response to Pathogens and Implications for Child Health. Annu Rev Nutr 2024; 44:99-124. [PMID: 38724105 DOI: 10.1146/annurev-nutr-062122-014910] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
Approximately five million children die each year from preventable causes, including respiratory infections, diarrhea, and malaria. Roughly half of those deaths are attributable to undernutrition, including micronutrient deficiencies (MNDs). The influence of infection on micronutrient status is well established: The inflammatory response to pathogens triggers anorexia, while pathogens and the immune response can both alter nutrient absorption and cause nutrient losses. We review the roles of vitamin A, vitamin D, iron, zinc, and selenium in the immune system, which act in the regulation of molecular- or cellular-level host defenses, directly affecting pathogens or protecting against oxidative stress or inflammation. We further summarize high-quality evidence regarding the synergistic or antagonistic interactions between MNDs, pathogens, and morbidity or mortality relevant to child health in low- and middle-income countries. We conclude with a discussion of gaps in the literature and future directions for multidisciplinary research on the interactions of MNDs, infection, and inflammation.
Collapse
Affiliation(s)
- Amanda C Palmer
- Center for Human Nutrition, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA;
| | - Jacquelyn R Bedsaul-Fryer
- Cancer Prevention Fellowship Program, Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland, USA
- Center for Human Nutrition, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA;
| | - Charles B Stephensen
- Department of Nutrition, University of California, Davis, California, USA
- Western Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Davis, California, USA
| |
Collapse
|
9
|
Chouraqui JP, Renella R, Turck D. Assessment of Dietary Iron Intake and Sources in Early Childhood in a Nationally Representative Cross-Sectional Survey. J Acad Nutr Diet 2024; 124:823-832.e1. [PMID: 38142742 DOI: 10.1016/j.jand.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND Iron deficiency has particular importance in early childhood because of its impact on growth and development. Preventive food-based strategies of iron deficiency require knowledge of intakes and sources of iron. OBJECTIVE This study aimed to assess daily iron intakes (DIIs) in 2013, to compare them with the dietary reference values, to assess their evolution since 1981, and to identify iron food sources among nonbreastfed French children younger than 3 years. DESIGN This was a nationwide cross-sectional survey conducted in 2013 in France to assess DIIs by means of comparing them with the dietary reference values. Parents' reported diet diaries were collected for 3 nonconsecutive days. PARTICIPANTS/SETTING Of the 1,184 children enrolled in the study, 1,035 nonbreastfed healthy children aged 0.5 to 35 months stratified into 11 age groups were included after informed consent was obtained from parents and according to a weighted quota sampling method. MAIN OUTCOME MEASURES DII from the different food sources and trends in their evolution from 1981 to 2013 was assessed. STATISTICAL ANALYSES PERFORMED Results are expressed as median with interquartile range and range and mean ± SD. Student t test was used with the 2-sided α level of significance set at 5%. RESULTS Mean ± SD DII was 6.7 ± 2.3 mg/d before 6 months, 8.2 ± 2.7 mg/d from 6 months to 1 year, and 7.0 ± 3.2 mg/d from 1 to 3 years. The prevalence of infants older than 7 months with a DII less than the dietary reference values was 52.5%, and that of young children was 30%. After slightly increasing until 2005, DIIs decreased thereafter. Formulas contributed to most of the DIIs up to 2 years of age. Cereals were the second largest contributor to DIIs, and meat accounted for a small part of iron intake. CONCLUSIONS A substantial number of children between 6 months and 3 years of age were at risk of insufficient iron intake. This risk increased from 2005 to 2013. The role of formula in ensuring iron intake is highlighted. More research on health outcomes of low iron intakes is needed.
Collapse
Affiliation(s)
- Jean-Pierre Chouraqui
- Paediatric Division of Nutrition and Gastroenterology, Pediatric Department, Grenoble-Alpes University Hospital, Grenoble, France.
| | - Raffaele Renella
- Pediatric Hematology-Oncology Unit, Division of Pediatrics, Department Woman-Mother-Child, Lausanne University Hospital, Switzerland
| | - Dominique Turck
- Department of Pediatrics, University of Lille, and INSERM U1286, Institute for Translational Research in Inflammation, Lille, France
| |
Collapse
|
10
|
Seyoum Y, Greffeuille V, Kouadio DKD, Kuong K, Turpin W, M'Rabt R, Chochois V, Fortin S, Perignon M, Fiorentino M, Berger J, Burja K, Ponce MC, Chamnan C, Wieringa FT, Humblot C. Faecal microbiota of schoolchildren is associated with nutritional status and markers of inflammation: a double-blinded cluster-randomized controlled trial using multi-micronutrient fortified rice. Nat Commun 2024; 15:5204. [PMID: 38890302 PMCID: PMC11189458 DOI: 10.1038/s41467-024-49093-4] [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: 11/10/2022] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
Faecal microbiota plays a critical role in human health, but its relationship with nutritional status among schoolchildren remains under-explored. Here, in a double-blinded cluster-randomized controlled trial on 380 Cambodian schoolchildren, we characterize the impact of six months consumption of two types of rice fortified with different levels of vitamins and minerals on pre-specified outcomes. We investigate the association between the faecal microbiota (16SrRNA sequencing) and age, sex, nutritional status (underweight, stunting), micronutrient status (iron, zinc and vitamin A deficiencies, anaemia, iron deficient anaemia, hemoglobinopathy), inflammation (systemic, gut), and parasitic infection. We show that the faecal microbiota is characterised by a surprisingly high proportion of Lactobacillaceae. We discover that deficiencies in specific micronutrients, such as iron and vitamin A, correlate with particular microbiota profiles, whereas zinc deficiency shows no such association. The nutritional intervention with the two rice treatments impacts both the composition and functions predicted from compositional analysis in different ways. (ClinicalTrials.gov (Identifier: NCT01706419)).
Collapse
Affiliation(s)
- Yohannes Seyoum
- Qualisud, Univ Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de La Réunion, Montpellier, France
| | - Valérie Greffeuille
- Qualisud, Univ Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de La Réunion, Montpellier, France
| | - Dorgeles Kouakou Dje Kouadio
- Qualisud, Univ Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de La Réunion, Montpellier, France
| | - Khov Kuong
- Department of Fisheries Post-Harvest Technologies and Quality Control, Ministry of Agriculture, Forestry and Fisheries, Phnom Penh, Cambodia
| | - Williams Turpin
- Qualisud, Univ Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de La Réunion, Montpellier, France
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, ON, Canada
- Division of Gastroenterology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Rachida M'Rabt
- Qualisud, Univ Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de La Réunion, Montpellier, France
| | - Vincent Chochois
- Qualisud, Univ Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de La Réunion, Montpellier, France
| | - Sonia Fortin
- Qualisud, Univ Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de La Réunion, Montpellier, France
| | - Marlène Perignon
- Qualisud, Univ Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de La Réunion, Montpellier, France
- MoISA, Univ Montpellier, CIHEAM-IAMM, CIRAD, INRAE, Institut Agro, IRD, Montpellier, France
| | - Marion Fiorentino
- Qualisud, Univ Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de La Réunion, Montpellier, France
- SESSTIM, INSERM, IRD, Aix Marseille Univ, Marseille, France
| | - Jacques Berger
- Qualisud, Univ Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de La Réunion, Montpellier, France
| | - Kurt Burja
- United Nations World Food Programme, Phnom Penh, Cambodia
| | - Maiza Campos Ponce
- Section Infectious Diseases, department of Health Sciences, Faculty of Earth and Life Sciences, VU University Amsterdam, Amsterdam, The Netherlands
| | - Chhoun Chamnan
- Department of Fisheries Post-Harvest Technologies and Quality Control, Ministry of Agriculture, Forestry and Fisheries, Phnom Penh, Cambodia
| | - Frank T Wieringa
- Qualisud, Univ Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de La Réunion, Montpellier, France
| | - Christèle Humblot
- Qualisud, Univ Montpellier, Avignon Université, CIRAD, Institut Agro, IRD, Université de La Réunion, Montpellier, France.
| |
Collapse
|
11
|
Bermúdez-Humarán LG, Chassaing B, Langella P. Exploring the interaction and impact of probiotic and commensal bacteria on vitamins, minerals and short chain fatty acids metabolism. Microb Cell Fact 2024; 23:172. [PMID: 38867272 PMCID: PMC11167913 DOI: 10.1186/s12934-024-02449-3] [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: 02/16/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024] Open
Abstract
There is increasing evidence that probiotic and commensal bacteria play a role in substrate metabolism, energy harvesting and intestinal homeostasis, and may exert immunomodulatory activities on human health. In addition, recent research suggests that these microorganisms interact with vitamins and minerals, promoting intestinal and metabolic well-being while producing vital microbial metabolites such as short-chain fatty acids (SCFAs). In this regard, there is a flourishing field exploring the intricate dynamics between vitamins, minerals, SCFAs, and commensal/probiotic interactions. In this review, we summarize some of the major hypotheses beyond the mechanisms by which commensals/probiotics impact gut health and their additional effects on the absorption and metabolism of vitamins, minerals, and SCFAs. Our analysis includes comprehensive review of existing evidence from preclinical and clinical studies, with particular focus on the potential interaction between commensals/probiotics and micronutrients. Finally, we highlight knowledge gaps and outline directions for future research in this evolving field.
Collapse
Affiliation(s)
- Luis G Bermúdez-Humarán
- Laboratory of Commensals and Probiotics-Host Interactions, Université Paris-Saclay, INRAE, Micalis Institute, Jouy-en-Josas, AgroParisTech, 78350, France.
| | - Benoit Chassaing
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, Paris, France
- INSERM U1016, team Mucosal microbiota in chronic inflammatory diseases, CNRS UMR 8104, Université de Paris, Paris, France
| | - Philippe Langella
- Laboratory of Commensals and Probiotics-Host Interactions, Université Paris-Saclay, INRAE, Micalis Institute, Jouy-en-Josas, AgroParisTech, 78350, France.
| |
Collapse
|
12
|
Álvarez-Herms J, Odriozola A. Microbiome and physical activity. ADVANCES IN GENETICS 2024; 111:409-450. [PMID: 38908903 DOI: 10.1016/bs.adgen.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Regular physical activity promotes health benefits and contributes to develop the individual biological potential. Chronical physical activity performed at moderate and high-intensity is the intensity more favorable to produce health development in athletes and improve the gut microbiota balance. The athletic microbiome is characterized by increased microbial diversity and abundance as well as greater phenotypic versatility. In addition, physical activity and microbiota composition have bidirectional effects, with regular physical activity improving microbial composition and microbial composition enhancing physical performance. The improvement of physical performance by a healthy microbiota is related to different phenotypes: i) efficient metabolic development, ii) improved regulation of intestinal permeability, iii) favourable modulation of local and systemic inflammatory and efficient immune responses, iv) efective regulation of systemic pH and, v) protection against acute stressful events such as environmental exposure to altitude or heat. The type of sport, both intensity or volume characteristics promote microbiota specialisation. Individual assessment of the state of the gut microbiota can be an effective biomarker for monitoring health in the medium to long term. The relationship between the microbiota and the rest of the body is bidirectional and symbiotic, with a full connection between the systemic functions of the nervous, musculoskeletal, endocrine, metabolic, acid-base and immune systems. In addition, circadian rhythms, including regular physical activity, directly influence the adaptive response of the microbiota. In conclusion, regular stimuli of moderate- and high-intensity physical activity promote greater diversity, abundance, resilience and versatility of the gut microbiota. This effect is highly beneficial for human health when healthy lifestyle habits including nutrition, hydration, rest, chronoregulation and physical activity.
Collapse
Affiliation(s)
- Jesús Álvarez-Herms
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain; Phymo® Lab, Physiology and Molecular Laboratory, Collado Hermoso, Segovia, Spain.
| | - Adrián Odriozola
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
13
|
EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA), Turck D, Bohn T, Castenmiller J, de Henauw S, Hirsch‐Ernst K, Knutsen HK, Maciuk A, Mangelsdorf I, McArdle HJ, Pentieva K, Siani A, Thies F, Tsabouri S, Vinceti M, Aggett P, Fairweather‐Tait S, de Sesmaisons Lecarré A, Fabiani L, Karavasiloglou N, Saad RM, Sofroniou A, Titz A, Naska A. Scientific opinion on the tolerable upper intake level for iron. EFSA J 2024; 22:e8819. [PMID: 38868106 PMCID: PMC11167337 DOI: 10.2903/j.efsa.2024.8819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
Following a request from the European Commission, the EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA) was asked to deliver a scientific opinion on the tolerable upper intake level (UL) for iron. Systematic reviews were conducted to identify evidence regarding high iron intakes and risk of chronic diseases, adverse gastrointestinal effects and adverse effects of iron supplementation in infancy, young childhood and pregnancy. It is established that systemic iron overload leads to organ toxicity, but no UL could be established. The only indicator for which a dose-response could be established was black stools, which reflect the presence of large amounts of unabsorbed iron in the gut. This is a conservative endpoint among the chain of events that may lead to systemic iron overload but is not adverse per se. Based on interventions in which black stools did not occur at supplemental iron intakes of 20-25 mg/day (added to a background intake of 15 mg/day), a safe level of intake for iron of 40 mg/day for adults (including pregnant and lactating women) was established. Using allometric scaling (body weight0.75), this value was scaled down to children and adolescents and safe levels of intakes between 10 mg/day (1-3 years) and 35 mg/day (15-17 years) were derived. For infants 7-11 months of age who have a higher iron requirement than young children, allometric scaling was applied to the supplemental iron intakes (i.e. 25 mg/day) and resulted in a safe level of supplemental iron intake of 5 mg/day. This value was extended to 4-6 month-old infants and refers to iron intakes from fortified foods and food supplements, not from infant and follow-on formulae. The application of the safe level of intake is more limited than a UL because the intake level at which the risk of adverse effects starts to increase is not defined.
Collapse
|
14
|
Yan Y, Zhang W, Wang Y, Yi C, Yu B, Pang X, Li K, Li H, Dai Y. Crosstalk between intestinal flora and human iron metabolism: the role in metabolic syndrome-related comorbidities and its potential clinical application. Microbiol Res 2024; 282:127667. [PMID: 38442456 DOI: 10.1016/j.micres.2024.127667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/31/2024] [Accepted: 02/25/2024] [Indexed: 03/07/2024]
Abstract
The interaction of iron and intestinal flora, both of which play crucial roles in many physiologic processes, is involved in the development of Metabolic syndrome (MetS). MetS is a pathologic condition represented by insulin resistance, obesity, dyslipidemia, and hypertension. MetS-related comorbidities including type 2 diabetes mellitus (T2DM), obesity, metabolism-related fatty liver (MAFLD), hypertension polycystic ovary syndrome (PCOS), and so forth. In this review, we examine the interplay between intestinal flora and human iron metabolism and its underlying mechanism in the pathogenesis of MetS-related comorbidities. The composition and metabolites of intestinal flora regulate the level of human iron by modulating intestinal iron absorption, the factors associated with iron metabolism. On the other hand, the iron level also affects the abundance, composition, and metabolism of intestinal flora. The crosstalk between these factors is of significant importance in human metabolism and exerts varying degrees of influence on the manifestation and progression of MetS-related comorbidities. The findings derived from these studies can enhance our comprehension of the interplay between intestinal flora and iron metabolism, and open up novel potential therapeutic approaches toward MetS-related comorbidities.
Collapse
Affiliation(s)
- Yijing Yan
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Wenlan Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yulin Wang
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chunmei Yi
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Bin Yu
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaoli Pang
- School of Nursing, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Kunyang Li
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - HuHu Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yongna Dai
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
15
|
Kaur T, Upadhyay J, Nandave M, Alsayari A, Alshehri SA, Pukale S, Wahab S, Ahmad W, Rashid S, Ansari MN. Exploring progress in iron supplement formulation approaches for treating iron deficiency anemia through bibliometric and thematic analysis. Heliyon 2024; 10:e29058. [PMID: 38623202 PMCID: PMC11016621 DOI: 10.1016/j.heliyon.2024.e29058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/04/2024] [Accepted: 03/28/2024] [Indexed: 04/17/2024] Open
Abstract
Anemia is a severe health issue that affects around one-third of the global population. Therefore, the present study aims to conduct a bibliometric analysis to investigate the research trends regarding advancements on iron formulations in treating iron deficiency anemia via oral or parenteral route. This study adopts thematic and bibliometric methods on existing research on novel iron formulations. It also provides perspective into the existing understanding on treatment strategies for iron deficiency anemia. This study is conducted on 543 papers on various ferrous and ferric formulations used in the treatment of iron deficiency anemia. The study period is from 1977 to 2022, and the papers are identified from the Scopus database. The bibliometric analysis was carried out using the R tool's Bibliometrix package. The study discusses performance analysis, including annual publications, geographic analysis, relevant affiliations, journal analysis, and citation analysis. In addition, the conceptual structure, including the co-occurrence network, thematic map, thematic evolution, intellectual structure highlighting co-citation analysis, and social structure depicting the collaboration network and collaboration world map, are presented. The results showed increased research on formulation strategies for the treatment of iron deficiency anemia from 2010 onwards. The top 5 contributing countries are the USA, Italy, India, Germany, and the UK, and peer-reviewed journals from the area of nutrition. The most trending areas of study are iron deficiency anemia in pregnancy, chronic kidney diseases, inflammatory bowel diseases, and various intravenous formulations used in its treatment. The authors from Europe collaborate the most with authors from other countries. The study concludes that a safer and more effective iron formulation is needed to reduce the prevalence of anemia. The findings of the study are helpful in advancing research on innovative formulations for treating iron deficiency anemia. The insights from the study are helpful to policymakers in designing specific health policies and investing more in research and development of novel formulations for the treatment of iron deficiency anemia.
Collapse
Affiliation(s)
- Tarnjot Kaur
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Bidholi Campus, Dehradun 248007, Uttarakhand, India
| | - Jyoti Upadhyay
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Bidholi Campus, Dehradun 248007, Uttarakhand, India
| | - Mukesh Nandave
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University New Delhi, 110017, India
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Saad Ali Alshehri
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Sudeep Pukale
- Lupin Research Park, Nande, Maharashtra 412115, India
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Wasim Ahmad
- Department of Pharmacy, Mohammed Al-Mana College for Medical Sciences, Dammam 34222, Saudi Arabia
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Mohd Nazam Ansari
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| |
Collapse
|
16
|
Choque-Quispe BM, Vásquez-Velásquez C, Gonzales GF. Evaluation of dietary composition between hemoglobin categories, total body iron content and adherence to multi-micronutrients in preschooler residents of the highlands of Puno, Peru. BMC Nutr 2024; 10:28. [PMID: 38347656 PMCID: PMC10860272 DOI: 10.1186/s40795-024-00837-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/05/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND The anemia prevalence is higher in highlands populations. It is assumed that iron deficiency anemia (IDA) in children is mainly due to low dietary intake. However, other suggest that high prevalence of anemia is due to an inappropriate hemoglobin (Hb) adjustment for altitude. MATERIALS AND METHODS Cross-sectional study conducted in 338 preschoolers (PSC) from Puno-Peru. Hb was measured in whole blood, and ferritin, Soluble transferrin receptor, and Interleukin 6 in serum.The dietary iron intake was assessed by 24-h dietary recall, using NutriCap Software. Hb concentration was assessed as adjusted or unadjusted for altitude. RESULTS With unadjusted Hb, the anemia prevalence was 4.7%, whereas after Hb correction, the prevalence raised-up to 65.6% (p < 0.001). Reciprocally, erythrocytosis proportion decreased from 20.35 to 0.30% (p < 0.001). Total Body Iron (TBI) showed that 7.44% had ID and 0.32% had IDA. PSC with normal unadjusted Hb levels have more protein and micronutrients intake than anemic ones. PSC with erythrocytosis consumed less fat, and more niacin and ascorbic acid than anemics. Total iron intake was lower in anemic than the other groups, but without statistical significance due to the standard deviation of the data in a small number of anemic PSC (n = 16). TBI, unadjusted Hb, and adjusted Hb were not different between groups consuming or not multimicronutrients. CONCLUSIONS The consumption of iron and iron status in children who live at high altitude is adequate, and that anemia could be due to other micronutrient deficiencies and/or that the adjustment of Hb by altitude is inappropriate.
Collapse
Affiliation(s)
| | - Cinthya Vásquez-Velásquez
- Laboratorio de Endocrinología y Reproducción (Laboratorios de Investigación y Desarrollo), Departamento de Ciencias Biológicas y Fisiológicas, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru.
- Instituto de Investigaciones de la Altura, Universidad Peruana Cayetano Heredia, Lima, Peru.
| | - Gustavo F Gonzales
- Laboratorio de Endocrinología y Reproducción (Laboratorios de Investigación y Desarrollo), Departamento de Ciencias Biológicas y Fisiológicas, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
- Instituto de Investigaciones de la Altura, Universidad Peruana Cayetano Heredia, Lima, Peru
| |
Collapse
|
17
|
Karamantziani T, Pouliakis A, Xanthos T, Ekmektzoglou K, Paliatsiou S, Sokou R, Iacovidou N. The Effect of Oral Iron Supplementation/Fortification on the Gut Microbiota in Infancy: A Systematic Review and Meta-Analysis. CHILDREN (BASEL, SWITZERLAND) 2024; 11:231. [PMID: 38397343 PMCID: PMC10887499 DOI: 10.3390/children11020231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024]
Abstract
(1) Background: Iron is an essential metal for the proper growth and neurodevelopment of infants. To prevent and treat iron deficiency, iron supplementation or fortification is often required. It has been shown, though, that it affects the synthesis of gut microbiota. (2) Methods: This paper is a systematic review and meta-analysis of the effect of oral iron supplementation/fortification on the gut microbiota in infancy. Studies in healthy neonates and infants who received per os iron with existing data on gut microbiota were included. Three databases were searched: PUBMED, Scopus, and Google Scholar. Randomized controlled trials (RCTs) were included. Quality appraisal was assessed using the ROB2Tool. (3) Results: A total of six RCTs met inclusion criteria for a systematic review, and four of them were included in the meta-analysis using both the fixed and random effects methods. Our results showed that there is very good heterogeneity in the iron group (I2 = 62%), and excellent heterogeneity in the non-iron group (I2 = 98%). According to the meta-analysis outcomes, there is a 10.3% (95% CI: -15.0--5.55%) reduction in the bifidobacteria population in the iron group and a -2.96% reduction for the non-iron group. There is a confirmed difference (p = 0.02) in the aggregated outcomes between iron and non-iron supplement, indicative that the bifidobacteria population is reduced when iron supplementation is given (total reduction 6.37%, 95%CI: 10.16-25.8%). (4) Conclusions: The abundance of bifidobacteria decreases when iron supplementation or fortification is given to infants.
Collapse
Affiliation(s)
- Theoni Karamantziani
- B’ Neonatal Intensive Care Unit and Neonatal High Dependency Unit, “Aghia Sofia” General Children’s Hospital, 11527 Athens, Greece;
| | - Abraham Pouliakis
- 2nd Department of Pathology, “Attikon” University Hospital, National and Kapodistrian University of Athens, 12464 Athens, Greece
| | - Theodoros Xanthos
- School of Health Sciences, University of West Attica, 12243 Athens, Greece;
| | | | - Styliani Paliatsiou
- 2nd Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens, Aretaieio Hospital, 11528 Athens, Greece;
| | - Rozeta Sokou
- Neonatal Intensive Care Unit, “Agios Panteleimon” General Hospital of Nikea, 3 D. Mantouvalou Str., Nikea, 18454 Piraeus, Greece;
| | - Nicoletta Iacovidou
- Neonatal Department, Aretaieio Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| |
Collapse
|
18
|
Sun B, Tan B, Zhang P, Zhu L, Wei H, Huang T, Li C, Yang W. Iron deficiency anemia: a critical review on iron absorption, supplementation and its influence on gut microbiota. Food Funct 2024; 15:1144-1157. [PMID: 38235788 DOI: 10.1039/d3fo04644c] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Iron deficiency anemia (IDA) caused by micronutrient iron deficiency has attracted global attention due to its adverse health effects. The regulation of iron uptake and metabolism is finely controlled by various transporters and hormones in the body. Dietary iron intake and regulation are essential in maintaining human health and iron requirements. The review aims to investigate literature concerning dietary iron intake and systemic regulation. Besides, recent IDA treatment and dietary iron supplementation are discussed. Considering the importance of the gut microbiome, the interaction between bacteria and micronutrient iron in the gut is also a focus of this review. The iron absorption efficiency varies considerably according to iron type and dietary factors. Iron fortification remains the cost-effective strategy, although challenges exist in developing suitable iron fortificants and food vehicles regarding bioavailability and acceptability. Iron deficiency may alter the microbiome structure and promote the growth of pathogenic bacteria in the gut, affecting immune balance and human health.
Collapse
Affiliation(s)
- Bolun Sun
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
- School of Nursing, Wenzhou Medical University, Wenzhou 325035, China
| | - Beibei Tan
- School of Agriculture and Food, Faculty of Science, University of Melbourne, Australia
| | - Panxue Zhang
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| | - Lianlian Zhu
- School of Nursing, Wenzhou Medical University, Wenzhou 325035, China
| | - Huamao Wei
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| | - Tao Huang
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| | - Chao Li
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| | - Wenge Yang
- College of Food and Pharmaceutical Sciences, Key Laboratory of Animal Protein Food Deep Processing Technology of Zhejiang Province, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
19
|
Mikulic N, Uyoga MA, Stoffel NU, Derrien M, Nyilima S, Kostopoulos I, Roeselers G, Chenoll E, Mwasi E, Pironaci G, Karanja S, Bourdet-Sicard R, Zimmermann MB. Prebiotics increase iron absorption and reduce the adverse effects of iron on the gut microbiome and inflammation: a randomized controlled trial using iron stable isotopes in Kenyan infants. Am J Clin Nutr 2024; 119:456-469. [PMID: 38042412 PMCID: PMC10884607 DOI: 10.1016/j.ajcnut.2023.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 11/18/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023] Open
Abstract
BACKGROUND Iron fortificants tend to be poorly absorbed and may adversely affect the gut, especially in African children. OBJECTIVE We assessed the effects of prebiotic galacto-oligosaccharides/fructo-oligosaccharides (GOS/FOS) on iron absorption and gut health when added to iron-fortified infant cereal. METHODS We randomly assigned Kenyan infants (n = 191) to receive daily for 3 wk a cereal containing iron and 7.5 g GOS/FOS (7.5 g+iron group), 3 g (3-g+iron group) GOS/FOS, or no prebiotics (iron group). A subset of infants in the 2 prebiotic+iron groups (n = 66) consumed 4 stable iron isotope-labeled test meals without and with prebiotics, both before and after the intervention. Primary outcome was fractional iron absorption (FIA) from the cereal with or without prebiotics regardless of dose, before and after 3 wk of consumption. Secondary outcomes included fecal gut microbiota, iron and inflammation status, and effects of prebiotic dose. RESULTS Median (25th-75th percentiles) FIAs from meals before intervention were as follows: 16.3% (8.0%-27.6%) without prebiotics compared with 20.5% (10.4%-33.4%) with prebiotics (Cohen d = 0.53; P < 0.001). FIA from the meal consumed without prebiotics after intervention was 22.9% (8.5%-32.4%), 41% higher than from the meal without prebiotics before intervention (Cohen d = 0.36; P = 0.002). FIA from the meal consumed with prebiotics after intervention was 26.0% (12.2%-36.1%), 60% higher than from the meal without prebiotics before intervention (Cohen d = 0.45; P = 0.007). After 3 wk, compared with the iron group, the following results were observed: 1) Lactobacillus sp. abundances were higher in both prebiotic+iron groups (P < 0.05); 2) Enterobacteriaceae sp. abundances (P = 0.022) and the sum of pathogens (P < 0.001) were lower in the 7.5-g+iron group; 3) the abundance of bacterial toxin-encoding genes was lower in the 3-g+iron group (false discovery rate < 0.05); 4) fecal pH (P < 0.001) and calprotectin (P = 0.033) were lower in the 7.5-g+iron group. CONCLUSIONS Adding prebiotics to iron-fortified infant cereal increases iron absorption and reduces the adverse effects of iron on the gut microbiome and inflammation in Kenyan infants. This trial was registered at clinicaltrials.gov as NCT03894358.
Collapse
Affiliation(s)
- Nadja Mikulic
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Mary A Uyoga
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Nicole U Stoffel
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | | | - Suzane Nyilima
- Public and Community Health Department, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | | | | | | | - Edith Mwasi
- Paediatrics Department, Msambweni County Referral Hospital, Msambweni, Kenya
| | - Giulia Pironaci
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Simon Karanja
- Public and Community Health Department, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | | | - Michael B Zimmermann
- Medical Research Council Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford, United Kingdom.
| |
Collapse
|
20
|
Adnan NA, Breen E, Tan CA, Wang CC, Jalaludin MY, Lum LCS. Iron deficiency in healthy, term infants aged five months, in a pediatric outpatient clinic: a prospective study. BMC Pediatr 2024; 24:74. [PMID: 38263022 PMCID: PMC10804717 DOI: 10.1186/s12887-023-04277-7] [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: 05/23/2023] [Accepted: 08/28/2023] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Iron deficiency (ID) is prevalent in Malaysian children. The incidence of ID in infants under 6 months of age is unknown. Our aim was to determine the prevalence of iron deficiency (ID) and iron deficiency anemia (IDA) in healthy, term infants aged below 6 months in our hospital population. METHODS A prospective longitudinal pilot study of mother-infant pairs was conducted on infants receiving routine immunizations in a mother and child clinic at a university hospital, in Kuala Lumpur, Malaysia. Mothers completed standardized questionnaires at 3- and 5-month postnatal visits. Maternal and infant full blood count, ferritin, and C-reactive protein (CRP) levels were measured at 3 months and for the infants repeated at 5 months. Infant anthropometric measurements were obtained at both visits. We conducted a univariate analysis to identify factors associated with ID and IDA. RESULTS Altogether, 91 mother-infant pairs were enrolled, with 88 completing the study. No infant had ID or IDA at 3 months; the lowest ferritin level was 16.6 µg/L. At 5 months, 5.9% (5/85) of infants had ID, and 2.4% (2/85) had IDA. Median (interquartile range) infant ferritin levels significantly declined from 113.4 (65.0-183.6) µg/L at 3 months to 50.9 (29.2-70.4) µg/L at 5 months, p < 0.001. Exclusive breastfeeding until 3 or 5 months was significantly associated with ID at 5 months (p = 0.020, and p = 0.008, respectively) on univariate analysis. The drop in ferritin between 3-5 months was significantly associated with weight and length gains between 0-3 months (p = 0.018, p = 0.009, respectively). Altogether, 14.3% of infants exclusively breastfed until 5 months developed ID. At 5 months, 3.4% of infants were underweight, 1.1% stunted, and 10.2% wasted. CONCLUSIONS In exclusively breastfed term infants, ID occurred by 5 months. Early introduction of iron-rich foods should be considered in exclusively breastfed babies. A high prevalence of wasting suggests a calorie deficit in this population and will lead to stunting if not addressed.
Collapse
Affiliation(s)
- Nur Aida Adnan
- Pediatric Department, Hospital Tunku Azizah, Kuala Lumpur, Malaysia
- Department of Pediatrics, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Emer Breen
- Clinical Investigation Center, University of Malaya Medical Center, 5th Floor East Tower, Kuala Lumpur, Malaysia.
| | - Chin Aun Tan
- Occupational Safety and Health Unit, Hospital Tunku Azizah, Kuala Lumpur, Malaysia
| | - Crystal C Wang
- Occupational Safety and Health Unit, Hospital Tunku Azizah, Kuala Lumpur, Malaysia
- Weill Cornell Medicine, New York, NY, USA
| | | | - Lucy Chai See Lum
- Department of Pediatrics, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
21
|
Hamamah S, Iatcu OC, Covasa M. Nutrition at the Intersection between Gut Microbiota Eubiosis and Effective Management of Type 2 Diabetes. Nutrients 2024; 16:269. [PMID: 38257161 PMCID: PMC10820857 DOI: 10.3390/nu16020269] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/15/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
Nutrition is one of the most influential environmental factors in both taxonomical shifts in gut microbiota as well as in the development of type 2 diabetes mellitus (T2DM). Emerging evidence has shown that the effects of nutrition on both these parameters is not mutually exclusive and that changes in gut microbiota and related metabolites such as short-chain fatty acids (SCFAs) and branched-chain amino acids (BCAAs) may influence systemic inflammation and signaling pathways that contribute to pathophysiological processes associated with T2DM. With this background, our review highlights the effects of macronutrients, carbohydrates, proteins, and lipids, as well as micronutrients, vitamins, and minerals, on T2DM, specifically through their alterations in gut microbiota and the metabolites they produce. Additionally, we describe the influences of common food groups, which incorporate varying combinations of these macronutrients and micronutrients, on both microbiota and metabolic parameters in the context of diabetes mellitus. Overall, nutrition is one of the first line modifiable therapies in the management of T2DM and a better understanding of the mechanisms by which gut microbiota influence its pathophysiology provides opportunities for optimizing dietary interventions.
Collapse
Affiliation(s)
- Sevag Hamamah
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
| | - Oana C. Iatcu
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania
| | - Mihai Covasa
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania
| |
Collapse
|
22
|
Momo Cabrera P, Rachmühl C, Derrien M, Bourdet-Sicard R, Lacroix C, Geirnaert A. Comparative prebiotic potential of galacto- and fructo-oligosaccharides, native inulin, and acacia gum in Kenyan infant gut microbiota during iron supplementation. ISME COMMUNICATIONS 2024; 4:ycae033. [PMID: 38774131 PMCID: PMC11107946 DOI: 10.1093/ismeco/ycae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 03/10/2024] [Indexed: 05/24/2024]
Abstract
Iron fortification to prevent anemia in African infants increases colonic iron levels, favoring the growth of enteropathogens. The use of prebiotics may be an effective strategy to reduce these detrimental effects. Using the African infant PolyFermS gut model, we compared the effect of the prebiotics short-chain galacto- with long-chain fructo-oligosaccharides (scGOS/lcFOS) and native inulin, and the emerging prebiotic acacia gum, a branched-polysaccharide-protein complex consisting of arabinose and galactose, during iron supplementation on four Kenyan infant gut microbiota. Iron supplementation did not alter the microbiota but promoted Clostridioides difficile in one microbiota. The prebiotic effect of scGOS/lcFOS and inulin was confirmed during iron supplementation in all investigated Kenyan infant gut microbiota, leading to higher abundance of bifidobacteria, increased production of acetate, propionate, and butyrate, and a significant shift in microbiota composition compared to non-supplemented microbiota. The abundance of the pathogens Clostridium difficile and Clostridium perfringens was also inhibited upon addition of the prebiotic fibers. Acacia gum had no effect on any of the microbiota. In conclusion, scGOS/lcFOS and inulin, but not acacia gum, showed a donor-independent strong prebiotic potential in Kenyan infant gut microbiota. This study demonstrates the relevance of comparing fibers in vitro prior to clinical studies.
Collapse
Affiliation(s)
- Paula Momo Cabrera
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zurich, Switzerland
| | - Carole Rachmühl
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zurich, Switzerland
| | - Muriel Derrien
- Danone Global Research & Innovation Center, 91190 Gif sur Yvette, France
- Present address: Department of Microbiology and Immunology, Laboratory of Molecular Bacteriology, Rega Institute KU, 3000 Leuven, Belgium
| | | | - Christophe Lacroix
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zurich, Switzerland
| | - Annelies Geirnaert
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
23
|
Nolte S, Krüger K, Lenz C, Zentgraf K. Optimizing the Gut Microbiota for Individualized Performance Development in Elite Athletes. BIOLOGY 2023; 12:1491. [PMID: 38132317 PMCID: PMC10740793 DOI: 10.3390/biology12121491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/23/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023]
Abstract
The human gut microbiota can be compared to a fingerprint due to its uniqueness, hosting trillions of living organisms. Taking a sport-centric perspective, the gut microbiota might represent a physiological system that relates to health aspects as well as individualized performance in athletes. The athletes' physiology has adapted to their exceptional lifestyle over the years, including the diversity and taxonomy of the microbiota. The gut microbiota is influenced by several physiological parameters and requires a highly individual and complex approach to unravel the linkage between performance and the microbial community. This approach has been taken in this review, highlighting the functions that the microbial community performs in sports, naming gut-centered targets, and aiming for both a healthy and sustainable athlete and performance development. With this article, we try to consider whether initiating a microbiota analysis is practicable and could add value in elite sport, and what possibilities it holds when influenced through a variety of interventions. The aim is to support enabling a well-rounded and sustainable athlete and establish a new methodology in elite sport.
Collapse
Affiliation(s)
- Svenja Nolte
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, University of Giessen, 35394 Giessen, Germany; (K.K.); (C.L.)
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, University of Giessen, 35394 Giessen, Germany; (K.K.); (C.L.)
| | - Claudia Lenz
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, University of Giessen, 35394 Giessen, Germany; (K.K.); (C.L.)
| | - Karen Zentgraf
- Department 5: Psychology & Sports Sciences, Institute for Sports Sciences, Goethe University Frankfurt, 60323 Frankfurt am Main, Germany;
| |
Collapse
|
24
|
Brittenham GM, Moir-Meyer G, Abuga KM, Datta-Mitra A, Cerami C, Green R, Pasricha SR, Atkinson SH. Biology of Anemia: A Public Health Perspective. J Nutr 2023; 153 Suppl 1:S7-S28. [PMID: 37778889 DOI: 10.1016/j.tjnut.2023.07.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/04/2023] [Accepted: 07/31/2023] [Indexed: 10/03/2023] Open
Abstract
Our goal is to present recent progress in understanding the biological mechanisms underlying anemia from a public health perspective. We describe important advances in understanding common causes of anemia and their interactions, including iron deficiency (ID), lack of other micronutrients, infection, inflammation, and genetic conditions. ID develops if the iron circulating in the blood cannot provide the amounts required for red blood cell production and tissue needs. ID anemia develops as iron-limited red blood cell production fails to maintain the hemoglobin concentration above the threshold used to define anemia. Globally, absolute ID (absent or reduced body iron stores that do not meet the need for iron of an individual but may respond to iron supplementation) contributes to only a limited proportion of anemia. Functional ID (adequate or increased iron stores that cannot meet the need for iron because of the effects of infection or inflammation and does not respond to iron supplementation) is frequently responsible for anemia in low- and middle-income countries. Absolute and functional ID may coexist. We highlight continued improvement in understanding the roles of infections and inflammation in causing a large proportion of anemia. Deficiencies of nutrients other than iron are less common but important in some settings. The importance of genetic conditions as causes of anemia depends upon the specific inherited red blood cell abnormalities and their prevalence in the settings examined. From a public health perspective, each setting has a distinctive composition of components underlying the common causes of anemia. We emphasize the coincidence between regions with a high prevalence of anemia attributed to ID (both absolute and functional), those with endemic infections, and those with widespread genetic conditions affecting red blood cells, especially in sub-Saharan Africa and regions in Asia and Oceania.
Collapse
Affiliation(s)
- Gary M Brittenham
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY, United States.
| | - Gemma Moir-Meyer
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Kelvin Mokaya Abuga
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Ananya Datta-Mitra
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, United States
| | - Carla Cerami
- The Medical Research Council Unit, The Gambia, London School of Hygiene and Tropical Medicine, London, UK
| | - Ralph Green
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, United States
| | - Sant-Rayn Pasricha
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia; Diagnostic Haematology, The Royal Melbourne Hospital; and Clinical Haematology at the Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Parkville, VIC Australia
| | - Sarah H Atkinson
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme, Kilifi, Kenya; Department of Paediatrics, University of Oxford, Oxford, UK
| |
Collapse
|
25
|
Loechl CU, Datta-Mitra A, Fenlason L, Green R, Hackl L, Itzkowitz L, Koso-Thomas M, Moorthy D, Owino VO, Pachón H, Stoffel N, Zimmerman MB, Raiten DJ. Approaches to Address the Anemia Challenge. J Nutr 2023; 153 Suppl 1:S42-S59. [PMID: 37714779 PMCID: PMC10797550 DOI: 10.1016/j.tjnut.2023.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 07/21/2023] [Accepted: 07/31/2023] [Indexed: 09/17/2023] Open
Abstract
Anemia is a multifactorial condition; approaches to address it must recognize that the causal factors represent an ecology consisting of internal (biology, genetics, and health) and external (social/behavioral/demographic and physical) environments. In this paper, we present an approach for selecting interventions, followed by a description of key issues related to the multiple available interventions for prevention and reduction of anemia. We address interventions for anemia using the following 2 main categories: 1) those that address nutrients alone, and, 2) those that address nonnutritional causes of anemia. The emphasis will be on interventions of public health relevance, but we also consider the clinical context. We also focus on interventions at different stages of the life course, with a particular focus on women of reproductive age and preschool-age children, and present evidence on various factors to consider when selecting an intervention-inflammation, genetic mutations, nutrient delivery, bioavailability, and safety. Each section on an intervention domain concludes with a brief discussion of key research areas.
Collapse
Affiliation(s)
- Cornelia U Loechl
- Division of Human Health, International Atomic Energy Agency, Vienna, Austria
| | - Ananya Datta-Mitra
- Department of Pathology and Laboratory Medicine, University of California, Davis, Davis, CA, United States
| | - Lindy Fenlason
- Bureau for Global Health, USAID, Washington, DC, United States
| | - Ralph Green
- Department of Pathology and Laboratory Medicine, University of California, Davis, Davis, CA, United States
| | - Laura Hackl
- USAID Advancing Nutrition, John Snow Inc., Arlington, VA, United States
| | - Laura Itzkowitz
- Bureau for Global Health, USAID, Washington, DC, United States
| | - Marion Koso-Thomas
- Eunice Kennedy Shriver National Institute of Child Health and Development, National Institutes of Health, Bethesda, MD, Unites States
| | - Denish Moorthy
- USAID Advancing Nutrition, John Snow Inc., Arlington, VA, United States.
| | | | - Helena Pachón
- Food Fortification Initiative, Emory University, Atlanta, GA, United States
| | - Nicole Stoffel
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Zu¨rich, Switzerland; MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Michael B Zimmerman
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Daniel J Raiten
- Eunice Kennedy Shriver National Institute of Child Health and Development, National Institutes of Health, Bethesda, MD, Unites States
| |
Collapse
|
26
|
Golbeck L, Cohrs I, Humann-Ziehank E, Grünberg W. Effects of oral or parenteral iron supplementation on haematological parameters, blood iron status and growth in newborn calves fed milk replacer. Vet Rec 2023; 193:e3265. [PMID: 37518851 DOI: 10.1002/vetr.3265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/28/2023] [Accepted: 07/05/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND This study aimed to assess the influence of early life iron supplementation on the haematological parameters of calves fed milk replacer on a high plane of nutrition. METHODS Thirty calves were allocated to receive either a sham treatment (CON), injection of 1000 mg Fe3+ (INJ) or oral administration of 1050 mg Fe3+ (ORAL), all administered less than 1 hour after birth. Blood was obtained before treatment, on days 1, 3, 5 and 7 of life and once weekly until week 9. Samples were analysed for haemoglobin (Hb), packed cell volume (PCV) and estimated transferrin saturation (%TSAT). RESULTS The Hb and PCV of calves in the INJ and ORAL groups were above the values of those in the CON group throughout the study. Hb and PCV of the CON group remained within the reference range for calves. The %TSAT exceeded reported ranges in the ORAL group in the first week. Values for the CON group were below the INJ and ORAL groups throughout the entire study. Average daily gain did not differ between treatments. LIMITATIONS The sample size was insufficient to assess the influence of iron supplementation on disease development. Furthermore, the study was set in a controlled environment and not performed under field conditions. CONCLUSIONS Oral and subcutaneous iron administration had similar effects on haematological development but resulted in numerically different transferrin saturation. Control animals showed lower Hb and PCV but did not develop overt anaemia.
Collapse
Affiliation(s)
- Lennart Golbeck
- Clinic for Cattle, University of Veterinary Medicine Hannover, Hanover, Germany
- Department of Internal Medicine, Reproduction and Population Medicine, Faculty of Veterinary Medicine, Merelbeke, Belgium
| | - Imke Cohrs
- Educational and Research Center for Animal Husbandry, Hofgut Neumühle, Münchweiler and der Alsenz, Germany
- Clinic for Ruminants, Justus-Liebig-University Giessen, Giessen, Germany
| | | | - Walter Grünberg
- Clinic for Cattle, University of Veterinary Medicine Hannover, Hanover, Germany
- Clinic for Ruminants, Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
27
|
von Siebenthal HK, Gessler S, Vallelian F, Steinwendner J, Kuenzi UM, Moretti D, Zimmermann MB, Stoffel NU. Alternate day versus consecutive day oral iron supplementation in iron-depleted women: a randomized double-blind placebo-controlled study. EClinicalMedicine 2023; 65:102286. [PMID: 38021373 PMCID: PMC10659995 DOI: 10.1016/j.eclinm.2023.102286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 12/01/2023] Open
Abstract
Background Guidelines to treat iron deficiency recommend daily provision of oral iron, but this may decrease fractional iron absorption and increase side effects. Our objective was to compare consecutive-day versus alternate-day iron supplementation. Methods In a double-masked, randomized, placebo-controlled trial, young Swiss women (n = 150; serum ferritin ≤30 μg/L) were assigned to: daily 100 mg iron for 90 d, followed by daily placebo for another 90 d (consecutive-day group) or the same daily dose of iron and placebo on alternate days for 180 d (alternate-day group). The study period was 24/11/2021-10/8/2022. Co-primary outcomes, at equal total iron doses, were serum ferritin and gastrointestinal side effects; secondary outcomes were iron deficiency and serum hepcidin. Compliance and side effects were recorded daily using a mobile application. Data were analysed using mixed models and longitudinal prevalence ratios (LPR). The trial was registered at ClinicalTrials.gov (NCT05105438). Findings 75 women were assigned to each group and included in the intention-to-treat analysis. Capsule adherence and side effect reporting was >97% in both groups. At equal total iron doses, comparing consecutive-day and alternate-day groups, median serum ferritin was 43.8 μg/L (31.7-58.2) versus 44.8 μg/L (33.8-53.6) (P = 0.98), the LPR for gastrointestinal side effects on days of iron intake was 1.56 (95% CI: 1.38, 1.77; P < 0.0001), and median serum hepcidin was 3.0 nM (IQR 2.0-5.0) versus 1.9 nM (1.4-2.9) (P < 0.0001). Iron deficiency prevalence after 3 months was 5.5% versus 4.3% (P = 0.74) and after 6 months was 11.4% and 3.0% (P = 0.049). Interpretation At equal total iron doses, compared to consecutive day dosing of iron, alternate day dosing did not result in higher serum ferritin but reduced iron deficiency at 6 months and triggered fewer gastrointestinal side effects. Funding Swiss National Science Foundation, Bern, Switzerland.
Collapse
Affiliation(s)
- Hanna K. von Siebenthal
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Sara Gessler
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
| | - Florence Vallelian
- Division of Internal Medicine, University Hospital and University of Zurich, Zurich, Switzerland
| | - Joachim Steinwendner
- Laboratory of Web Science, Swiss Distance University of Applied Sciences, Zürich, Switzerland
| | - Urs-Martin Kuenzi
- Laboratory of Web Science, Swiss Distance University of Applied Sciences, Zürich, Switzerland
| | - Diego Moretti
- Nutrition Research, Department of Health, Swiss Distance University of Applied Sciences, Zürich, Switzerland
| | - Michael B. Zimmermann
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
- Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Nicole U. Stoffel
- Laboratory of Human Nutrition, Department of Health Sciences and Technology, ETH Zurich, Switzerland
- Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
28
|
de Romaña DL, Mildon A, Golan J, Jefferds MED, Rogers LM, Arabi M. Review of intervention products for use in the prevention and control of anemia. Ann N Y Acad Sci 2023; 1529:42-60. [PMID: 37688369 PMCID: PMC10876383 DOI: 10.1111/nyas.15062] [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] [Indexed: 09/10/2023]
Abstract
Anemia remains a major public health problem, especially in low- and middle-income countries. The World Health Organization recommends several interventions to prevent and manage anemia in vulnerable population groups, including young children, menstruating adolescent girls and women, and pregnant and postpartum women. Daily iron supplementation reduces the risk of anemia in infants, children, and pregnant women, and intermittent iron supplementation reduces anemia risk in menstruating girls and women. Micronutrient powders reduce the risk of anemia in children. Fortifying wheat flour with iron reduces the risk of anemia in the overall population, whereas the effect of fortifying maize flour and rice is still uncertain. Regarding non-nutrition-related interventions, malaria treatment and deworming have been reported to decrease anemia prevalence. Promising interventions to prevent anemia include vitamin A supplementation, multiple micronutrient supplementation for pregnant women, small-quantity lipid-based supplements, and fortification of salt with iodine and iron. Future research could address the efficacy and safety of different iron supplementation formulations, identify the most bioavailable form of iron for fortification, examine adherence to supplementation regimens and fortification standards, and investigate the effectiveness of integrating micronutrient, helminth, and malaria control programs.
Collapse
Affiliation(s)
| | - Alison Mildon
- Global Technical Services, Nutrition International, Ottawa, Ontario, Canada
| | - Jenna Golan
- Global Technical Services, Nutrition International, Ottawa, Ontario, Canada
| | | | - Lisa M. Rogers
- Department of Nutrition and Food Safety, World Health Organization, Geneva, Switzerland
| | - Mandana Arabi
- Global Technical Services, Nutrition International, Ottawa, Ontario, Canada
| |
Collapse
|
29
|
Soriano-Lerma A, García-Burgos M, Alférez MJ, Crespo-Pérez JV, Pérez-Carrasco V, Ortiz-Gonzalez M, Linde-Rodriguez Á, Sanchez-Martin V, Soriano M, Garcia-Salcedo JA, López-Aliaga I. Fermented Goat's Milk Contributes to the Recovery of Iron Deficiency Anemia via Modulation of the Gut Microbiome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15668-15679. [PMID: 37830350 PMCID: PMC11006235 DOI: 10.1021/acs.jafc.3c05560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/19/2023] [Accepted: 09/29/2023] [Indexed: 10/14/2023]
Abstract
Iron deficiency anemia (IDA) is a global public health concern affecting 1.6 billion people worldwide. The administration of iron supplements during the treatment of IDA adversely affects the intestinal barrier function and the composition and functionality of the intestinal microbiome, both of which are already altered during IDA. For this reason, it is of great interest to develop nutritional strategies aimed at alleviating these gut alterations associated with IDA and its treatment. In this sense, fermented goat's milk (FGM) was studied due to its nutritional quality. Our findings showed that in anemic animals the consumption of a FGM-based diet, compared to a standard diet, had positive modulatory effects on the intestinal microbiome. FGM-based diet restored intestinal dysbiosis, the intestinal barrier functionality, and bacterial translocation, contributing to a more efficient recovery of IDA. Therefore, FGM is a useful nutritional tool to ease intestinal alterations occurring during IDA and during its treatment.
Collapse
Affiliation(s)
- Ana Soriano-Lerma
- Department
of Physiology (Faculty of Pharmacy, Campus Universitario de Cartuja),
Institute of Nutrition and Food Technology “José Mataix
Verdú”, University of Granada, E-18071 Granada, Spain
- GENYO,
Centre for Genomics and Oncological Research: Pfizer/University of
Granada/Andalusian Regional Government, PTS Granada, E-18016 Granada, Spain
- Instituto
de Investigación Biosanitaria ibs.GRANADA, E-18012 Granada, Spain
| | - María García-Burgos
- Department
of Physiology (Faculty of Pharmacy, Campus Universitario de Cartuja),
Institute of Nutrition and Food Technology “José Mataix
Verdú”, University of Granada, E-18071 Granada, Spain
- GENYO,
Centre for Genomics and Oncological Research: Pfizer/University of
Granada/Andalusian Regional Government, PTS Granada, E-18016 Granada, Spain
| | - María José
M. Alférez
- Department
of Physiology (Faculty of Pharmacy, Campus Universitario de Cartuja),
Institute of Nutrition and Food Technology “José Mataix
Verdú”, University of Granada, E-18071 Granada, Spain
| | - Jorge Valentín Crespo-Pérez
- Service
of Anatomical Pathology, Intercenter Regional Unit Granada, University Hospital Virgen de las Nieves, E-18014 Granada, Spain
| | - Virginia Pérez-Carrasco
- GENYO,
Centre for Genomics and Oncological Research: Pfizer/University of
Granada/Andalusian Regional Government, PTS Granada, E-18016 Granada, Spain
- Instituto
de Investigación Biosanitaria ibs.GRANADA, E-18012 Granada, Spain
- Microbiology
Unit, University Hospital Virgen de las
Nieves, E-18014 Granada, Spain
| | - Matilde Ortiz-Gonzalez
- GENYO,
Centre for Genomics and Oncological Research: Pfizer/University of
Granada/Andalusian Regional Government, PTS Granada, E-18016 Granada, Spain
- Center for
Intensive Mediterranean Agrosystems and Agri-Food Biotechnology (CIAIMBITAL), University of Almeria, E-04120 Almería, Spain
| | - Ángel Linde-Rodriguez
- GENYO,
Centre for Genomics and Oncological Research: Pfizer/University of
Granada/Andalusian Regional Government, PTS Granada, E-18016 Granada, Spain
- Instituto
de Investigación Biosanitaria ibs.GRANADA, E-18012 Granada, Spain
- Microbiology
Unit, University Hospital Virgen de las
Nieves, E-18014 Granada, Spain
| | - Victoria Sanchez-Martin
- GENYO,
Centre for Genomics and Oncological Research: Pfizer/University of
Granada/Andalusian Regional Government, PTS Granada, E-18016 Granada, Spain
- Instituto
de Investigación Biosanitaria ibs.GRANADA, E-18012 Granada, Spain
- Microbiology
Unit, University Hospital Virgen de las
Nieves, E-18014 Granada, Spain
| | - Miguel Soriano
- Center for
Intensive Mediterranean Agrosystems and Agri-Food Biotechnology (CIAIMBITAL), University of Almeria, E-04120 Almería, Spain
| | - Jose A. Garcia-Salcedo
- GENYO,
Centre for Genomics and Oncological Research: Pfizer/University of
Granada/Andalusian Regional Government, PTS Granada, E-18016 Granada, Spain
- Instituto
de Investigación Biosanitaria ibs.GRANADA, E-18012 Granada, Spain
- Microbiology
Unit, University Hospital Virgen de las
Nieves, E-18014 Granada, Spain
| | - Inmaculada López-Aliaga
- Department
of Physiology (Faculty of Pharmacy, Campus Universitario de Cartuja),
Institute of Nutrition and Food Technology “José Mataix
Verdú”, University of Granada, E-18071 Granada, Spain
| |
Collapse
|
30
|
SHI J, XIE Y, LI Y, REN D, ZHANG Y, SHAO H, LIU Y, WANG X, LI Y. Effects of food-grade iron(III) oxide nanoparticles on cecal digesta- and mucosa-associated microbiota and short-chain fatty acids in rats. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2023; 43:43-54. [PMID: 38188661 PMCID: PMC10767317 DOI: 10.12938/bmfh.2023-012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 08/21/2023] [Indexed: 01/09/2024]
Abstract
Although iron(III) oxide nanoparticles (IONPs) are widely used in diverse applications ranging from food to biomedicine, the effects of IONPs on different locations of gut microbiota and short-chain fatty acids (SCFAs) are unclear. So, a subacute repeated oral toxicity study on Sprague Dawley (SD) rats was performed, administering low (50 mg/kg·bw), medium (100 mg/kg·bw), and high (200 mg/kg·bw) doses of IONPs. In this study, we found that a high dose of IONPs increased animal weight, and 16S rRNA sequencing revealed that IONPs caused intestinal flora disorders in both the cecal digesta- and mucosa-associated microbiota. However, only high-dose IONP exposure changed the abundance and composition of the mucosa-associated microbiota. IONPs increased the relative abundances of Firmicutes, Ruminococcaceae_UCG-014, Ruminiclostridium_9, Romboutsia, and Bilophila and decreased the relative abundance of Bifidobacterium, and many of these microorganisms are associated with weight gain, obesity, inflammation, diabetes, and mucosal damage. Functional analysis showed that changes in the gut microbiota induced by a high dose of IONPs were mainly related to metabolism, infection, immune, and endocrine disease functions. IONPs significantly elevated the levels of valeric, isobutyric, and isovaleric acid, promoting the absorption of iron. This is the first description of intestinal microbiota dysbiosis in SD rats caused by IONPs, and the effects and mechanisms of action of IONPs on intestinal and host health need to be further studied and confirmed.
Collapse
Affiliation(s)
- Jiangchun SHI
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Yumeng XIE
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Yulin LI
- Department of Hospital-acquired Infection Management, Guizhou
Provincial People’s Hospital, Guiyang 550002, China
| | - Dongxia REN
- Department of Blood Transfusion, Tangdu Hospital, Fourth
Military Medical University, Xi’an 710032, China
| | - Yiqi ZHANG
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Huangfang SHAO
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Yang LIU
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Xue WANG
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
| | - Yun LI
- West China School of Public Health and West China Fourth
Hospital, Sichuan University, Chengdu 610041, China
- Provincial Key Laboratory of Food Safety Monitoring and Risk
Assessment of Sichuan, Chengdu 610041, China
| |
Collapse
|
31
|
Gnatzy L, Ismailos G, Vertzoni M, Reppas C. Managing the clinical effects of drug-induced intestinal dysbiosis with a focus to antibiotics: Challenges and opportunities. Eur J Pharm Sci 2023; 188:106510. [PMID: 37380062 DOI: 10.1016/j.ejps.2023.106510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 06/30/2023]
Abstract
The term "intestinal dysbiosis" is used for indicating change(s) of the intestinal microbiota which have been associated with the development of diseases and the deterioration of disease treatments in humans. In this review, documented clinical effects of drug-induced intestinal dysbiosis are briefly presented, and methodologies which could be considered for the management of drug-induced intestinal dysbiosis based on clinical data are critically reviewed. Until relevant methodologies are optimized and/or their effectiveness to the general population is confirmed, and, since drug-induced intestinal dysbiosis refers predominantly to antibiotic-specific intestinal dysbiosis, a pharmacokinetically-based approach for mitigating the impact of antimicrobial therapy on intestinal dysbiosis is proposed.
Collapse
Affiliation(s)
- Lea Gnatzy
- Department of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece
| | - George Ismailos
- Experimental, Research and Training Center ELPEN, ELPEN Pharmaceuticals, Pikermi, Greece; National Antimicrobial Testing Committee, Athens, Greece
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece
| | - Christos Reppas
- Department of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece.
| |
Collapse
|
32
|
Xiao L, Tang R, Wang J, Wan D, Yin Y, Xie L. Gut microbiota bridges the iron homeostasis and host health. SCIENCE CHINA. LIFE SCIENCES 2023; 66:1952-1975. [PMID: 37515687 DOI: 10.1007/s11427-022-2302-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/08/2023] [Indexed: 07/31/2023]
Abstract
The gut microbiota acts as a symbiotic microecosystem that plays an indispensable role in the regulation of a number of metabolic processes in the host by secreting secondary metabolites and impacting the physiology and pathophysiology of numerous organs and tissues through the circulatory system. This relationship, referred to as the "gut-X axis", is associated with the development and progression of disorders, including obesity, fatty liver and Parkinson's disease. Given its importance, the gut flora is a vital research area for the understanding and development of the novel therapeutic approaches for multiple disorders. Iron is a common but necessary element required by both mammals and bacteria. As a result, iron metabolism is closely intertwined with the gut microbiota. The host's iron homeostasis affects the composition of the gut microbiota and the interaction between host and gut microbiota through various mechanisms such as nutrient homeostasis, intestinal peaceability, gut immunity, and oxidative stress. Therefore, understanding the relationship between gut microbes and host iron metabolism is not only of enormous significance to host health but also may offer preventative and therapeutic approaches for a number of disorders that impact both parties. In this review, we delve into the connection between the dysregulation of iron metabolism and dysbiosis of gut microbiota, and how it contributes to the onset and progression of metabolic and chronic diseases.
Collapse
Affiliation(s)
- Lanling Xiao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Rui Tang
- Department of Psychiatry, The First Affiliated Hospital of Jinan University, Guangzhou, 510000, China
| | - Jie Wang
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Dan Wan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 101408, China.
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
- University of Chinese Academy of Sciences, Beijing, 101408, China.
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China.
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China.
- Department of Stomatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan, 528308, China.
| |
Collapse
|
33
|
Dong Z, Liu S, Deng Q, Li G, Tang Y, Wu X, Wan D, Yin Y. Role of iron in host-microbiota interaction and its effects on intestinal mucosal growth and immune plasticity in a piglet model. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2086-2098. [PMID: 37530911 DOI: 10.1007/s11427-022-2409-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/15/2023] [Indexed: 08/03/2023]
Abstract
Iron is an essential trace element for both the host and resident microbes in the gut. In this study, iron was administered orally and parenterally to anemic piglets to investigate the role of iron in host-microbiota interaction and its effects on intestinal mucosal growth and immune plasticity. We found that oral iron administration easily increased the abundance of Proteobacteria and Escherichia-Shigella, and decreased the abundance of Lactobacillus in the ileum. Furthermore, similar bacterial changes, namely an increase in Proteobacteria, Escherichia-Shigella, and Fusobacterium and a reduction in the Christensenellaceae_R-7_group, were observed in the colon of both iron-supplemented groups. Spearman's correlation analysis indicated that the changed Fusobacterium, Fusobacteria and Proteobacteria in the colon were positively correlated with hemoglobin, colon and spleen iron levels. Nevertheless, it was found that activated mTOR1 signaling, improved villous height and crypt depth in the ileum, enhanced immune communication, and increased protein expression of IL-22 and IL-10 in the colon of both iron-supplemented groups. In conclusion, the benefits of improved host iron outweigh the risks of altered gut microbiota for intestinal mucosal growth and immune regulation in treating iron deficiency anemia.
Collapse
Affiliation(s)
- Zhenglin Dong
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Shuan Liu
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Qingqing Deng
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Guanya Li
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Yulong Tang
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Xin Wu
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Dan Wan
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| |
Collapse
|
34
|
Karthikappallil R, Atkinson SH. Universal iron supplementation: the best strategy to tackle childhood anaemia in malaria-endemic countries? Wellcome Open Res 2023; 8:345. [PMID: 37786779 PMCID: PMC10541535 DOI: 10.12688/wellcomeopenres.19750.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2023] [Indexed: 10/04/2023] Open
Abstract
Iron deficiency presents a major public health concern in many malaria-endemic regions, and both conditions affect young children most severely. Daily iron supplementation is the standard public health intervention recommended to alleviate rates of iron deficiency in children, but there is controversy over whether universal supplementation could increase the incidence and severity of malaria infection. Current evidence suggests that iron supplementation of deficient individuals is safe and effective in high-transmission settings when accompanied by malaria prevention strategies. However, low-resource settings often struggle to effectively control the spread of malaria, and it remains unclear whether supplementation of iron replete individuals could increase their risk of malaria and other infections. This review explores the evidence for and against universal iron supplementation programmes, and alternative strategies that could be used to alleviate iron deficiency in malaria-endemic areas, while minimising potential harm.
Collapse
Affiliation(s)
- Roshan Karthikappallil
- Department of Paediatrics, University of Oxford, Oxford, England, UK
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Sarah H. Atkinson
- Department of Paediatrics, University of Oxford, Oxford, England, UK
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
- Kenya Medical Research Institute (KEMRI) Centre for Geographic Medicine Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| |
Collapse
|
35
|
Georgieff MK. The importance of iron deficiency in pregnancy on fetal, neonatal, and infant neurodevelopmental outcomes. Int J Gynaecol Obstet 2023; 162 Suppl 2:83-88. [PMID: 37538010 PMCID: PMC10421617 DOI: 10.1002/ijgo.14951] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
The role of iron in neurodevelopment has long been recognized, and the adverse effects of early-life iron deficiency on brain development and subsequent function across the lifespan continue to be a subject of research. A greater appreciation of the contribution of maternal preconceptional iron status and fetal iron accretion to offspring, postnatal iron status, and brain health across the lifespan has occurred over the past decade. This paradigm shift in thinking links two previously relatively siloed literatures: neonatal iron deficiency and postnatal iron deficiency. The understanding that iron accretion during the fetal period strongly influences postnatal iron balance has led to an appreciation of the importance and value of ensuring proper fetal iron loading. This article reviews the dynamics of fetal iron metabolism, the role of iron in the developing fetal brain, the short- and long-term neurobehavioral consequences of fetal iron underloading, and the potential mechanisms that account for the long-term effects of fetal/neonatal iron deficiency.
Collapse
|
36
|
Fischer JAJ, Pei LX, Elango R, Hou K, Goldfarb DM, Karakochuk CD. Is a Lower Dose of More Bioavailable Iron (18-mg Ferrous Bisglycinate) Noninferior to 60-mg Ferrous Sulfate in Increasing Ferritin Concentrations While Reducing Gut Inflammation and Enteropathogen Detection in Cambodian Women? A Randomized Controlled Noninferiority Trial. J Nutr 2023; 153:2453-2462. [PMID: 37271416 DOI: 10.1016/j.tjnut.2023.05.029] [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: 03/31/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/06/2023] Open
Abstract
BACKGROUND Global guidelines recommend untargeted iron supplementation for women in regions of anemia prevalence ≥40%, such as Cambodia. However, the potential harms of untargeted iron on the gut have not been rigorously studied in women and likely vary depending on iron dose and form. OBJECTIVES We investigated if a lower dose of a highly bioavailable iron amino acid chelate was as effective as the standard dose of iron salts in increasing ferritin concentrations and whether any differences were observed in gut inflammation or enteropathogen detection. METHODS A double-blind, randomized placebo-controlled noninferiority trial was conducted in Cambodia. Nonpregnant women (n = 480, 18-45 y) were randomly assigned to 60-mg ferrous sulfate, 18-mg ferrous bisglycinate, or placebo for 12 wk. Nonfasting blood and stool specimens were collected at baseline and 12 wk. Ferritin and fecal calprotectin were measured with an ELISA. A molecular assay was used to detect 11 enteropathogens in a random subset of n = 100 women. Generalized linear mixed-effects models were used to estimate the adjusted mean difference in ferritin concentrations at 12 wk (primary outcome), as compared with our 'a priori' noninferiority margin of 20 μg/L. RESULTS Baseline anemia and iron deficiency prevalence was low (17% and 6%, respectively). The adjusted mean difference in ferritin concentrations between the iron groups was 14.6 (95% confidence interval [CI]: 7.6, 21.6) μg/L. Mean ferritin concentration at 12 wk was higher in the ferrous sulfate (99 [95% CI: 95, 103] μg/L, P < 0.001) than in ferrous bisglycinate (84 [95% CI: 80, 88] μg/L) and placebo groups (78 [95% CI: 74, 82] μg/L). No differences in fecal calprotectin concentrations or enteropathogen detection were observed across groups at 12 wk. CONCLUSIONS Ferrous bisglycinate (18-mg) was not as effective as ferrous sulfate (60-mg) in increasing ferritin concentrations and did not differentially influence biomarkers of gut health in this predominantly iron-replete population of Cambodian women. This trial was registered at clinicaltrials.gov registry as NCT04017598.
Collapse
Affiliation(s)
- Jordie A J Fischer
- Food, Nutrition and Health, The University of British Columbia, Vancouver, Canada; BC Children's Hospital Research Institute, Vancouver, Canada
| | - Lulu X Pei
- Food, Nutrition and Health, The University of British Columbia, Vancouver, Canada; BC Children's Hospital Research Institute, Vancouver, Canada
| | - Rajavel Elango
- BC Children's Hospital Research Institute, Vancouver, Canada; Department of Pediatrics, the University of British Columbia, Vancouver, Canada
| | - Kroeun Hou
- Helen Keller International, Phnom Penh, Cambodia
| | - David M Goldfarb
- BC Children's Hospital Research Institute, Vancouver, Canada; Department of Pathology and Laboratory Medicine, the University of British Columbia, Vancouver, Canada
| | - Crystal D Karakochuk
- Food, Nutrition and Health, The University of British Columbia, Vancouver, Canada; BC Children's Hospital Research Institute, Vancouver, Canada.
| |
Collapse
|
37
|
Fischer JAJ, Cherian AM, Bone JN, Karakochuk CD. The effects of oral ferrous bisglycinate supplementation on hemoglobin and ferritin concentrations in adults and children: a systematic review and meta-analysis of randomized controlled trials. Nutr Rev 2023; 81:904-920. [PMID: 36728680 PMCID: PMC10331582 DOI: 10.1093/nutrit/nuac106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
CONTEXT Iron deficiency and anemia have serious consequences, especially for children and pregnant women. Iron salts are commonly provided as oral supplements to prevent and treat iron deficiency, despite poor bioavailability and frequently reported adverse side effects. Ferrous bisglycinate is a novel amino acid iron chelate that is thought to be more bioavailable and associated with fewer gastrointestinal (GI) adverse events as compared with iron salts. OBJECTIVE A systematic review and meta-analysis of randomized controlled trials (RCTs) was conducted to evaluate the effects of ferrous bisglycinate supplementation compared with other iron supplements on hemoglobin and ferritin concentrations and GI adverse events. DATA SOURCES A systematic search of electronic databases and grey literature was performed up to July 17, 2020, yielding 17 RCTs that reported hemoglobin or ferritin concentrations following at least 4 weeks' supplementation of ferrous bisglycinate compared with other iron supplements in any dose or frequency. DATA EXTRACTION Random-effects meta-analyses were conducted among trials of pregnant women (n = 9) and children (n = 4); pooled estimates were expressed as standardized mean differences (SMDs). Incidence rate ratios (IRRs) were estimated for GI adverse events, using Poisson generalized linear mixed-effects models. The remaining trials in other populations (n = 4; men and nonpregnant women) were qualitatively evaluated. DATA ANALYSIS Compared with other iron supplements, supplementation with ferrous bisglycinate for 4-20 weeks resulted in higher hemoglobin concentrations in pregnant women (SMD, 0.54 g/dL; 95% confidence interval [CI], 0.15-0.94; P < 0.01) and fewer reported GI adverse events (IRR, 0.36; 95%CI, 0.17-0.76; P < 0.01). We observed a non-significant trend for higher ferritin concentrations in pregnant women supplemented with ferrous bisglycinate. No significant differences in hemoglobin or ferritin concentrations were detected among children. CONCLUSION Ferrous bisglycinate shows some benefit over other iron supplements in increasing hemoglobin concentration and reducing GI adverse events among pregnant women. More trials are needed to assess the efficacy of ferrous bisglycinate against other iron supplements in other populations. PROSPERO REGISTRATION NO CRD42020196984.
Collapse
Affiliation(s)
- Jordie A J Fischer
- Department of Food, Nutrition, and Health, The University of British Columbia, Vancouver, British Columbia, Canada. Healthy Starts, BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Arlin M Cherian
- Department of Family Practice, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Jeffrey N Bone
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Crystal D Karakochuk
- Department of Food, Nutrition, and Health, The University of British Columbia, Vancouver, British Columbia, Canada. Healthy Starts, BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
38
|
Wang Y, Jian C. Novel plant-based meat alternatives: Implications and opportunities for consumer nutrition and health. ADVANCES IN FOOD AND NUTRITION RESEARCH 2023; 106:241-274. [PMID: 37722774 DOI: 10.1016/bs.afnr.2023.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
Abstract
Against the backdrop of the global protein transition needed to remain within planetary boundaries, there is an influx of plant-based meat alternatives that seek to approximate the texture, flavor and/or nutrient profiles of conventional animal meat. These novel plant-based meat alternatives, enabled by advances in food technology, can be fundamentally different from the whole-plant foods from which they are derived. One of the reasons is the necessity to use food additives on various occasions, since consumers' acceptance of plant-based meat products primarily depends on the organoleptic properties. Consequently, a high degree of heterogeneity in formulation and nutritional profiles exists both within and between product categories of plant-based meat alternatives with unknown effects on several aspects of human health. This is further complicated by the differences in digestibility and bioavailability between proteins from animal and plant sources, which have a profound impact on colonic fermentation, nutritional adequacy and potential health effects. On the other hand, emerging strategies provide opportunities to develop affordable, delicious and nutritious plant-based meat alternatives that align with consumer interests.
Collapse
Affiliation(s)
- Yaqin Wang
- Department of Food and Nutrition, University of Helsinki, Helsinki, Finland.
| | - Ching Jian
- Department of Food and Nutrition, University of Helsinki, Helsinki, Finland; Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
39
|
Simic S, Karczewski M, Klapdor S, Nowak A, Schubert M, Moretti D, Swinkels DW, Beuschlein F, Saleh L, Suter P, Krayenbuehl PA. Effectiveness of low-dose iron treatment in non-anaemic iron-deficient women: a prospective open-label single-arm trial. Swiss Med Wkly 2023; 153:40079. [PMID: 37229775 DOI: 10.57187/smw.2023.40079] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Iron deficiency without anaemia is highly prevalent and is particularly associated with fatigue, cognitive impairment, or poor physical endurance. Standard oral iron therapy often results in intestinal irritation with associated side effects and premature discontinuation of therapy, therefore, optimal oral iron therapy with sufficient iron absorption and minimal side effects is desirable. METHODS Thirty-six iron-deficient non-anaemic premenopausal women (serum ferritin ≤30 ng/ml, haemoglobin ≥117 g/l) with normal body mass index (BMI) and no hypermenorrhea received 6 mg of elemental oral iron (corresponding to 18.6 mg ferrous sulphate) twice daily for 8 weeks. RESULTS Participants treated with low-dose iron had an average age of 28 years and a BMI of 21 kg/m2. Their serum ferritin and haemoglobin increased significantly from 18 ng/ml to 33 ng/ml (p <0.001) and from 135 g/l to 138 g/l (p = 0.014), respectively. Systolic blood pressure increased from 114 mmHg to 120 mmHg (p = 0.003). Self-reported health status improved after 8 weeks (p <0.001) and only one woman reported gastrointestinal side effects (3%). CONCLUSION This prospective open-label single-arm trial shows that oral iron treatment of 6 mg of elemental iron twice daily over 8 weeks is effective in iron-deficient non-anaemic women. Due to the negligible side effects, low-dose iron treatment is a valuable therapeutic option for iron-deficient non-anaemic women with normal BMI and menstruation. Further placebo-controlled studies with a larger number of participants are needed to confirm these results. CLINICALTRIALS gov NCT04636060.
Collapse
Affiliation(s)
- Stana Simic
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Switzerland
| | - Maximilian Karczewski
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Switzerland
| | - Silke Klapdor
- Department of Internal Medicine, Lucerne Cantonal Hospital, Switzerland
| | - Albina Nowak
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Switzerland
- Department of Internal Medicine, Psychiatric University Clinic Zurich, Switzerland
| | - Morton Schubert
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Switzerland
| | - Diego Moretti
- ETH Zurich, Laboratory of Human Nutrition, Institute of Food Nutrition and Health, Switzerland
| | - Dorine W Swinkels
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Switzerland
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Switzerland
- Translational Metabolic Laboratory (TML), Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen and Sanquin Blood Bank, Amsterdam, the Netherlands
| | - Lanja Saleh
- Department of General Routine and Emergency Analysis, Institute of Clinical Chemistry, University Hospital of Zurich, Switzerland
| | - Paolo Suter
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Switzerland
| | - Pierre-Alexandre Krayenbuehl
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich, Switzerland
- General Practice, Brauereistrasse, Uster-Zurich, Switzerland
| |
Collapse
|
40
|
Tsilika M, Mitrou J, Antonakos N, Tseti IK, Damoraki G, Leventogiannis K, Giamarellos-Bourboulis EJ. An active new formulation of iron carried by aspartyl casein for iron-deficiency anemia: results of the ACCESS trial. Ann Hematol 2023; 102:1341-1349. [PMID: 37022473 PMCID: PMC10182142 DOI: 10.1007/s00277-023-05197-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 03/23/2023] [Indexed: 04/07/2023]
Abstract
Oral iron supplementation is the cornerstone for the management of iron-deficiency anemia. A new oral formulation of iron conjugated with N-aspartyl-casein (Fe-ASP) (Omalin®, Uni-Pharma) is studied in the ACCESS double-blind, double-dummy randomized clinical trial; 60 patients were randomized to 12-week oral treatment twice every day either with oral ferrous sulfate (FeSO4) delivering 47 mg elementary iron or oral Fe-ASP delivering 40 mg elementary iron. Participants had hemoglobin less than 10 g/dl, decreased red blood cell (RBC) count, and ferritin lower than 30 ng/ml; patients with a medical history of malignancy were excluded. The primary endpoint was the increase of Hb in the first 4 weeks of treatment, and the study was powered for non-inferiority. A new score of global improvement was introduced where all participants were given one point for any at least 10% increase of Hb, RBC, and reticulocytes. At week 4, the mean (SE) change of Hb was 0.76 g/dl in the FeSO4 group and 0.83 g/dl in the Fe-ASP group (p: 0.876). The odds for worse allocation of the global score were 0.35 in the Fe-ASP group compared to the FeSO4 group. Patients in the Fe-ASP group experienced a significant decrease in the number of IDA-related physical signs by week 4. No differences were found between the two groups in any of the patient-reported outcomes of fatigue and of gastrointestinal adverse events either at week 4 or at week 12. ACCESS is the most recent clinical trial showing the non-inferiority of Fe-ASP to FeSO4 for the primary endpoint of the Hb change.
Collapse
Affiliation(s)
- Maria Tsilika
- 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - John Mitrou
- 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Antonakos
- 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Georgia Damoraki
- 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Leventogiannis
- 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos J Giamarellos-Bourboulis
- 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
- 4th Department of Internal Medicine, ATTIKON University General Hospital, 1 Rimini Street, 124 62, Athens, Greece.
| |
Collapse
|
41
|
Gupta MS, Kumar TP, Reddy D, Pathak K, Gowda DV, Babu AVN, Aodah AH, Khafagy ES, Alotaibi HF, Abu Lila AS, Moin A, Hussin T. Development and Characterization of Pullulan-Based Orodispersible Films of Iron. Pharmaceutics 2023; 15:pharmaceutics15031027. [PMID: 36986887 PMCID: PMC10056616 DOI: 10.3390/pharmaceutics15031027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Iron deficiency is the principal cause of nutritional anemia and it constitutes a major health problem, especially during pregnancy. Despite the availability of various non-invasive traditional oral dosage forms such as tablets, capsules, and liquid preparations of iron, they are hard to consume for special populations such as pregnant women, pediatric, and geriatric patients with dysphagia and vomiting tendency. The objective of the present study was to develop and characterize pullulan-based iron-loaded orodispersible films (i-ODFs). Microparticles of iron were formulated by a microencapsulation technique, to mask the bitter taste of iron, and ODFs were fabricated by a modified solvent casting method. Morphological characteristics of the microparticles were identified by optical microscopy and the percentage of iron loading was evaluated by inductively coupled plasma optical emission spectroscopy (ICP-OES). The fabricated i-ODFs were evaluated for their morphology by scanning electron microscopy. Other parameters including thickness, folding endurance, tensile strength, weight variation, disintegration time, percentage moisture loss, surface pH, and in vivo animal safety were evaluated. Lastly, stability studies were carried out at a temperature of 25 °C/60% RH. The results of the study confirmed that pullulan-based i-ODFs had good physicochemical properties, excellent disintegration time, and optimal stability at specified storage conditions. Most importantly, the i-ODFs were free from irritation when administered to the tongue as confirmed by the hamster cheek pouch model and surface pH determination. Collectively, the present study suggests that the film-forming agent, pullulan, could be successfully employed on a lab scale to formulate orodispersible films of iron. In addition, i-ODFs can be processed easily on a large scale for commercial use.
Collapse
Affiliation(s)
- Maram Suresh Gupta
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysore 570015, India
| | - Tegginamath Pramod Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysore 570015, India
| | - Dinesh Reddy
- Aavishkar Oral Strips Private Limited, Plot No. 109/3, Phase-II, Sector 2, Lane No. 6 IDA Cherlapally, Hyderabad 500051, India
| | - Kamla Pathak
- Pharmacy College Saifai, Uttar Pradesh University of Medical Sciences, Etawah 206130, India
| | | | - A V Naresh Babu
- Aavishkar Oral Strips Private Limited, Plot No. 109/3, Phase-II, Sector 2, Lane No. 6 IDA Cherlapally, Hyderabad 500051, India
| | - Alhussain H Aodah
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj 11942, Saudi Arabia
| | - El-Sayed Khafagy
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj 11942, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Hadil Faris Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Amr Selim Abu Lila
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il 81442, Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Afrasim Moin
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il 81442, Saudi Arabia
| | - Talib Hussin
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Ha'il, Ha'il 81442, Saudi Arabia
| |
Collapse
|
42
|
Urrutia-Baca V, Hernández-Hernández S, Martínez L, Dávila-Vega J, Chuck-Hernández C. The Role of Probiotics in Dairy Foods and Strategies to Evaluate Their Functional Modifications. FOOD REVIEWS INTERNATIONAL 2023. [DOI: 10.1080/87559129.2023.2172426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Affiliation(s)
- V.H Urrutia-Baca
- Tecnologico de Monterrey, The Institute for Obesity Research, Monterrey, NL, México
| | | | - L.M. Martínez
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey, NL, México
| | - J.P Dávila-Vega
- Tecnologico de Monterrey, The Institute for Obesity Research, Monterrey, NL, México
| | - C. Chuck-Hernández
- Tecnologico de Monterrey, The Institute for Obesity Research, Monterrey, NL, México
| |
Collapse
|
43
|
Lin D, Medeiros DM. The microbiome as a major function of the gastrointestinal tract and its implication in micronutrient metabolism and chronic diseases. Nutr Res 2023; 112:30-45. [PMID: 36965327 DOI: 10.1016/j.nutres.2023.02.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/22/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023]
Abstract
The composition and function of microbes harbored in the human gastrointestinal lumen have been underestimated for centuries because of the underdevelopment of nucleotide sequencing techniques and the lack of humanized gnotobiotic models. Now, we appreciate that the gut microbiome is an integral part of the human body and exerts considerable roles in host health and diseases. Dietary factors can induce changes in the microbial community composition, metabolism, and function, thereby altering the host immune response, and consequently, may influence disease risks. An imbalance of gut microbiome homeostasis (i.e., dysbiosis) has been linked to several chronic diseases, such as inflammatory bowel diseases, obesity, and diabetes. Remarkable progress has recently been made in better understanding the extent to which the influence of the diet-microbiota interaction on host health outcomes in both animal models and human participants. However, the exact causality of the gut microbiome on the development of diseases is still controversial. In this review, we will briefly describe the general structure and function of the intestine and the process of nutrient absorption in humans. This is followed by a summarization of the recent updates on interactions between gut microbiota and individual micronutrients, including carotenoids, vitamin A, vitamin D, vitamin C, folate, iron, and zinc. In the opinion of the authors, these nutrients were identified as representative of vitamins and minerals with sufficient research on their roles in the microbiome. The host responses to the gut microbiome will also be discussed. Future direction in microbiome research, for example, precision microbiome, will be proposed.
Collapse
Affiliation(s)
- Dingbo Lin
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078.
| | - Denis M Medeiros
- Division of Molecular Biology and Biochemistry, University of Missouri-Kansas City, Kansas City, MO 64108
| |
Collapse
|
44
|
Adetola OY, Taylor JRN, Duodu KG. Can consumption of local micronutrient- and absorption enhancer-rich plant foods together with starchy staples improve bioavailable iron and zinc in diets of at-risk African populations? Int J Food Sci Nutr 2023; 74:188-208. [PMID: 36843328 DOI: 10.1080/09637486.2023.2182740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Abstract
Iron and zinc deficiencies remain prevalent in developing countries, often due to monotonous starchy diets that are low in bioavailable minerals. This review addresses the question as to whether consumption of starchy staple foods in Africa together with micronutrient-dense and absorption enhancer-rich plant foods can enhance iron and zinc bioavailability in the diets of at-risk populations. While green leafy vegetables (GLVs) fortification of starchy staples can improve mineral contents, especially iron, it may not improve bioavailable iron and zinc, due to GLVs' high contents of mineral absorption inhibitors, notably polyphenols, phytate and calcium. Fruits, although low in minerals, could improve bioavailable iron and zinc in the staples because of their high ascorbic and citric acid and/or β-carotene contents, which can form soluble chelates with the minerals. More human studies are needed to establish whether such a technology or fortification strategy can improve bioavailable iron and zinc in African-type plant-based diets.
Collapse
Affiliation(s)
- Oluyimika Y Adetola
- Department of Consumer and Food Sciences, University of Pretoria, Hatfield, South Africa
| | - John R N Taylor
- Department of Consumer and Food Sciences, University of Pretoria, Hatfield, South Africa
| | - K G Duodu
- Department of Consumer and Food Sciences, University of Pretoria, Hatfield, South Africa
| |
Collapse
|
45
|
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: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 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
|
46
|
Huynh U, Zastrow ML. Metallobiology of Lactobacillaceae in the gut microbiome. J Inorg Biochem 2023; 238:112023. [PMID: 36270041 PMCID: PMC9888405 DOI: 10.1016/j.jinorgbio.2022.112023] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/26/2022] [Accepted: 10/03/2022] [Indexed: 11/21/2022]
Abstract
Lactobacillaceae are a diverse family of lactic acid bacteria found in the gut microbiota of humans and many animals. These bacteria exhibit beneficial effects on intestinal health, including modulating the immune system and providing protection against pathogens, and many species are frequently used as probiotics. Gut bacteria acquire essential metal ions, like iron, zinc, and manganese, through the host diet and changes to the levels of these metals are often linked to alterations in microbial community composition, susceptibility to infection, and gastrointestinal diseases. Lactobacillaceae are frequently among the organisms increased or decreased in abundance due to changes in metal availability, yet many of the molecular mechanisms underlying these changes have yet to be defined. Metal requirements and metallotransporters have been studied in some species of Lactobacillaceae, but few of the mechanisms used by these bacteria to respond to metal limitation or excess have been investigated. This review provides a current overview of these mechanisms and covers how iron, zinc, and manganese impact Lactobacillaceae in the gut microbiota with an emphasis on their biochemical roles, requirements, and homeostatic mechanisms in several species.
Collapse
Affiliation(s)
- Uyen Huynh
- Department of Chemistry, University of Houston, Houston, TX, USA
| | | |
Collapse
|
47
|
Husmann FMD, Zimmermann MB, Herter-Aeberli I. The Effect of Prebiotics on Human Iron Absorption: A Review. Adv Nutr 2022; 13:2296-2304. [PMID: 35816457 PMCID: PMC9776726 DOI: 10.1093/advances/nmac079] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/22/2022] [Accepted: 07/06/2022] [Indexed: 01/29/2023] Open
Abstract
Iron deficiency remains the most common nutritional deficiency. Oral iron supplementation is the recommended first-line treatment and used as a preventive measure as well. Enhancers of iron absorption are highly sought after to improve supplementation outcomes. Evidence from animal and human studies exists that prebiotics can enhance iron absorption. The purpose of this present narrative review of the literature is to summarize the existing evidence on the effects of prebiotics on human iron absorption. Relevant articles were identified from PUBMED, Scopus, and Web of Science from inception to November 2021. Only human trials investigating the effect of prebiotics on iron absorption were included. Eleven articles were identified and included for review. There are promising findings supporting an enhancing effect of certain prebiotics, but inconsistencies between the studies and results exist. The most convincing evidence exists for the prebiotics galacto-oligosaccharides and fructo-oligosaccharides combined with the commonly used iron compound ferrous fumarate, from studies in adult women with low iron stores and in anemic infants. Many factors seem to play a role in the enhancing effect of prebiotics on iron absorption such as type of prebiotic, dose, acute (single-dose) or chronic (long-term) prebiotic consumption, iron compound, iron status, inflammatory status, and age of the population studied. More research investigating the optimal combination of prebiotic, iron compound, and dose as well as the effect of long-term application on iron status outcomes is needed.
Collapse
Affiliation(s)
- Frederike M D Husmann
- Laboratory of Human Nutrition, Institute of Food, Nutrition and
Health, ETH Zurich, Zurich, Switzerland
| | - Michael B Zimmermann
- Laboratory of Human Nutrition, Institute of Food, Nutrition and
Health, ETH Zurich, Zurich, Switzerland
| | - Isabelle Herter-Aeberli
- Laboratory of Human Nutrition, Institute of Food, Nutrition and
Health, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
48
|
Davis EW, Wong CP, Arnold HK, Kasschau K, Gaulke CA, Sharpton TJ, Ho E. Age and micronutrient effects on the microbiome in a mouse model of zinc depletion and supplementation. PLoS One 2022; 17:e0275352. [PMID: 36534653 PMCID: PMC9762596 DOI: 10.1371/journal.pone.0275352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Older adult populations are at risk for zinc deficiency, which may predispose them to immune dysfunction and age-related chronic inflammation that drives myriad diseases and disorders. Recent work also implicates the gut microbiome in the onset and severity of age-related inflammation, indicating that dietary zinc status and the gut microbiome may interact to impact age-related host immunity. We hypothesize that age-related alterations in the gut microbiome contribute to the demonstrated zinc deficits in host zinc levels and increased inflammation. We tested this hypothesis with a multifactor two-part study design in a C57BL/6 mouse model. The two studies included young (2 month old) and aged (24 month old) mice fed either (1) a zinc adequate or zinc supplemented diet, or (2) a zinc adequate or marginal zinc deficient diet, respectively. Overall microbiome composition did not significantly change with zinc status; beta diversity was driven almost exclusively by age effects. Microbiome differences due to age are evident at all taxonomic levels, with more than half of all taxonomic units significantly different. Furthermore, we found 150 out of 186 genera were significantly different between the two age groups, with Bacteriodes and Parabacteroides being the primary taxa of young and old mice, respectively. These data suggest that modulating individual micronutrient concentrations does not lead to comprehensive microbiome shifts, but rather affects specific components of the gut microbiome. However, a phylogenetic agglomeration technique (ClaaTU) revealed phylogenetic clades that respond to modulation of dietary zinc status and inflammation state in an age-dependent manner. Collectively, these results suggest that a complex interplay exists between host age, gut microbiome composition, and dietary zinc status.
Collapse
Affiliation(s)
- Edward W. Davis
- Center for Quantitative Life Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Carmen P. Wong
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, United States of America
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Holly K. Arnold
- Department of Microbiology, Oregon State University, Corvallis, Oregon, United States of America
| | - Kristin Kasschau
- Department of Microbiology, Oregon State University, Corvallis, Oregon, United States of America
| | - Christopher A. Gaulke
- Department of Microbiology, Oregon State University, Corvallis, Oregon, United States of America
| | - Thomas J. Sharpton
- Department of Microbiology, Oregon State University, Corvallis, Oregon, United States of America
- Department of Statistics, Oregon State University, Corvallis, Oregon, United States of America
| | - Emily Ho
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, United States of America
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, United States of America
- * E-mail:
| |
Collapse
|
49
|
Effects of Different Doses, Forms, and Frequencies of Zinc Supplementation on Biomarkers of Iron and Zinc Status among Young Children in Dhaka, Bangladesh. Nutrients 2022; 14:nu14245334. [PMID: 36558493 PMCID: PMC9781687 DOI: 10.3390/nu14245334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Young children in resource-constrained settings are susceptible to zinc deficiency and its deleterious health effects. The objective of this secondary analysis was to evaluate the effects of the following six interventions on biomarkers of iron and zinc status among a subgroup of young children in Dhaka, Bangladesh, who participated in the Zinc in Powders Trial (ZiPT): (1) standard micronutrient powders (MNPs) containing 4.1 mg zinc and 10 mg iron, daily; (2) high-zinc (10 mg) and low-iron (6 mg) (HiZn LoFe) MNP, daily; (3) HiZn (10 mg) and LoFe (6 mg)/HiZn (10 mg) and no-iron MNPs on alternating days; (4) dispersible zinc tablet (10 mg), daily; (5) dispersible zinc tablet (10 mg), daily for 2 weeks at enrollment and at 12 weeks; (6) placebo powder, daily. At the end of the 24 week intervention period, children in the daily dispersible zinc tablet group exhibited a mean serum zinc concentration (SZC) of 92.5 μg/dL, which was significantly higher than all other groups except the HiZn LoFe MNP alternating group (81.3 μg/dL). MNPs containing 10 mg and 6 mg of iron had a similar impact on biomarkers of iron status, with no evidence of an adverse interaction with zinc.
Collapse
|
50
|
Sneak-peek into iron deficiency anemia in India: The need for food-based interventions and enhancing iron bioavailability. Food Res Int 2022; 162:111927. [DOI: 10.1016/j.foodres.2022.111927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 09/02/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022]
|