1
|
Wang X, Lenartowicz M, Mazgaj R, Ogłuszka M, Szkopek D, Zaworski K, Kopeć Z, Żelazowska B, Lipiński P, Woliński J, Starzyński RR. Preterm Piglets Born by Cesarean Section as a Suitable Animal Model for the Study of Iron Metabolism in Premature Infants. Int J Mol Sci 2024; 25:11215. [PMID: 39456997 PMCID: PMC11508764 DOI: 10.3390/ijms252011215] [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: 09/14/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Preterm infants are most at risk of iron deficiency. However, our knowledge of the regulation of iron homeostasis in preterm infants is poor. The main goal of our research was to develop and validate an animal model of human prematurity to assess iron status in preterm infants. We performed a cesarean section on sows on the 109th day of pregnancy, which corresponds to the last trimester of human pregnancy. Preterm piglets showed decreased body weight, red blood cell indices, plasma iron level and transferrin saturation. Interestingly, higher hepatic and splenic non-heme iron content and plasma and hepatic ferritin levels were found in premature piglets compared with term ones. In addition, premature piglets showed higher mRNA levels of iron-regulatory hormone hepcidin in the liver than term animals, which have not been reflected in higher plasma hepcidin-25 levels. We also showed changes in hepcidin regulators, including hepatic bone morphogenetic protein 6, plasma erythroferrone and growth differentiation factor 15 in preterm piglets. Consequently, no difference was observed in iron-exporter ferroportin levels in the spleen and liver. Overall, it seems that premature piglets show a pattern of iron metabolism characteristic of functional iron deficiency and iron accumulation in the tissue.
Collapse
Affiliation(s)
- Xiuying Wang
- Laboratory of Iron Molecular Biology, Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland; (X.W.); (R.M.); (Z.K.); (B.Ż.); (P.L.)
| | - Małgorzata Lenartowicz
- Laboratory of Genetics and Evolutionism, Institute of Zoology and Biomedical Research, Jagiellonian University, 30-387 Kraków, Poland
| | - Rafał Mazgaj
- Laboratory of Iron Molecular Biology, Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland; (X.W.); (R.M.); (Z.K.); (B.Ż.); (P.L.)
| | - Magdalena Ogłuszka
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland;
| | - Dominika Szkopek
- Laboratory of Large Animal Models, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland; (D.S.); (J.W.)
| | - Kamil Zaworski
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland;
| | - Zuzanna Kopeć
- Laboratory of Iron Molecular Biology, Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland; (X.W.); (R.M.); (Z.K.); (B.Ż.); (P.L.)
| | - Beata Żelazowska
- Laboratory of Iron Molecular Biology, Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland; (X.W.); (R.M.); (Z.K.); (B.Ż.); (P.L.)
| | - Paweł Lipiński
- Laboratory of Iron Molecular Biology, Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland; (X.W.); (R.M.); (Z.K.); (B.Ż.); (P.L.)
| | - Jarosław Woliński
- Laboratory of Large Animal Models, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland; (D.S.); (J.W.)
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland;
| | - Rafał Radosław Starzyński
- Laboratory of Iron Molecular Biology, Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland; (X.W.); (R.M.); (Z.K.); (B.Ż.); (P.L.)
| |
Collapse
|
2
|
Grander M, Haschka D, Indelicato E, Kremser C, Amprosi M, Nachbauer W, Henninger B, Stefani A, Högl B, Fischer C, Seifert M, Kiechl S, Weiss G, Boesch S. Genetic Determined Iron Starvation Signature in Friedreich's Ataxia. Mov Disord 2024; 39:1088-1098. [PMID: 38686449 DOI: 10.1002/mds.29819] [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: 12/28/2023] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Early studies in cellular models suggested an iron accumulation in Friedreich's ataxia (FA), yet findings from patients are lacking. OBJECTIVES The objective is to characterize systemic iron metabolism, body iron storages, and intracellular iron regulation in FA patients. METHODS In FA patients and matched healthy controls, we assessed serum iron parameters, regulatory hormones as well as the expression of regulatory proteins and iron distribution in peripheral blood mononuclear cells (PBMCs). We applied magnetic resonance imaging with R2*-relaxometry to quantify iron storages in the liver, spleen, and pancreas. Across all evaluations, we assessed the influence of the genetic severity as expressed by the length of the shorter GAA-expansion (GAA1). RESULTS We recruited 40 FA patients (19 women). Compared to controls, FA patients displayed lower serum iron and transferrin saturation. Serum ferritin, hepcidin, mean corpuscular hemoglobin and mean corpuscular volume in FA inversely correlated with the GAA1-repeat length, indicating iron deficiency and restricted availability for erythropoiesis with increasing genetic severity. R2*-relaxometry revealed a reduction of splenic and hepatic iron stores in FA. Liver and spleen R2* values inversely correlated with the GAA1-repeat length. FA PBMCs displayed downregulation of ferritin and upregulation of transferrin receptor and divalent metal transporter-1 mRNA, particularly in patients with >500 GAA1-repeats. In FA PBMCs, intracellular iron was not increased, but shifted toward mitochondria. CONCLUSIONS We provide evidence for a previously unrecognized iron starvation signature at systemic and cellular levels in FA patients, which is related to the underlying genetic severity. These findings challenge the use of systemic iron lowering therapies in FA. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Manuel Grander
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - David Haschka
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Elisabetta Indelicato
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Kremser
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Matthias Amprosi
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Wolfgang Nachbauer
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Benjamin Henninger
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ambra Stefani
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Birgit Högl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christine Fischer
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Kiechl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- VASCage, Centre on Clinical Stroke Research, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Sylvia Boesch
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
3
|
Schleh MW, Ameka M, Rodriguez A, Hasty AH. Deficiency of the hemoglobin-haptoglobin receptor, CD163, worsens insulin sensitivity in obese male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596887. [PMID: 38895370 PMCID: PMC11185572 DOI: 10.1101/2024.05.31.596887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Excessive iron accumulation in metabolic organs such as the adipose tissue, liver, and skeletal muscle is associated with increased diabetes risk. Tissue-resident macrophages serve multiple roles including managing inflammatory tone and regulating parachymal iron homeostasis; thus protecting against metabolic dysfunction upon iron overload. The scavenger receptor CD163 is uniquely present on tissue-resident macrophages, and plays a significant role in iron homeostasis by clearing extracellular hemoglobin-haptoglobin complexes, thereby limiting oxidative damage caused by free hemoglobin in metabolic tissues. We show that the absence of CD163 exacerbates glucose intolerance and insulin resistance in male mice with obesity. Additionally, loss of CD163 reduced the expression of iron regulatory genes (Tfr1, Cisd1, Slc40a1) in adipose tissue macrophages and anti-inflammatory (M2-like) bone marrow-derived macrophages (BMDMs). Further, CD163 deficiency mediated a pro-inflammatory shift and limited hemoglobin scavenging specifically in M2-like BMDMs. To this end, iron buffering was diminished in inguinal white adipose tissue (iWAT) macrophages in vivo, which culminated in iron spillover into adipocytes and CD45+CD11B- non-myeloid immune cells in iWAT. These findings show that CD163 on tissue-resident macrophages is critical for their anti-inflammatory and hemoglobin scavenging roles, and its absence results in impaired systemic insulin action in an obese setting.
Collapse
Affiliation(s)
- Michael W. Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine; Nashville, TN 37232, USA
| | - Magdalene Ameka
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine; Nashville, TN 37232, USA
| | - Alec Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine; Nashville, TN 37232, USA
| | - Alyssa H. Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine; Nashville, TN 37232, USA
- VA Tennessee Valley Healthcare System; Nashville, TN 37212, USA
| |
Collapse
|
4
|
Țichil I, Mitre I, Zdrenghea MT, Bojan AS, Tomuleasa CI, Cenariu D. A Review of Key Regulators of Steady-State and Ineffective Erythropoiesis. J Clin Med 2024; 13:2585. [PMID: 38731114 PMCID: PMC11084473 DOI: 10.3390/jcm13092585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Erythropoiesis is initiated with the transformation of multipotent hematopoietic stem cells into committed erythroid progenitor cells in the erythroblastic islands of the bone marrow in adults. These cells undergo several stages of differentiation, including erythroblast formation, normoblast formation, and finally, the expulsion of the nucleus to form mature red blood cells. The erythropoietin (EPO) pathway, which is activated by hypoxia, induces stimulation of the erythroid progenitor cells and the promotion of their proliferation and survival as well as maturation and hemoglobin synthesis. The regulation of erythropoiesis is a complex and dynamic interaction of a myriad of factors, such as transcription factors (GATA-1, STAT5), cytokines (IL-3, IL-6, IL-11), iron metabolism and cell cycle regulators. Multiple microRNAs are involved in erythropoiesis, mediating cell growth and development, regulating oxidative stress, erythrocyte maturation and differentiation, hemoglobin synthesis, transferrin function and iron homeostasis. This review aims to explore the physiology of steady-state erythropoiesis and to outline key mechanisms involved in ineffective erythropoiesis linked to anemia, chronic inflammation, stress, and hematological malignancies. Studying aberrations in erythropoiesis in various diseases allows a more in-depth understanding of the heterogeneity within erythroid populations and the development of gene therapies to treat hematological disorders.
Collapse
Affiliation(s)
- Ioana Țichil
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- Department of Haematology, “Ion Chiricuta” Institute of Oncology, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Ileana Mitre
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
| | - Mihnea Tudor Zdrenghea
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- Department of Haematology, “Ion Chiricuta” Institute of Oncology, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Anca Simona Bojan
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- Department of Haematology, “Ion Chiricuta” Institute of Oncology, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Ciprian Ionuț Tomuleasa
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- Department of Haematology, “Ion Chiricuta” Institute of Oncology, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
- MEDFUTURE—Research Centre for Advanced Medicine, 8 Louis Pasteur Street, 400347 Cluj-Napoca, Romania
| | - Diana Cenariu
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- MEDFUTURE—Research Centre for Advanced Medicine, 8 Louis Pasteur Street, 400347 Cluj-Napoca, Romania
| |
Collapse
|
5
|
Youssry I, Samy RM, AbdelMohsen M, Salama NM. The association between growth differentiation factor-15, erythroferrone, and iron status in thalassemic patients. Pediatr Res 2024; 95:1095-1100. [PMID: 37464096 PMCID: PMC10920194 DOI: 10.1038/s41390-023-02729-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/20/2023]
Abstract
BACKGROUND Iron overload can result in grave consequences in thalassemic patients, despite the availability of iron chelators. Therefore, alternative pathways aiming to reduce iron toxicity are currently investigated. Among which, reduction of iron absorption through control of hepcidin production appears to be promising. In this study, we investigated growth differentiation factor-15 (GDF15) and erythroferrone (ERFE) as potential suppressors of hepcidin. METHODS This cross-sectional study was conducted on 61 thalassemic patients and 60 healthy controls. The frequency of GDF15 gene polymorphism (rs4808793) (-3148C/G), serum level of GDF15 and erythroferrone were measured and correlated with those of hepcidin and serum ferritin. RESULTS The presence of GDF15 gene mutations were significantly higher in the patients' group compared to controls (P value 0.035). Also, thalassemia patients had significantly higher levels of GDF15 and ERFE and lower hepcidin levels than controls (P value < 0.001). Serum hepcidin level showed significantly negative correlations with GDF15, ERFE, reticulocyte count, LDH level, and serum ferritin. Contrarily, it had highly significant positive correlation with hemoglobin. CONCLUSIONS High level of GDF15 and/or ERFE may inhibit hepcidin production and increase iron load in patients with thalassemia; therefore, medications that suppress their actions may provide new therapeutic potentials for iron toxicity. IMPACT Iron overload continues to be a major contributor to high morbidity and mortality in patients with thalassemia. New strategies together with proper chelation, need to be developed to minimize the effect of iron toxicity. Growth differentiation factor-15 (GDF15) and erythroferrone (ERFE) inhibit hepcidin production and increase iron levels in conditions with ineffective erythropoiesis. Medications that suppress the production or interfere with the action of GDF15 or ERFE may represent new therapeutic potentials for iron toxicity. Prevention of iron toxicity will significantly reduce morbidity and mortality and improve the quality of life of thalassemia patients.
Collapse
Affiliation(s)
- Ilham Youssry
- Pediatric Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Rania M Samy
- Clinical Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Niveen M Salama
- Pediatric Department, Faculty of Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
6
|
Settakorn K, Hantrakool S, Petiwathayakorn T, Hutachok N, Tantiworawit A, Charoenkwan P, Chalortham N, Chompupoung A, Paradee N, Koonyosying P, Srichairatanakool S. A randomized placebo-controlled clinical trial of oral green tea epigallocatechin 3-gallate on erythropoiesis and oxidative stress in transfusion-dependent β-thalassemia patients. Front Mol Biosci 2024; 10:1248742. [PMID: 38328786 PMCID: PMC10848917 DOI: 10.3389/fmolb.2023.1248742] [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: 06/27/2023] [Accepted: 12/28/2023] [Indexed: 02/09/2024] Open
Abstract
β-Thalassemia patients suffer from ineffective erythropoiesis and increased red blood cell (RBC) hemolysis. Blood transfusion, erythropoietic enhancement, and antioxidant supplementation can ameliorate chronic anemia. Green tea extract (GTE) is comprised of catechin derivatives, of which epigallocatechin-3-gallate (EGCG) is the most abundant, presenting free-radical scavenging, iron-chelating, and erythropoiesis-protective effects. The present study aimed to evaluate the effects of GTE tablets on the primary outcome of erythropoiesis and oxidative stress parameters in transfusion-dependent β-thalassemia (TDT) patients. Twenty-seven TDT patients were randomly divided into placebo and GTE tablet (50 and 100 mg EGCG equivalent) groups and assigned to consume the product once daily for 60 days. Blood was collected for analysis of hematological, biochemical, and oxidative stress parameters. Accordingly, consumption of GTE tablets improved blood hemoglobin levels when compared with the placebo; however, there were more responders to the GTE tablets. Interestingly, amounts of nonheme iron in RBC membranes tended to decrease in both GTE tablet groups when compared with the placebo. Importantly, consumption of GTE tablets lowered plasma levels of erythroferrone (p < 0.05) and reduced bilirubin non-significantly and dose-independently. Thus, GTE tablets could improve RBC hemolysis and modulate erythropoiesis regulators in transfusion-dependent thalassemia patients.
Collapse
Affiliation(s)
- Kornvipa Settakorn
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sasinee Hantrakool
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Thalassemia and Hematology Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Nuntouchaporn Hutachok
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Adisak Tantiworawit
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Thalassemia and Hematology Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pimlak Charoenkwan
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nopphadol Chalortham
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| | | | - Narisara Paradee
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pimpisid Koonyosying
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | |
Collapse
|
7
|
Tseng YJ, Kageyama Y, Murdaugh RL, Kitano A, Kim JH, Hoegenauer KA, Tiessen J, Smith MH, Uryu H, Takahashi K, Martin JF, Samee MAH, Nakada D. Increased iron uptake by splenic hematopoietic stem cells promotes TET2-dependent erythroid regeneration. Nat Commun 2024; 15:538. [PMID: 38225226 PMCID: PMC10789814 DOI: 10.1038/s41467-024-44718-0] [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: 10/01/2021] [Accepted: 01/02/2024] [Indexed: 01/17/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are capable of regenerating the blood system, but the instructive cues that direct HSCs to regenerate particular lineages lost to the injury remain elusive. Here, we show that iron is increasingly taken up by HSCs during anemia and induces erythroid gene expression and regeneration in a Tet2-dependent manner. Lineage tracing of HSCs reveals that HSCs respond to hemolytic anemia by increasing erythroid output. The number of HSCs in the spleen, but not bone marrow, increases upon anemia and these HSCs exhibit enhanced proliferation, erythroid differentiation, iron uptake, and TET2 protein expression. Increased iron in HSCs promotes DNA demethylation and expression of erythroid genes. Suppressing iron uptake or TET2 expression impairs erythroid genes expression and erythroid differentiation of HSCs; iron supplementation, however, augments these processes. These results establish that the physiological level of iron taken up by HSCs has an instructive role in promoting erythroid-biased differentiation of HSCs.
Collapse
Affiliation(s)
- Yu-Jung Tseng
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yuki Kageyama
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Rebecca L Murdaugh
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ayumi Kitano
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jong Hwan Kim
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kevin A Hoegenauer
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jonathan Tiessen
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Mackenzie H Smith
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Hidetaka Uryu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - James F Martin
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, 77030, USA
| | - Md Abul Hassan Samee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Daisuke Nakada
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
8
|
Liongue C, Ward AC. Myeloproliferative Neoplasms: Diseases Mediated by Chronic Activation of Signal Transducer and Activator of Transcription (STAT) Proteins. Cancers (Basel) 2024; 16:313. [PMID: 38254802 PMCID: PMC10813624 DOI: 10.3390/cancers16020313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs) are hematopoietic diseases characterized by the clonal expansion of single or multiple lineages of differentiated myeloid cells that accumulate in the blood and bone marrow. MPNs are grouped into distinct categories based on key clinical presentations and distinctive mutational hallmarks. These include chronic myeloid leukemia (CML), which is strongly associated with the signature BCR::ABL1 gene translocation, polycythemia vera (PV), essential thrombocythemia (ET), and primary (idiopathic) myelofibrosis (PMF), typically accompanied by molecular alterations in the JAK2, MPL, or CALR genes. There are also rarer forms such as chronic neutrophilic leukemia (CNL), which involves mutations in the CSF3R gene. However, rather than focusing on the differences between these alternate disease categories, this review aims to present a unifying molecular etiology in which these overlapping diseases are best understood as disruptions of normal hematopoietic signaling: specifically, the chronic activation of signaling pathways, particularly involving signal transducer and activator of transcription (STAT) transcription factors, most notably STAT5B, leading to the sustained stimulation of myelopoiesis, which underpins the various disease sequalae.
Collapse
Affiliation(s)
- Clifford Liongue
- School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia;
- Institute for Mental and Physical Health and Clinical Translation, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Alister C. Ward
- School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia;
- Institute for Mental and Physical Health and Clinical Translation, Deakin University, Waurn Ponds, VIC 3216, Australia
| |
Collapse
|
9
|
Tang P, Wang H. Regulation of erythropoiesis: emerging concepts and therapeutic implications. Hematology 2023; 28:2250645. [PMID: 37639548 DOI: 10.1080/16078454.2023.2250645] [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: 04/19/2023] [Accepted: 08/17/2023] [Indexed: 08/31/2023] Open
Abstract
The process of erythropoiesis is complex and involves the transfer of cells from the yolk sac to the fetal hepar and, ultimately, to the bone marrow during embryonic development. Within the bone marrow, erythroid progenitor cells undergo several stages to generate reticulocytes that enter the bloodstream. Erythropoiesis is regulated by various factors, with erythropoietin (EPO) synthesized by the kidney being the promoting factor and hepcidin synthesized by the hepar inhibiting iron mobilization. Transcription factors, such as GATA and KLF, also play a crucial role in erythropoiesis. Disruption of any of these factors can lead to abnormal erythropoiesis, resulting in red cell excess, red cell deficiency, or abnormal morphological function. This review provides a general description of erythropoiesis, as well as its regulation, highlighting the significance of understanding the process for the diagnosis and treatment of various hematological disorders.
Collapse
Affiliation(s)
- Pu Tang
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| | - Huaquan Wang
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
10
|
Hoving V, Nijssen LE, Donker AE, Roelofs R, Schols SEM, Swinkels DW. Erythropoiesis-hepcidin-iron axis in patients with X-linked sideroblastic anaemia: An explorative biomarker study. Br J Haematol 2023; 202:1216-1219. [PMID: 37469032 DOI: 10.1111/bjh.18986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/28/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023]
Affiliation(s)
- Vera Hoving
- Department of Haematology, Radboud university medical center, Nijmegen, The Netherlands
| | - Lieke E Nijssen
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Department of Paediatrics, Máxima Medical Center, Veldhoven, The Netherlands
| | - Albertine E Donker
- Department of Paediatrics, Máxima Medical Center, Veldhoven, The Netherlands
| | - Rian Roelofs
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Saskia E M Schols
- Department of Haematology, Radboud university medical center, Nijmegen, The Netherlands
| | - Dorine W Swinkels
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Sanquin Blood Bank, Sanquin Diagnostics BV, Amsterdam, The Netherlands
| |
Collapse
|
11
|
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: 16.0] [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
|
12
|
Williams A, Bissinger R, Shamaa H, Patel S, Bourne L, Artunc F, Qadri SM. Pathophysiology of Red Blood Cell Dysfunction in Diabetes and Its Complications. PATHOPHYSIOLOGY 2023; 30:327-345. [PMID: 37606388 PMCID: PMC10443300 DOI: 10.3390/pathophysiology30030026] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 08/23/2023] Open
Abstract
Diabetes Mellitus (DM) is a complex metabolic disorder associated with multiple microvascular complications leading to nephropathy, retinopathy, and neuropathy. Mounting evidence suggests that red blood cell (RBC) alterations are both a cause and consequence of disturbances related to DM-associated complications. Importantly, a significant proportion of DM patients develop varying degrees of anemia of confounding etiology, leading to increased morbidity. In chronic hyperglycemia, RBCs display morphological, enzymatic, and biophysical changes, which in turn prime them for swift phagocytic clearance from circulation. A multitude of endogenous factors, such as oxidative and dicarbonyl stress, uremic toxins, extracellular hypertonicity, sorbitol accumulation, and deranged nitric oxide metabolism, have been implicated in pathological RBC changes in DM. This review collates clinical laboratory findings of changes in hematology indices in DM patients and discusses recent reports on the putative mechanisms underpinning shortened RBC survival and disturbed cell membrane architecture within the diabetic milieu. Specifically, RBC cell death signaling, RBC metabolism, procoagulant RBC phenotype, RBC-triggered endothelial cell dysfunction, and changes in RBC deformability and aggregation in the context of DM are discussed. Understanding the mechanisms of RBC alterations in DM provides valuable insights into the clinical significance of the crosstalk between RBCs and microangiopathy in DM.
Collapse
Affiliation(s)
- Alyssa Williams
- Faculty of Science, Ontario Tech University, Oshawa, ON L1G 0C5, Canada
- School of Biomedical Engineering, McMaster University, Hamilton, ON L8S 4M1, Canada
| | - Rosi Bissinger
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Hala Shamaa
- Faculty of Health Sciences, Ontario Tech University, Oshawa, ON L1G 0C5, Canada
| | - Shivani Patel
- Faculty of Health Sciences, Ontario Tech University, Oshawa, ON L1G 0C5, Canada
| | - Lavern Bourne
- Faculty of Health Sciences, Ontario Tech University, Oshawa, ON L1G 0C5, Canada
| | - Ferruh Artunc
- Division of Endocrinology, Diabetology and Nephrology, Department of Internal Medicine, University Hospital Tübingen, 72076 Tübingen, Germany
- Institute of Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- German Center for Diabetes Research at the University of Tübingen, 72076 Tübingen, Germany
| | - Syed M. Qadri
- Faculty of Health Sciences, Ontario Tech University, Oshawa, ON L1G 0C5, Canada
| |
Collapse
|
13
|
Cardona CJ, Montgomery MR. Iron regulatory proteins: players or pawns in ferroptosis and cancer? Front Mol Biosci 2023; 10:1229710. [PMID: 37457833 PMCID: PMC10340119 DOI: 10.3389/fmolb.2023.1229710] [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: 05/26/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
Cells require iron for essential functions like energy production and signaling. However, iron can also engage in free radical formation and promote cell proliferation thereby contributing to both tumor initiation and growth. Thus, the amount of iron within the body and in individual cells is tightly regulated. At the cellular level, iron homeostasis is maintained post-transcriptionally by iron regulatory proteins (IRPs). Ferroptosis is an iron-dependent form of programmed cell death with vast chemotherapeutic potential, yet while IRP-dependent targets have established roles in ferroptosis, our understanding of the contributions of IRPs themselves is still in its infancy. In this review, we present the growing circumstantial evidence suggesting that IRPs play critical roles in the adaptive response to ferroptosis and ferroptotic cell death and describe how this knowledge can be leveraged to target neoplastic iron dysregulation more effectively.
Collapse
|
14
|
Akiyama H, Carter BZ, Andreeff M, Ishizawa J. Molecular Mechanisms of Ferroptosis and Updates of Ferroptosis Studies in Cancers and Leukemia. Cells 2023; 12:1128. [PMID: 37190037 PMCID: PMC10136912 DOI: 10.3390/cells12081128] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/04/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
Ferroptosis is a mode of cell death regulated by iron-dependent lipid peroxidation. Growing evidence suggests ferroptosis induction as a novel anti-cancer modality that could potentially overcome therapy resistance in cancers. The molecular mechanisms involved in the regulation of ferroptosis are complex and highly dependent on context. Therefore, a comprehensive understanding of its execution and protection machinery in each tumor type is necessary for the implementation of this unique cell death mode to target individual cancers. Since most of the current evidence for ferroptosis regulation mechanisms is based on solid cancer studies, the knowledge of ferroptosis with regard to leukemia is largely lacking. In this review, we summarize the current understanding of ferroptosis-regulating mechanisms with respect to the metabolism of phospholipids and iron as well as major anti-oxidative pathways that protect cells from ferroptosis. We also highlight the diverse impact of p53, a master regulator of cell death and cellular metabolic processes, on the regulation of ferroptosis. Lastly, we discuss recent ferroptosis studies in leukemia and provide a future perspective for the development of promising anti-leukemia therapies implementing ferroptosis induction.
Collapse
Affiliation(s)
| | | | | | - Jo Ishizawa
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (H.A.); (B.Z.C.); (M.A.)
| |
Collapse
|
15
|
Zaman BA, Rasool SO, Abdo JM. The effect of erythroferrone suppression by transfusion on the erythropoietin–erythroferrone–hepcidin axis in transfusion‐dependent thalassaemia: A pre–post cohort study. Br J Haematol 2022; 201:547-551. [PMID: 36535905 DOI: 10.1111/bjh.18619] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
To track post-transfusion changes on the erythropoietin (EPO)-erythroferrone (ERFE)-hepcidin axis, we collected blood samples from 82 regularly transfused patients with β-thalassaemia major (β-TM) immediately before and 4-6 days after transfusion. The post-transfusion haemoglobin, hepcidin, and ferritin levels were increased, while the EPO, ERFE, and soluble transferrin receptor were suppressed. In addition, hepcidin change was inversely associated with erythropoietic change, which was confirmed by an increase in the hepcidin-to-ERFE ratio after transfusion. Age was the main predictor of serum ERFE, followed by EPO, transfusion frequencies, and ferritin. We found ERFE to be a highly sensitive indicator of erythroid activity in β-TM and that the hepcidin-to-ERFE ratio after transfusion may be used as an appropriateness index of serum hepcidin regulation relative to the degree of erythropoiesis.
Collapse
Affiliation(s)
- Burhan Abdullah Zaman
- Department of Basic Sciences, College of Pharmacy University of Duhok Duhok City Iraq
| | - Suzan Omer Rasool
- Department of clinical pharmacy, College of Pharmacy University of Duhok Duhok City Iraq
| | - Jassim Mohammed Abdo
- Department of Basic Sciences, College of Pharmacy University of Duhok Duhok City Iraq
| |
Collapse
|
16
|
Tabu K, Taga T. Cancer ego-system in glioma: an iron-replenishing niche network systemically self-organized by cancer stem cells. Inflamm Regen 2022; 42:54. [PMID: 36451253 PMCID: PMC9710158 DOI: 10.1186/s41232-022-00240-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022] Open
Abstract
For all living organisms, the adaptation to outside environments is an essential determinant to survive natural and artificial selections and to sustain the whole ecosystem intact with functional biodiversity. Likewise, cancer cells have similar characteristics that evade not only stresses from the host-internal innate and adaptive immune systems but also those from host-externally administered therapeutic interventions. Such selfish characteristics of cancer cells lead to the formation of cancerous ecosystem with a wide variety of phenotypic heterogeneity, which should be called cancer "egosystem" from the host point of view. Recently increasing evidence demonstrates that cancer stem cells (CSCs) are responsible for this cancer egosystem by effectively exploiting host inflammatory and hematopoietic cells and thereby reconstructing their own advantageous niches, which may well be a driving force in cancer recurrence. CSCs are further likely to render multiple niches mutually interconnected and cooperating as a network to support back CSCs themselves. Here, we summarize a recently identified iron-replenishing niche network self-organized by glioma CSCs (GSCs) through remote regulation of host myeloid and erythroid lineage cells. GSCs recruit bone marrow (BM)-derived inflammatory monocytes into tumor parenchyma, facilitate their differentiation into macrophages (Mφs) and skew their polarization into pro-tumoral phenotype, i.e., tumor-associated Mφs (TAMs). Meanwhile, GSCs distantly enhance erythropoiesis in host hematopoietic organs like BM and spleen potentially by secreting some soluble mediators that maintain continuous supply of erythrocytes within tumors. In addition, as normal red pulp Mφs (RPMs) under steady state conditions in spleen recycle iron by phagocytosing the aged or damaged erythrocytes (a/dECs) and release it in time of need, TAMs at least in gliomas phagocytose the hemorrhaged erythrocytes within tumors and potentially serve as a source of iron, an important nutrient indispensable to GSC survival and glioma progression. Taken together, these studies provide the substantial evidence that CSCs have a unique strategy to orchestrate multiple niches as an ecosystem that threatens the host living, which in this sense must be an egosystem. Targeting such an adaptive subpopulation of CSCs could achieve drastic disturbance of the CSC niches and subsequent extinction of malignant neoplasms.
Collapse
Affiliation(s)
- Kouichi Tabu
- grid.265073.50000 0001 1014 9130Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8510 Japan
| | - Tetsuya Taga
- grid.265073.50000 0001 1014 9130Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, 113-8510 Japan
| |
Collapse
|
17
|
Rieg CEH, Cattani D, Naspolini NF, Cenci VH, de Liz Oliveira Cavalli VL, Jacques AV, Nascimento MVPDS, Dalmarco EM, De Moraes ACR, Santos-Silva MC, Silva FRMB, Parisotto EB, Zamoner A. Perinatal exposure to a glyphosate pesticide formulation induces offspring liver damage. Toxicol Appl Pharmacol 2022; 454:116245. [PMID: 36116562 DOI: 10.1016/j.taap.2022.116245] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 11/28/2022]
Abstract
The present study investigated the effects of perinatal exposure to glyphosate-based herbicide (GBH) in offspring's liver. Pregnant Wistar rats were exposed to GBH (70 mg glyphosate/Kg body weight/day) in drinking water from gestation day 5 to postnatal day 15. The perinatal exposure to GBH increased 45Ca2+ influx in offspring's liver. Pharmacological tools indicated a role played by oxidative stress, phospholipase C (PLC) and Akt pathways, as well as voltage-dependent Ca2+ channel modulation on GBH-induced Ca2+ influx in offspring's liver. In addition, changes in the enzymatic antioxidant defense system, decreased GSH content, lipid peroxidation and protein carbonylation suggest a connection between GBH-induced hepatotoxic mechanism and redox imbalance. The perinatal exposure to GBH also increased the enzymatic activities of transaminases and gamma-glutamyl transferase in offspring's liver and blood, suggesting a pesticide-induced liver injury. Moreover, we detected increased iron levels in liver, blood and bone marrow of GBH-exposed rats, which were accompanied by increased transferrin saturation and decreased transferrin levels in blood. The levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) were increased in the liver of rats perinatally exposed to GBH, which were associated with. Increased phospho-p65NFκB immunocontent. Therefore, we propose that excessive amounts of iron in offspring's liver, blood and bone marrow induced by perinatal exposure to GBH may account for iron-driven hepatotoxicity, which was associated with Ca2+ influx, oxidative damage and inflammation. Further studies will clarify whether these events can ultimately impact on liver function.
Collapse
Affiliation(s)
- Carla Elise Heinz Rieg
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Daiane Cattani
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Nathalia Ferrazzo Naspolini
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Vitoria Hayduck Cenci
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | | | - Amanda Virtuoso Jacques
- Department of Clinical Analysis, Center of Health Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | | | - Eduardo Monguilhott Dalmarco
- Department of Clinical Analysis, Center of Health Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Ana Carolina Rabello De Moraes
- Department of Clinical Analysis, Center of Health Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Maria Cláudia Santos-Silva
- Department of Clinical Analysis, Center of Health Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Fátima Regina Mena Barreto Silva
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Eduardo Benedetti Parisotto
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Ariane Zamoner
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
18
|
Günther A, Hose M, Abberger H, Schumacher F, Veith Y, Kleuser B, Matuschewski K, Lang KS, Gulbins E, Buer J, Westendorf A, Hansen W. The acid ceramidase/ceramide axis controls parasitemia in Plasmodium yoelii-infected mice by regulating erythropoiesis. eLife 2022; 11:77975. [PMID: 36094170 PMCID: PMC9499531 DOI: 10.7554/elife.77975] [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: 02/17/2022] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
Acid ceramidase (Ac) is part of the sphingolipid metabolism and responsible for the degradation of ceramide. As bioactive molecule, ceramide is involved in the regulation of many cellular processes. However, the impact of cell-intrinsic Ac activity and ceramide on the course of Plasmodium infection remains elusive. Here, we use Ac-deficient mice with ubiquitously increased ceramide levels to elucidate the role of endogenous Ac activity in a murine malaria model. Interestingly, ablation of Ac leads to alleviated parasitemia associated with decreased T cell responses in the early phase of Plasmodium yoelii infection. Mechanistically, we identified dysregulated erythropoiesis with reduced numbers of reticulocytes, the preferred host cells of P. yoelii, in Ac-deficient mice. Furthermore, we demonstrate that administration of the Ac inhibitor carmofur to wildtype mice has similar effects on P. yoelii infection and erythropoiesis. Notably, therapeutic carmofur treatment after manifestation of P. yoelii infection is efficient in reducing parasitemia. Hence, our results provide evidence for the involvement of Ac and ceramide in controlling P. yoelii infection by regulating red blood cell development.
Collapse
Affiliation(s)
- Anne Günther
- Institute of Medical Microbiology, University of Duisburg-Essen, Essen, Germany
| | - Matthias Hose
- Institute of Medical Microbiology, University of Duisburg-Essen, Essen, Germany
| | - Hanna Abberger
- Institute of Medical Microbiology, University of Duisburg-Essen, Essen, Germany
| | | | - Ylva Veith
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Burkhard Kleuser
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Kai Matuschewski
- Institute of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Erich Gulbins
- Institute of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Jan Buer
- Institute of Medical Microbiology, University of Duisburg-Essen, Essen, Germany
| | - Astrid Westendorf
- Institute of Medical Microbiology, University of Duisburg-Essen, Duisburg, Germany
| | - Wiebke Hansen
- Institute of Medical Microbiology, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
19
|
Zaman BA, Ibrahim SA. Hepcidin-to-Ferritin Ratio as an Early Diagnostic Index of Iron Overload in β-Thalassemia Major Patients. Hemoglobin 2022; 46:106-113. [PMID: 35930276 DOI: 10.1080/03630269.2022.2083969] [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: 01/19/2023]
Abstract
Hepcidin (HEPC) hormone production is expected to be elevated in cases accompanying iron overload, but the opposite impact of ineffective erythropoiesis in β-thalassemia major (β-TM) patients overrides this effect. The role of the HEPC-to-ferritin (FER) ratio and its components in iron metabolism along with their diagnostic cutoff values, sensitivity, specificity, and accuracy in β-TM patients with iron overload, were examined in this study. This was a 1:1 case-control study with 120 participants, ages ranging from 2 to 30 years of both sexes, who were assigned into two groups: 60 β-TM patients with iron overload, and a control group, comprising 60 healthy individuals matched by gender and age. In the present study, we found slightly elevated serum HEPC concentration (21.9 ng/mL) compared to the controls (9.9 ng/mL), which was not statistically significant (p =0.1), and the median HEPC-to-FER ratio of the cases was significantly lower than the controls, with the median case-control difference of (-0.366; p < 0.001). Our results revealed a statistically significant impact (p < 0.001) of mean age on the serum HEPC level with the inverse linear correlation of (-0.487, p < 0.001). The area under the curve of the HEPC-to-FER ratio was 0.999 and the optimum cutoff value was 0.046 ng/mL (p < 0.001) with 100.0% sensitivity and 98.3% specificity. In conclusion, we found that serum HEPC-to-FER ratio, with an accuracy of 99.2%, may serve as an excellent index for the diagnosis of iron overload in β-TM patients differentiating them from nonthalassemic controls.
Collapse
Affiliation(s)
- Burhan A Zaman
- Department of Basic Sciences, College of Pharmacy, University of Duhok, Duhok City, Iraq
| | - Shereen A Ibrahim
- Department of Physiology and Pharmacology, College of Medicine, University of Duhok, Duhok City, Iraq
| |
Collapse
|
20
|
Man Y, Lu Z, Yao X, Gong Y, Yang T, Wang Y. Recent Advancements in Poor Graft Function Following Hematopoietic Stem Cell Transplantation. Front Immunol 2022; 13:911174. [PMID: 35720412 PMCID: PMC9202575 DOI: 10.3389/fimmu.2022.911174] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/06/2022] [Indexed: 01/05/2023] Open
Abstract
Poor graft function (PGF) is a life-threatening complication that occurs after transplantation and has a poor prognosis. With the rapid development of haploidentical hematopoietic stem cell transplantation, the pathogenesis of PGF has become an important issue. Studies of the pathogenesis of PGF have resulted in some success in CD34+-selected stem cell boosting. Mesenchymal stem cells, N-acetyl-l-cysteine, and eltrombopag have also been investigated as therapeutic strategies for PGF. However, predicting and preventing PGF remains challenging. Here, we propose that the seed, soil, and insect theories of aplastic anemia also apply to PGF; CD34+ cells are compared to seeds; the bone marrow microenvironment to soil; and virus infection, iron overload, and donor-specific anti-human leukocyte antigen antibodies to insects. From this perspective, we summarize the available information on the common risk factors of PGF, focusing on its potential mechanism. In addition, the safety and efficacy of new strategies for treating PGF are discussed to provide a foundation for preventing and treating this complex clinical problem.
Collapse
Affiliation(s)
- Yan Man
- Department of Hematology, National Key Clinical Specialty of Hematology, Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Zhixiang Lu
- Department of Hematology, National Key Clinical Specialty of Hematology, Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Xiangmei Yao
- Department of Hematology, National Key Clinical Specialty of Hematology, Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Yuemin Gong
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Tonghua Yang
- Department of Hematology, National Key Clinical Specialty of Hematology, Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, The First People’s Hospital of Yunnan Province, Kunming, China,*Correspondence: Tonghua Yang, ; Yajie Wang,
| | - Yajie Wang
- Department of Hematology, National Key Clinical Specialty of Hematology, Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, The First People’s Hospital of Yunnan Province, Kunming, China,*Correspondence: Tonghua Yang, ; Yajie Wang,
| |
Collapse
|
21
|
Correnti M, Gammella E, Cairo G, Recalcati S. Iron Mining for Erythropoiesis. Int J Mol Sci 2022; 23:ijms23105341. [PMID: 35628152 PMCID: PMC9140467 DOI: 10.3390/ijms23105341] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 01/27/2023] Open
Abstract
Iron is necessary for essential processes in every cell of the body, but the erythropoietic compartment is a privileged iron consumer. In fact, as a necessary component of hemoglobin and myoglobin, iron assures oxygen distribution; therefore, a considerable amount of iron is required daily for hemoglobin synthesis and erythroid cell proliferation. Therefore, a tight link exists between iron metabolism and erythropoiesis. The liver-derived hormone hepcidin, which controls iron homeostasis via its interaction with the iron exporter ferroportin, coordinates erythropoietic activity and iron homeostasis. When erythropoiesis is enhanced, iron availability to the erythron is mainly ensured by inhibiting hepcidin expression, thereby increasing ferroportin-mediated iron export from both duodenal absorptive cells and reticuloendothelial cells that process old and/or damaged red blood cells. Erythroferrone, a factor produced and secreted by erythroid precursors in response to erythropoietin, has been identified and characterized as a suppressor of hepcidin synthesis to allow iron mobilization and facilitate erythropoiesis.
Collapse
|
22
|
Stephenson D, Nemkov T, Qadri SM, Sheffield WP, D’Alessandro A. Inductively-Coupled Plasma Mass Spectrometry-Novel Insights From an Old Technology Into Stressed Red Blood Cell Physiology. Front Physiol 2022; 13:828087. [PMID: 35197866 PMCID: PMC8859330 DOI: 10.3389/fphys.2022.828087] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/17/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Ion and metal homeostasis are critical to red blood cell physiology and Inductively Coupled Plasma (ICP) is a decades old approach to pursue elemental analysis. Recent evolution of ICP has resulted in its coupling to mass spectrometry (MS) instead of atomic absorption/emission. METHODS Here we performed Inductively-coupled plasma mass spectrometry (ICP-MS) measurements of intra- and extra-cellular Na, K, Ca, Mg, Fe, and Cu in red blood cells undergoing ionic, heat, or starvation stress. Results were correlated with Ca measurements from other common platforms (e.g., fluorescence-based approaches) and extensive measurements of red blood cell metabolism. RESULTS All stresses induced significant intra- and extracellular alterations of all measured elements. In particular, ionomycin treatment or hypertonic stress significantly impacted intracellular sodium and extracellular potassium and magnesium levels. Iron efflux was observed as a function of temperatures, with ionic and heat stress at 40°C causing the maximum decrease in intracellular iron pools and increases in the supernatants. Strong positive correlation was observed between calcium measurements via ICP-MS and fluorescence-based approaches. Correlation analyses with metabolomics data showed a strong positive association between extracellular calcium and intracellular sodium or magnesium levels and intracellular glycolysis. Extracellular potassium or iron were positively correlated with free fatty acids (especially mono-, poly-, and highly-unsaturated or odd-chain fatty acid products of lipid peroxidation). Intracellular iron was instead positively correlated with saturated fatty acids (palmitate, stearate) and negatively with methionine metabolism (methionine, S-adenosylmethionine), phosphatidylserine exposure and glycolysis. CONCLUSION In the era of omics approaches, ICP-MS affords a comprehensive characterization of intracellular elements that provide direct insights on red blood cell physiology and represent meaningful covariates for data generated via other omics platforms such as metabolomics.
Collapse
Affiliation(s)
- Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver–Anschutz Medical Campus, Aurora, CO, United States
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver–Anschutz Medical Campus, Aurora, CO, United States
| | - Syed M. Qadri
- Faculty of Health Sciences, Ontario Tech University, Oshawa, ON, Canada
| | - William P. Sheffield
- Centre for Innovation, Canadian Blood Services, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver–Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
23
|
Zeidan RS, Han SM, Leeuwenburgh C, Xiao R. Iron homeostasis and organismal aging. Ageing Res Rev 2021; 72:101510. [PMID: 34767974 DOI: 10.1016/j.arr.2021.101510] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/21/2022]
Abstract
Iron is indispensable for normal body functions across species because of its critical roles in red blood cell function and many essential proteins and enzymes required for numerous physiological processes. Regulation of iron homeostasis is an intricate process involving multiple modulators at the systemic, cellular, and molecular levels. Interestingly, emerging evidence has demonstrated that many modulators of iron homeostasis contribute to organismal aging and longevity. On the other hand, the age-related dysregulation of iron homeostasis is often associated with multiple age-related pathologies including bone resorption and neurodegenerative diseases such as Alzheimer's disease. Thus, a thorough understanding on the interconnections between systemic and cellular iron balance and organismal aging may help decipher the etiologies of multiple age-related diseases, which could ultimately lead to developing therapeutic strategies to delay aging and treat various age-related diseases. Here we present the current understanding on the mechanisms of iron homeostasis. We also discuss the impacts of aging on iron homeostatic processes and how dysregulated iron metabolism may affect aging and organismal longevity.
Collapse
|
24
|
Aimaitijiang A, Tabu K, Wang W, Nobuhisa I, Taga T. Glioma cells remotely promote erythropoiesis as a self-expanding strategy of cancer stem cells. Genes Cells 2021; 27:25-42. [PMID: 34837452 DOI: 10.1111/gtc.12908] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 11/18/2021] [Indexed: 11/27/2022]
Abstract
Cancer stem cells are a promising target for cancer eradication due to their responsibility for therapy-resistance and cancer recurrence. Previously, we have demonstrated that glioma stem cells (GSCs) recruit and induce the differentiation of bone marrow (BM) monocytes into tumor-infiltrating macrophages, which phagocytose hemorrhaged erythrocytes and store GSC-beneficial iron in mouse xenografts, suggesting a self-expanding strategy of GSCs that exploits host hematopoiesis of myeloid cells. However, it remains unclear whether a self-advantageous effect of GSCs also occurs on erythroid cells during glioma development. Here, we found that, in the primary cultures of mouse fetal liver proerythroblasts (proEs), conditioned media prepared from glioma cells including patient-derived glioblastoma (GBM) cells significantly facilitated the differentiation of proEs into erythroblasts. Importantly, in-vivo erythroid analysis in intracranially GSC-transplanted mice showed an enhanced erythropoiesis in the BM. In addition, the sphere forming ability of patient-derived GBM cells was significantly suppressed by hypoxia treatment and iron chelation, suggesting higher demands of GSCs for oxygen and iron, which may be supplied by GSCs- and their progeny-induced erythrocyte production. Our findings provide a new insight into survival and expanding strategies of GSCs that systemically exploit host erythropoiesis.
Collapse
Affiliation(s)
- Alapati Aimaitijiang
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kouichi Tabu
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Wenqian Wang
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Ikuo Nobuhisa
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tetsuya Taga
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
25
|
Diepeveen L, Roelofs R, Grebenchtchikov N, van Swelm R, Kautz L, Swinkels D. Differentiating iron-loading anemias using a newly developed and analytically validated ELISA for human serum erythroferrone. PLoS One 2021; 16:e0254851. [PMID: 34283879 PMCID: PMC8291690 DOI: 10.1371/journal.pone.0254851] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/03/2021] [Indexed: 12/26/2022] Open
Abstract
Erythroferrone (ERFE), the erythroid regulator of iron metabolism, inhibits hepcidin to increase iron availability for erythropoiesis. ERFE plays a pathological role during ineffective erythropoiesis as occurs in X-linked sideroblastic anemia (XLSA) and β-thalassemia. Its measurement might serve as an indicator of severity for these diseases. However, for reliable quantification of ERFE analytical characterization is indispensable to determine the assay’s limitations and define proper methodology. We developed a sandwich ELISA for human serum ERFE using polyclonal antibodies and report its extensive analytical validation. This new assay showed, for the first time, the differentiation of XLSA and β-thalassemia major patients from healthy controls (p = 0.03) and from each other (p<0.01), showing the assay provides biological plausible results. Despite poor dilution linearity, parallelism and recovery in patient serum matrix, which indicated presence of a matrix effect and/or different immunoreactivity of the antibodies to the recombinant standard and the endogenous analyte, our assay correlated well with two other existing ERFE ELISAs (both R2 = 0.83). Nevertheless, employment of one optimal dilution of all serum samples is warranted to obtain reliable results. When adequately performed, the assay can be used to further unravel the human erythropoiesis-hepcidin-iron axis in various disorders and assess the added diagnostic value of ERFE.
Collapse
Affiliation(s)
- Laura Diepeveen
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- * E-mail:
| | - Rian Roelofs
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nicolai Grebenchtchikov
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rachel van Swelm
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leon Kautz
- Institut de Recherche en Santé Digestive (IRSD), Université de Toulouse, INSERM U1220, Institut National de la Recherche Agronomique (INRA) U1416, École Nationale Vétérinaire de Toulouse (ENVT), Université Paul Sabatier (UPS), Toulouse, France
| | - Dorine Swinkels
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
26
|
The critical roles of iron during the journey from fetus to adolescent: Developmental aspects of iron homeostasis. Blood Rev 2021; 50:100866. [PMID: 34284901 DOI: 10.1016/j.blre.2021.100866] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/12/2022]
Abstract
Iron is indispensable for human life. However, it is also potentially toxic, since it catalyzes the formation of harmful oxidative radicals in unbound form and may facilitate pathogen growth. Therefore, iron homeostasis needs to be tightly regulated. Rapid growth and development require large amounts of iron, while (especially young) children are vulnerable to infections with iron-dependent pathogens due to an immature immune system. Moreover, unbalanced iron status early in life may have effects on the nervous system, immune system and gut microbiota that persist into adulthood. In this narrative review, we assess the critical roles of iron for growth and development and elaborate how the body adapts to physiologically high iron demands during the journey from fetus to adolescent. As a first step towards the development of clinical guidelines for the management of iron disorders in children, we summarize the unmet needs regarding the developmental aspects of iron homeostasis.
Collapse
|
27
|
Zhang J, Tang K, Zhang Y, Ma Y, Zhang C, Hu H, Jia X, Zhuang R, Jin B, Wang M, Zhang X, Liu D, Zhang Y. The Presence of Circulating Nucleated Red Blood Cells Is Associated With Disease Severity in Patients of Hemorrhagic Fever With Renal Syndrome. Front Med (Lausanne) 2021; 8:665410. [PMID: 34113638 PMCID: PMC8186265 DOI: 10.3389/fmed.2021.665410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/19/2021] [Indexed: 12/31/2022] Open
Abstract
Hemorrhagic fever with renal syndrome (HFRS) is a regional infectious disease of epidemic potential caused by the Hantaan virus (HTNV). Red blood cells (RBCs) are the major components of peripheral blood. However, pathological changes in RBCs and the underlying mechanisms during HTNV infection remain largely unclear. Therefore, this study sought to explore changes in RBCs in the peripheral blood of HFRS patients. We isolated PBMCs from HFRS patients and performed single-cell RNA sequencing. The results showed that clusters of RBCs in the peripheral blood of HFRS could be classified as nucleated red blood cells (NRBC) based on their cellular components, gene expression profiles and cell surface markers. In addition, it was shown that the higher the count of NRBC in peripheral blood, the more severe the disease status was. Moreover, hematological indices related to RBCs were analyzed and the results showed that impairment in the folate pathway might be the possible reason behind the presence of NRBCs. This study, for the first time showed that the presence of NRBCs in the peripheral blood of HFRS patients was associated with disease severity. This was also the first study to show that infection with the HTNV virus hindered the maturation of RBCs. Therefore, this work provides further insights on the role of and pathological changes in RBCs during HTNV infection.
Collapse
Affiliation(s)
- Jingang Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China.,Brigade of Cadet, Air Force Medical University, Xi'an, China
| | - Kang Tang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Yun Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Ying Ma
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Chunmei Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Haifeng Hu
- Center for Infectious Diseases, Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Xiaozhou Jia
- Department of Infectious Disease, Xi'an Eighth Hospital, Xi'an, China
| | - Ran Zhuang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Boquan Jin
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Meng Wang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Xiyue Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| | - Dalu Liu
- Department of Radiation Medicine and Protection, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi'an, China
| | - Yusi Zhang
- Department of Immunology, Basic Medicine School, Air Force Medical University, Xi'an, China
| |
Collapse
|
28
|
Iron Status in Newly Diagnosed β-Thalassemia Major: High Rate of Iron Status due to Erythropoiesis Drive. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5560319. [PMID: 33954177 PMCID: PMC8068532 DOI: 10.1155/2021/5560319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/23/2021] [Accepted: 04/10/2021] [Indexed: 11/25/2022]
Abstract
Background Iron overload in severe β-thalassemia is a serious complication that occurs during the course of the disease. Information about the iron status during initial illness with β-thalassemia major seemed to be limited. This study is aimed at analyzing iron status, serum hepcidin, and growth differentiation factor 15 (GDF15) levels in newly diagnosed β-thalassemia major. Methods A case-control study was performed at Dr. Hasan Sadikin General Hospital, which included 41 children with newly diagnosed β-thalassemia major. Age- and sex-matched controls were enrolled. The subjects had no blood transfusion, had normal liver function, and had no sign of inflammation. The groups were compared in terms of the levels of hemoglobin (Hb), serum ferritin (SF), transferrin saturation (TS), serum hepcidin, and GDF15 as iron homeostasis parameters. Results Of the 41 newly diagnosed β-thalassemia major patients, those who were less than 24 months old had significantly lower median Hb levels than controls (5.0 vs. 11.7 g/dL, P < 0.001). The median SF and TS levels were significantly higher than those in controls (315.0 vs. 29.0 ng/mL, P < 0.001; 70.6 vs. 16.5%, P < 0.001), and median hepcidin was at the normal limit, but the value was higher in patients (251.0 vs. 123.1 ng/mL, P < 0.001). The median GDF15 level was significantly higher in patients (2,095.3 vs. 342.4 pg/mL, P < 0.001). There was a positive correlation between SF-TS, SF-hepcidin, TS-hepcidin, SF-GDF15, TS-GDF15, and hepcidin-GDF15 (P < 0.001). Conclusion In newly diagnosed β-thalassemia major, an increase in iron status occurred. This may be caused by increased iron absorption due to massive erythropoietic activity, characterized by an increase in GDF15 levels, which does not cause hepcidin suppression. The iron homeostasis response seems to be physiologically indicated by a tendency to increase hepcidin levels.
Collapse
|
29
|
Han Y, Huang W, Meng H, Zhan Y, Hou J. Pro-inflammatory cytokine interleukin-6-induced hepcidin, a key mediator of periodontitis-related anemia of inflammation. J Periodontal Res 2021; 56:690-701. [PMID: 33656216 DOI: 10.1111/jre.12865] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/05/2021] [Accepted: 02/06/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To investigate whether anemia of inflammation (AI) occurs in periodontitis patients and to further explore underlying pathogenesis of periodontitis-related AI by an experimental periodontitis model. BACKGROUND Previous studies have reported periodontitis patients could show a tendency toward AI. However, the relationship between periodontitis and AI remains unclear, and the related pathological mechanisms have not been identified. MATERIALS AND METHODS Periodontal clinical parameters, inflammatory markers, and anemia-related indicators were compared between 98 aggressive periodontitis (AgP) patients and 103 healthy subjects. An experimental periodontitis model was induced by ligature placement in mice. The changes in mice inflammatory markers, anemia indicators, hepcidin mRNA expression, and serum hepcidin concentrations were measured. Human and mouse liver cells were treated with interleukin-6 (IL-6) for analyzing the changes in hepcidin expression based on mRNA and protein levels. RESULTS AgP patients exhibited higher white blood cell counts, IL-6, and C-reactive protein. Adjusted linear regression analyses showed correlations between AgP and decreased hemoglobin (HGB) and hematocrit (HCT). The ligature-induced periodontitis caused systemic inflammation and elevated IL-6 levels. Lower red blood cell counts, HGB, and HCT were detected, whereas the levels of hepcidin mRNA expression and serum hepcidin concentrations increased. The treatment of hepatocytes with IL-6 induced both hepcidin mRNA expression and hepcidin secretion. CONCLUSIONS Systemic inflammation induced by periodontitis leads to an increased risk for AI. IL-6-induced hepcidin could play a central mediator role and act as a key pathologic mechanism. Our results demonstrate periodontitis may be considered as an additional inflammatory disease contributing to the development of AI.
Collapse
Affiliation(s)
- Ye Han
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Wenxue Huang
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Huanxin Meng
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yalin Zhan
- First Clinical Division, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Jianxia Hou
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
30
|
Gonzalez-Menendez P, Romano M, Yan H, Deshmukh R, Papoin J, Oburoglu L, Daumur M, Dumé AS, Phadke I, Mongellaz C, Qu X, Bories PN, Fontenay M, An X, Dardalhon V, Sitbon M, Zimmermann VS, Gallagher PG, Tardito S, Blanc L, Mohandas N, Taylor N, Kinet S. An IDH1-vitamin C crosstalk drives human erythroid development by inhibiting pro-oxidant mitochondrial metabolism. Cell Rep 2021; 34:108723. [PMID: 33535038 PMCID: PMC9169698 DOI: 10.1016/j.celrep.2021.108723] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/26/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
The metabolic changes controlling the stepwise differentiation of hematopoietic stem and progenitor cells (HSPCs) to mature erythrocytes are poorly understood. Here, we show that HSPC development to an erythroid-committed proerythroblast results in augmented glutaminolysis, generating alpha-ketoglutarate (αKG) and driving mitochondrial oxidative phosphorylation (OXPHOS). However, sequential late-stage erythropoiesis is dependent on decreasing αKG-driven OXPHOS, and we find that isocitrate dehydrogenase 1 (IDH1) plays a central role in this process. IDH1 downregulation augments mitochondrial oxidation of αKG and inhibits reticulocyte generation. Furthermore, IDH1 knockdown results in the generation of multinucleated erythroblasts, a morphological abnormality characteristic of myelodysplastic syndrome and congenital dyserythropoietic anemia. We identify vitamin C homeostasis as a critical regulator of ineffective erythropoiesis; oxidized ascorbate increases mitochondrial superoxide and significantly exacerbates the abnormal erythroblast phenotype of IDH1-downregulated progenitors, whereas vitamin C, scavenging reactive oxygen species (ROS) and reprogramming mitochondrial metabolism, rescues erythropoiesis. Thus, an IDH1-vitamin C crosstalk controls terminal steps of human erythroid differentiation.
Collapse
Affiliation(s)
- Pedro Gonzalez-Menendez
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France.
| | - Manuela Romano
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Hongxia Yan
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; New York Blood Center, New York, NY, USA
| | - Ruhi Deshmukh
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Julien Papoin
- The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Leal Oburoglu
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Marie Daumur
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Anne-Sophie Dumé
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Ira Phadke
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France; Pediatric Oncology Branch, NCI, CCR, NIH, Bethesda, MD, USA
| | - Cédric Mongellaz
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Xiaoli Qu
- New York Blood Center, New York, NY, USA
| | - Phuong-Nhi Bories
- Service d'Hématologie Biologique, Assistance Publique-Hôpitaux de Paris, Institut Cochin, Paris, France
| | - Michaela Fontenay
- Laboratory of Excellence GR-Ex, Paris 75015, France; Service d'Hématologie Biologique, Assistance Publique-Hôpitaux de Paris, Institut Cochin, Paris, France
| | - Xiuli An
- New York Blood Center, New York, NY, USA
| | - Valérie Dardalhon
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Marc Sitbon
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Valérie S Zimmermann
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France
| | - Patrick G Gallagher
- Departments of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Saverio Tardito
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Lionel Blanc
- The Feinstein Institute for Medical Research, Manhasset, NY, USA
| | | | - Naomi Taylor
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France; Pediatric Oncology Branch, NCI, CCR, NIH, Bethesda, MD, USA.
| | - Sandrina Kinet
- Institut de Génétique Moléculaire de Montpellier, Univ. Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Paris 75015, France.
| |
Collapse
|
31
|
Kobayashi Y, Tanizawa M, Ogata M, Aoi W, Kuwahata M. Changes in iron metabolism centered on hepcidin due to high-intensity exercise under restricted food intake. Nutrition 2021; 86:111179. [PMID: 33690026 DOI: 10.1016/j.nut.2021.111179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/03/2021] [Accepted: 01/21/2021] [Indexed: 01/24/2023]
Abstract
OBJECTIVES The purpose of this study was to observe changes in iron metabolism of rats due to high-intensity exercise during reduced food intake. METHODS Thirty-six, 4-wk-old male Wistar rats were divided into six groups after being acclimated to running: rested control, intense exercise, 80% feeding and rested, 80% feeding and intense exercise, 70% feeding diet and rested, and 70% feeding and intense exercise groups. Only three intense exercise groups underwent acute running sessions for 30 min (30 m/min) once a day for 7 d. RESULTS Rats did not develop anemia with exercise and food intake restriction. Serum iron levels, transferrin saturation, hepcidin-25, and hepatic iron levels in the exercise groups were lower than those in the respective resting groups. However, these values in the 70% diet groups were slightly higher than those in the 80% diet groups. Serum erythropoietin levels decreased as food intake decreased, but the serum erythropoietin level in the 70% feeding and intense exercise group was similar to that in the rested control group. Serum interleukin-6 levels were significantly lower in the groups with restricted food intake than in the free-fed group, and exercise had no effect. CONCLUSIONS High-intensity exercise under restricted food intake may significantly alter the iron metabolism to maintain blood cell levels due to the strong promotion of metabolic iron utilization in response to dietary iron depletion, increased iron demand, and excretion.
Collapse
Affiliation(s)
- Yukiko Kobayashi
- Department of Nutrition Science, Graduate school of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan.
| | - Midori Tanizawa
- Department of Nutrition Science, Graduate school of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Midori Ogata
- Department of Nutrition Science, Graduate school of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Wataru Aoi
- Department of Nutrition Science, Graduate school of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Masashi Kuwahata
- Department of Nutrition Science, Graduate school of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| |
Collapse
|
32
|
Liu X, Hu J, Hu XR, Li XX, Guan DR, Liu JQ, Zhang YL, Zhang FK. [Expression of iron-regulating erythroid factors in different types of erythropoiesis disorders]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2021; 42:52-57. [PMID: 33677869 PMCID: PMC7957252 DOI: 10.3760/cma.j.issn.0253-2727.2021.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Indexed: 11/29/2022]
Abstract
Objective: To investigate the expression of iron-regulating erythroid factors in different types of erythropoiesis disorders. Methods: From January 2016 to November 2019, the plasma concentrations of iron-regulating erythroid factors were measured by ELISA methods in 47 patients with different types of erythropoiesis disorders. The adaptation orientation of iron-regulating erythroid factor expression with bone marrow erythropoiesis activities (represented by bone marrow-nucleated erythrocytes ratio) was analyzed. Results: The median plasma growth differentiation factor (GDF) 15 levels in patients with polycythemia vera (PV) , pure red cell aplasia (PRCA) , autoimmune hemolytic anemia (AIHA) , and myelodysplastic syndrome (MDS) were 266.01 ng/L (112.40, 452.37) , 110.63 ng/L (81.41, 220.42) , 52.11 ng/L (32.61, 171.66) , and 276.53 (132.16, 525.70) ng/L, respectively, which were significantly higher than those in normal patients with 37.45 (19.65, 57.72) ng/L (all P < 0.01) . The plasma TWSG1 expression levels were not significantly different in patients with PV, PRCA, AIHA, and MDS from those of normal patients (P>0.05) . The median plasma GDF11 level in PV was 74.75 (10.95, 121.32) ng/L, which was significantly higher than 36.90 (3.38, 98.34) ng/L in normal control subjects (P<0.01) . However, no statistical differences were observed in the other three subjects (P>0.05) . The median plasma erythroferrone (ERFE) levels in AIHA and PV were 121.76 ng/L (68.12, 343.11) and 129.63 (47.02, 170.03) ng/L, respectively, with the highest level in AIHA in all the studied types of erythropoiesis disorders. The bone marrow-nucleated erythrocytes ratio was significantly and positively correlated with ERFE (r=0.458, P=0.001) but not with GDF15 (r=-0.163, P=0.274) , GDF11 (r=0.120, P=0.421) , and TWSG1 (r=-0.166, P=0.269) . Conclusion: The expression profile of iron-regulating erythroid factors is not exactly the same in different types of erythropoiesis disorders. ERFE demonstrated the highest correlation with erythropoiesis activities.
Collapse
Affiliation(s)
- X Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - J Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - X R Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - X X Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - D R Guan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - J Q Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Y L Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - F K Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
33
|
Lira Zidanes A, Marchi G, Busti F, Marchetto A, Fermo E, Giorgetti A, Vianello A, Castagna A, Olivieri O, Bianchi P, Girelli D. A Novel ALAS2 Missense Mutation in Two Brothers With Iron Overload and Associated Alterations in Serum Hepcidin/Erythroferrone Levels. Front Physiol 2020; 11:581386. [PMID: 33281618 PMCID: PMC7689258 DOI: 10.3389/fphys.2020.581386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/01/2020] [Indexed: 01/19/2023] Open
Abstract
Iron loading anemias are characterized by ineffective erythropoiesis and iron overload. The prototype is non-transfusion dependent ß-thalassemia (NTDT), with other entities including congenital sideroblastic anemias, congenital dyserythropoietic anemias, some hemolytic anemias, and myelodysplastic syndromes. Differential diagnosis of iron loading anemias may be challenging due to heterogeneous genotype and phenotype. Notwithstanding the recent advances in linking ineffective erythropoiesis to iron overload, many pathophysiologic aspects are still unclear. Moreover, measurement of hepcidin and erythroferrone (ERFE), two key molecules in iron homeostasis and erythropoiesis, is scarcely used in clinical practice and of uncertain utility. Here, we describe a comprehensive diagnostic approach, including next-generation sequencing (NGS), in silico modeling, and measurement of hepcidin and erythroferrone (ERFE), in two brothers eventually diagnosed as X-linked sideroblastic anemia (XLSA). A novel pathogenic ALAS2 missense mutation (c.1382T>A, p.Leu461His) is described. Hyperferritinemia with high hepcidin-25 levels (but decreased hepcidin:ferritin ratio) and mild-to-moderate iron overload were detected in both patients. ERFE levels were markedly elevated in both patients, especially in the proband, who had a more expressed phenotype. Our study illustrates how new technologies, such as NGS, in silico modeling, and measurement of serum hepcidin-25 and ERFE, may help in diagnosing and studying iron loading anemias. Further studies on the hepcidin-25/ERFE axis in additional patients with XLSA and other iron loading anemias may help in establishing its usefulness in differential diagnosis, and it may also aid our understanding of the pathophysiology of these genetically and phenotypically heterogeneous entities.
Collapse
Affiliation(s)
- Acaynne Lira Zidanes
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy.,EuroBloodNet Referral Center for Rare Disorders of Iron Metabolism, University Hospital of Verona, Verona, Italy
| | - Giacomo Marchi
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy.,EuroBloodNet Referral Center for Rare Disorders of Iron Metabolism, University Hospital of Verona, Verona, Italy
| | - Fabiana Busti
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy.,EuroBloodNet Referral Center for Rare Disorders of Iron Metabolism, University Hospital of Verona, Verona, Italy
| | | | - Elisa Fermo
- Hematology and Pathophysiology of Anemias Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) Ca' Granda Foundation, Policlinico Milano, Milan, Italy
| | | | - Alice Vianello
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy.,EuroBloodNet Referral Center for Rare Disorders of Iron Metabolism, University Hospital of Verona, Verona, Italy
| | - Annalisa Castagna
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy.,EuroBloodNet Referral Center for Rare Disorders of Iron Metabolism, University Hospital of Verona, Verona, Italy
| | - Oliviero Olivieri
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy.,EuroBloodNet Referral Center for Rare Disorders of Iron Metabolism, University Hospital of Verona, Verona, Italy
| | - Paola Bianchi
- Hematology and Pathophysiology of Anemias Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) Ca' Granda Foundation, Policlinico Milano, Milan, Italy
| | - Domenico Girelli
- Section of Internal Medicine, Department of Medicine, University of Verona, Verona, Italy.,EuroBloodNet Referral Center for Rare Disorders of Iron Metabolism, University Hospital of Verona, Verona, Italy
| |
Collapse
|
34
|
Gräfe C, Müller EK, Gresing L, Weidner A, Radon P, Friedrich RP, Alexiou C, Wiekhorst F, Dutz S, Clement JH. Magnetic hybrid materials interact with biological matrices. PHYSICAL SCIENCES REVIEWS 2020. [DOI: 10.1515/psr-2019-0114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Abstract
Magnetic hybrid materials are a promising group of substances. Their interaction with matrices is challenging with regard to the underlying physical and chemical mechanisms. But thinking matrices as biological membranes or even structured cell layers they become interesting with regard to potential biomedical applications. Therefore, we established in vitro blood-organ barrier models to study the interaction and processing of superparamagnetic iron oxide nanoparticles (SPIONs) with these cellular structures in the presence of a magnetic field gradient. A one-cell-type–based blood-brain barrier model was used to investigate the attachment and uptake mechanisms of differentially charged magnetic hybrid materials. Inhibition of clathrin-dependent endocytosis and F-actin depolymerization led to a dramatic reduction of cellular uptake. Furthermore, the subsequent transportation of SPIONs through the barrier and the ability to detect these particles was of interest. Negatively charged SPIONs could be detected behind the barrier as well as in a reporter cell line. These observations could be confirmed with a two-cell-type–based blood-placenta barrier model. While positively charged SPIONs heavily interact with the apical cell layer, neutrally charged SPIONs showed a retarded interaction behavior. Behind the blood-placenta barrier, negatively charged SPIONs could be clearly detected. Finally, the transfer of the in vitro blood-placenta model in a microfluidic biochip allows the integration of shear stress into the system. Even without particle accumulation in a magnetic field gradient, the negatively charged SPIONs were detectable behind the barrier. In conclusion, in vitro blood-organ barrier models allow the broad investigation of magnetic hybrid materials with regard to biocompatibility, cell interaction, and transfer through cell layers on their way to biomedical application.
Collapse
Affiliation(s)
- Christine Gräfe
- Department of Internal Medicine II, Hematology and Medical Oncology , Jena University Hospital , Jena , Germany
| | - Elena K. Müller
- Department of Internal Medicine II, Hematology and Medical Oncology , Jena University Hospital , Jena , Germany
| | - Lennart Gresing
- Department of Internal Medicine II, Hematology and Medical Oncology , Jena University Hospital , Jena , Germany
| | - Andreas Weidner
- Institute of Biomedical Engineering and Informatics (BMTI), Technische Universität Ilmenau , Ilmenau , Germany
| | - Patricia Radon
- Physikalisch-Technische Bundesanstalt , Berlin , Germany
| | - Ralf P. Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON) , Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen , Erlangen , Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON) , Else Kröner-Fresenius-Stiftung-Professorship, Universitätsklinikum Erlangen , Erlangen , Germany
| | | | - Silvio Dutz
- Institute of Biomedical Engineering and Informatics (BMTI), Technische Universität Ilmenau , Ilmenau , Germany
| | - Joachim H. Clement
- Department of Internal Medicine II, Hematology and Medical Oncology , Jena University Hospital , Jena , Germany
| |
Collapse
|
35
|
Gaharwar US, Kumar S, Rajamani P. Iron oxide nanoparticle-induced hematopoietic and immunological response in rats. RSC Adv 2020; 10:35753-35764. [PMID: 35517102 PMCID: PMC9056920 DOI: 10.1039/d0ra05901c] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/14/2020] [Indexed: 12/28/2022] Open
Abstract
The application and use of iron oxide nanoparticless (IONPs) in the biomedical field are steadily increasing, although it remains uncertain whether IONPs are safe or should be used with caution. In the present study, we investigated the toxicity profile of ultrafine IONPs in rats administered with 7.5, 15 and 30 mg IONPs/kg body wt intravenously once a week for 4 weeks. IONP treatment reduces bone marrow-mononuclear cell proliferation, increases free radical species and DNA damage leading to growth arrest and subsequently apoptosis induction at 15 and 30 mg doses. It also induces apoptosis in undifferentiated hematopoietic stem cells. IONP treatment significantly increased the pro-inflammatory cytokine (Interleukin (IL)-1β, TNF-α, and IL-6) level in serum. The induction in inflammation was likely mediated by splenic M1 macrophages (IL-6 and TNF-α secretion). IONP treatment induces splenocyte apoptosis and alteration in the immune system represented by reduced CD4+/CD8+ ratio and increased B cells. It also reduces innate defense represented by lower natural killer cell cytotoxicity. IONP administration markedly increased lipid peroxidation in the spleen, while the glutathione level was reduced. Similarly, superoxide dismutase activity was increased and catalase activity was reduced in the spleen of IONP-treated rats. At an organ level, IONP treatment did not cause any significant injury or structural alteration in the spleen. Collectively, our results suggest that a high dose of ultrafine IONPs may cause oxidative stress, cell death, and inflammation in a biological system.
Collapse
Affiliation(s)
- Usha Singh Gaharwar
- School of Environmental Sciences, Jawaharlal Nehru University New Delhi 110067 India +91-11-26741586 +91-11-26704162
| | - Sumit Kumar
- School of Life Sciences, Jawaharlal Nehru University New Delhi India
| | - Paulraj Rajamani
- School of Environmental Sciences, Jawaharlal Nehru University New Delhi 110067 India +91-11-26741586 +91-11-26704162
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Erythropoiesis is a complex multistep process going from committed erythroid progenitors to mature red cells. Although recent advances allow the characterization of some components of erythropoiesis, much still remains to be investigated particularly on stress erythropoiesis. This review summarizes recent progresses made to understand the impact of oxidative stress on normal and pathologic erythropoiesis. RECENT FINDINGS During erythroid maturation, reactive oxygen species might function as second messenger through either transient oxidation of cysteine residues on signaling targets or modulation of intracellular signaling pathways. Thus, in erythropoiesis, efficient cytoprotective systems are required to limit possible reactive oxygen species-related toxic effects especially in stress erythropoiesis characterized by severe oxidation such as β-thalassemia. In addition, prolonged or severe oxidative stress impairs autophagy, which might contribute to the block of erythroid maturation in stress erythropoiesis. Understanding the functional role of cytoprotective systems such as peroxiredoxin-2 or classical molecular chaperones such as the heat shock proteins will contribute to develop innovative therapeutic strategies for ineffective erythropoiesis. SUMMARY We provide an update on cytoprotective mechanisms against oxidation in normal and stress erythropoiesis. We discuss the role of oxidative sensors involved in modulation of intracellular signaling during erythroid maturation process in normal and stress erythropoiesis.
Collapse
|
37
|
Hennigar SR, Berryman CE, Harris MN, Karl JP, Lieberman HR, McClung JP, Rood JC, Pasiakos SM. Testosterone Administration During Energy Deficit Suppresses Hepcidin and Increases Iron Availability for Erythropoiesis. J Clin Endocrinol Metab 2020; 105:5693356. [PMID: 31894236 DOI: 10.1210/clinem/dgz316] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 12/30/2019] [Indexed: 12/18/2022]
Abstract
CONTEXT Severe energy deprivation markedly inhibits erythropoiesis by restricting iron availability for hemoglobin synthesis. OBJECTIVE The objective of this study was to determine whether testosterone supplementation during energy deficit increased indicators of iron turnover and attenuated the decline in erythropoiesis compared to placebo. DESIGN This was a 3-phase, randomized, double-blind, placebo-controlled trial. SETTING The study was conducted at the Pennington Biomedical Research Center. PATIENTS OR OTHER PARTICIPANTS Fifty healthy young males. INTERVENTION(S) Phase 1 was a 14-day free-living eucaloric controlled-feeding phase; phase 2 was a 28-day inpatient phase where participants were randomized to 200 mg testosterone enanthate/week or an isovolumetric placebo/week during an energy deficit of 55% of total daily energy expenditure; phase 3 was a 14-day free-living, ad libitum recovery period. MAIN OUTCOME MEASURE(S) Indices of erythropoiesis, iron status, and hepcidin and erythroferrone were determined. RESULTS Hepcidin declined by 41%, indicators of iron turnover increased, and functional iron stores were reduced with testosterone administration during energy deficit compared to placebo. Testosterone administration during energy deficit increased circulating concentrations of erythropoietin and maintained erythropoiesis, as indicated by an attenuation in the decline in hemoglobin and hematocrit with placebo. Erythroferrone did not differ between groups, suggesting that the reduction in hepcidin with testosterone occurs through an erythroferrone-independent mechanism. CONCLUSION These findings indicate that testosterone suppresses hepcidin, through either direct or indirect mechanisms, to increase iron turnover and maintain erythropoiesis during severe energy deficit. This trial was registered at www.clinicaltrials.gov as #NCT02734238.
Collapse
Affiliation(s)
- Stephen R Hennigar
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, Florida
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, Massachusetts
- Oak Ridge Institute of Science and Technology, Belcamp, Maryland
| | - Claire E Berryman
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, Florida
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, Massachusetts
- Oak Ridge Institute of Science and Technology, Belcamp, Maryland
| | | | - J Philip Karl
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, Massachusetts
| | - Harris R Lieberman
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, Massachusetts
| | - James P McClung
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, Massachusetts
| | - Jennifer C Rood
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Stefan M Pasiakos
- Military Nutrition Division, U.S. Army Research Institute of Environmental Medicine, Natick, Massachusetts
| |
Collapse
|
38
|
Chen Y, Wan J, Xia H, Li Y, Xu Y, Lin H, Iftikhar H. Total iron binding capacity (TIBC) is a potential biomarker of left ventricular remodelling for patients with iron deficiency anaemia. BMC Cardiovasc Disord 2020; 20:4. [PMID: 31914941 PMCID: PMC6947819 DOI: 10.1186/s12872-019-01320-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/24/2019] [Indexed: 01/21/2023] Open
Abstract
Background Preclinical studies indicate iron deficiency (ID) plays an important role in cardiac remodelling. However, the relationship between ID and cardiac remodelling remains unknown in clinical setting. This retrospective study aims to identify a potential biomarker for the myocardial remodelling in patients with ID. Due to limited patients with ID are identified without iron deficiency anaemia (IDA), we analyse the relationship of total iron binding capacity (TIBC) and the left ventricular mass index (LVMI) in patients with iron deficiency anaemia. Methods A total of 82 patients with IDA exhibiting the diagnostic criteria for IDA were enrolled in the study. Among the patients, 65 had reported LVMI values. Subsequently, these patients were divided into two groups according to abnormal LVMI (> 115 g/m2 in men and > 95 g/m2 in women). Linear bivariate analysis was performed to detect the associations of haemoglobin or TIBC with clinical and echocardiographic characteristics. Simple linear regression analysis was used to evaluate the correlation between LVMI and the parameters of IDA, while multivariable linear analysis was used to assess the association of LVMI with age, TIBC and haemoglobin. Logistic regression analysis was utilized to determine the relationship of LV remodelling with anaemia severity and TIBC. Results As compared with control group, the levels of TIBC in abnormal LVMI group are increased. Using log transformed LVMI as the dependent variable, simultaneously introducing age, TIBC, and haemoglobin into the simple linear regression or multivariable linear regression analysis confirmed the positive association among these factors. Bivariate correlation analysis reveals the irrelevance between haemoglobin and TIBC. In logistic regression analysis, TIBC is associated with the risk of LV remodelling. Conclusions Results of study indicate that TIBC exhibit an explicit association with LVMI in patients with iron deficiency anaemia. Logistic analysis further confirms the contribution of TIBC to abnormal LVMI incidence among this population with IDA.
Collapse
Affiliation(s)
- Yan Chen
- Division of Cardiology, Department of Medicine, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, People's Republic of China, 430071
| | - Jing Wan
- Division of Cardiology, Department of Medicine, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, People's Republic of China, 430071.
| | - Haidan Xia
- Division of Cardiology, Department of Medicine, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, People's Republic of China, 430071
| | - Ya Li
- Division of Cardiology, Department of Medicine, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, People's Republic of China, 430071
| | - Yufeng Xu
- Division of Hematology, Department of Medicine, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, People's Republic of China, 430071
| | - Haiyan Lin
- Division of Hematology, Department of Medicine, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, People's Republic of China, 430071
| | - Hassah Iftikhar
- Division of Cardiology, Department of Medicine, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, People's Republic of China, 430071
| |
Collapse
|
39
|
Brissot E, Bernard DG, Loréal O, Brissot P, Troadec MB. Too much iron: A masked foe for leukemias. Blood Rev 2020; 39:100617. [DOI: 10.1016/j.blre.2019.100617] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/20/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023]
|
40
|
Hall R, Peeling P, Nemeth E, Bergland D, McCluskey WTP, Stellingwerff T. Single versus Split Dose of Iron Optimizes Hemoglobin Mass Gains at 2106 m Altitude. Med Sci Sports Exerc 2019; 51:751-759. [PMID: 30882751 DOI: 10.1249/mss.0000000000001847] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE To determine if a single versus a split equivalent daily dose of elemental iron was superior for hemoglobin mass (Hbmass) gains at altitude while minimizing gastrointestinal (GI) discomfort. METHODS Twenty-four elite runners attended a 3.1 ± 0.3 wk training camp (Flagstaff, AZ; 2106 m). A two-group design, randomized and stratified to baseline Hbmass, sex, and ferritin (>30 μ·L), was implemented daily as: 1) single dose of 1 × 200 mg (PM only, SINGLE) versus 2) split dose of 2 × 100 mg (AM and PM; SPLIT) elemental iron (ferrous fumarate). The Hbmass and venipuncture assessments were completed upon arrival and departure (±2 d) from camp for ferritin, hepcidin, and erythroferrone (ERFE) concentrations. Validated food frequency, GI distress, menstrual blood loss (MBL) and training questionnaires were implemented throughout. Univariate analysis was used to compare Hbmass, with baseline ferritin, dietary iron intake, MBL, and training volume used as covariates. RESULTS Both conditions increased Hbmass from baseline (P < 0.05), with SINGLE (867.3 ± 47.9 g) significantly higher than SPLIT (828.9 ± 48.9 g) (P = 0.048). The GI scores were worse in SINGLE for weeks 1 and 2 combined (SINGLE, 18.0 ± 6.7 points; SPLIT, 11.3 ± 6.9 points; P = 0.025); however, GI scores improved by week 3, resulting in no between-group differences (P = 0.335). Hepcidin significantly decreased over time (P = 0.043) in SINGLE, with a nonsignificant decrease evident in SPLIT (~22%). ERFE significantly decreased in both groups (~28.5%; P < 0.05). No between-group differences existed for ERFE, hepcidin, food frequency, MBL, or daily training outcomes (P > 0.05). CONCLUSIONS A single nightly 200-mg dose of elemental iron was superior to a split dose for optimizing Hbmass changes at altitude in runners over an approximately 3-wk training camp.
Collapse
Affiliation(s)
- Rebecca Hall
- Canadian Sport Institute Pacific, Vancouver & Victoria, British Columbia, CANADA
| | - Peter Peeling
- School of Human Sciences (Exercise and Sport Science), University of Western Australia, Crawley, Western Australia, AUSTRALIA.,Western Australian Institute of Sport, Mt Claremont, Western Australia, AUSTRALIA
| | - Elizabeta Nemeth
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Dan Bergland
- Hypo2 High Performance Sport Center, Flagstaff, AZ
| | - Walter T P McCluskey
- Canadian Sport Institute Pacific, Vancouver & Victoria, British Columbia, CANADA
| | - Trent Stellingwerff
- Canadian Sport Institute Pacific, Vancouver & Victoria, British Columbia, CANADA.,Department of Exercise Science, Physical & Health Education, University of Victoria, British Columbia, CANADA
| |
Collapse
|
41
|
Matte A, Federti E, Winter M, Koerner A, Harmeier A, Mazer N, Tomka T, Di Paolo ML, De Falco L, Andolfo I, Beneduce E, Iolascon A, Macias-Garcia A, Chen JJ, Janin A, Lebouef C, Turrini F, Brugnara C, De Franceschi L. Bitopertin, a selective oral GLYT1 inhibitor, improves anemia in a mouse model of β-thalassemia. JCI Insight 2019; 4:130111. [PMID: 31593554 DOI: 10.1172/jci.insight.130111] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/02/2019] [Indexed: 01/09/2023] Open
Abstract
Anemia of β-thalassemia is caused by ineffective erythropoiesis and reduced red cell survival. Several lines of evidence indicate that iron/heme restriction is a potential therapeutic strategy for the disease. Glycine is a key initial substrate for heme and globin synthesis. We provide evidence that bitopertin, a glycine transport inhibitor administered orally, improves anemia, reduces hemolysis, diminishes ineffective erythropoiesis, and increases red cell survival in a mouse model of β-thalassemia (Hbbth3/+ mice). Bitopertin ameliorates erythroid oxidant damage, as indicated by a reduction in membrane-associated free α-globin chain aggregates, in reactive oxygen species cellular content, in membrane-bound hemichromes, and in heme-regulated inhibitor activation and eIF2α phosphorylation. The improvement of β-thalassemic ineffective erythropoiesis is associated with diminished mTOR activation and Rab5, Lamp1, and p62 accumulation, indicating an improved autophagy. Bitopertin also upregulates liver hepcidin and diminishes liver iron overload. The hematologic improvements achieved by bitopertin are blunted by the concomitant administration of the iron chelator deferiprone, suggesting that an excessive restriction of iron availability might negate the beneficial effects of bitopertin. These data provide important and clinically relevant insights into glycine restriction and reduced heme synthesis strategies for the treatment of β-thalassemia.
Collapse
Affiliation(s)
- Alessandro Matte
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Verona, Policlinico GB Rossi, Verona, Italy
| | - Enrica Federti
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Verona, Policlinico GB Rossi, Verona, Italy
| | - Michael Winter
- Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Annette Koerner
- Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Anja Harmeier
- Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Norman Mazer
- Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Tomas Tomka
- Pharmaceutical Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Luigia De Falco
- Department of Molecular Medicine and Medical Biotechnology, University Federico II and CEINGE, Naples, Italy
| | - Immacolata Andolfo
- Department of Molecular Medicine and Medical Biotechnology, University Federico II and CEINGE, Naples, Italy
| | - Elisabetta Beneduce
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Verona, Policlinico GB Rossi, Verona, Italy
| | - Achille Iolascon
- Department of Molecular Medicine and Medical Biotechnology, University Federico II and CEINGE, Naples, Italy
| | - Alejandra Macias-Garcia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jane-Jane Chen
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Anne Janin
- INSERM, U1165, Paris, France.,Université Paris 7 - Denis Diderot, Paris, France.,AP-HP, Hôpital Saint-Louis, Paris, France
| | - Christhophe Lebouef
- INSERM, U1165, Paris, France.,Université Paris 7 - Denis Diderot, Paris, France.,AP-HP, Hôpital Saint-Louis, Paris, France
| | - Franco Turrini
- Department of Oncology, University of Torino, Torino, Italy
| | - Carlo Brugnara
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lucia De Franceschi
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Verona, Policlinico GB Rossi, Verona, Italy
| |
Collapse
|
42
|
Association of Daily Dietary Intake and Inflammation Induced by Marathon Race. Mediators Inflamm 2019; 2019:1537274. [PMID: 31686980 PMCID: PMC6800895 DOI: 10.1155/2019/1537274] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023] Open
Abstract
Daily food intake is crucial to maintain health and determine endogenous fuel to practice endurance exercise. We investigated the association between quantity of macronutrient and micronutrient daily intake and inflammation induced by long-distance exercise. Methods. Forty-four Brazilian male amateurs' marathon finishers from 30 to 55 years old participated in this study. Blood samples were collected 1 day before, immediately after, and 1 day and 3 days after São Paulo International Marathon. The serum levels of IL-6, IL-1β, IL-10, IL-8, IL-12p70, and TNF-α were measured to evaluate inflammation. Dietary intake was determined using a prospective method of three food records in the week before marathon race. Results. Marathon race promoted an elevation on IL-6, IL-8, IL-1-β, and IL-10 immediately after the race. The energy intake (EI), carbohydrate, fiber, folic acid, vitamin E, vitamin D, calcium, magnesium, and potassium intakes was below recommended. Immediately after the marathon race, we observed a negative correlation between IL-8 and daily EI, carbohydrate, fiber, fat, iron, calcium, potassium, and sodium intakes, and higher levels of IL-8 on runners with <3 g/kg/day of carbohydrate intake compared to runners with >5 g/kg/day. We demonstrated a positive correlation between daily carbohydrate intake and IL-10 and a negative correlation between TNF-α and % of energy intake recommended, carbohydrate and fiber intakes. Finally, runners with adequate EI had lower levels of IL-1β and TNF-α compared with low EI immediately after the race. Conclusion. Nutrition strategies to promote balanced diet in amateur runners seem to be as important as immunonutrition sports market. Daily food intake, mainly EI, electrolyte and carbohydrate intakes, may modulate exacerbated inflammation after endurance exercise.
Collapse
|
43
|
Pek RH, Yuan X, Rietzschel N, Zhang J, Jackson L, Nishibori E, Ribeiro A, Simmons W, Jagadeesh J, Sugimoto H, Alam MZ, Garrett L, Haldar M, Ralle M, Phillips JD, Bodine DM, Hamza I. Hemozoin produced by mammals confers heme tolerance. eLife 2019; 8:e49503. [PMID: 31571584 PMCID: PMC6773446 DOI: 10.7554/elife.49503] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/24/2019] [Indexed: 12/28/2022] Open
Abstract
Free heme is cytotoxic as exemplified by hemolytic diseases and genetic deficiencies in heme recycling and detoxifying pathways. Thus, intracellular accumulation of heme has not been observed in mammalian cells to date. Here we show that mice deficient for the heme transporter SLC48A1 (also known as HRG1) accumulate over ten-fold excess heme in reticuloendothelial macrophage lysosomes that are 10 to 100 times larger than normal. Macrophages tolerate these high concentrations of heme by crystallizing them into hemozoin, which heretofore has only been found in blood-feeding organisms. SLC48A1 deficiency results in impaired erythroid maturation and an inability to systemically respond to iron deficiency. Complete heme tolerance requires a fully-operational heme degradation pathway as haplo insufficiency of HMOX1 combined with SLC48A1 inactivation causes perinatal lethality demonstrating synthetic lethal interactions between heme transport and degradation. Our studies establish the formation of hemozoin by mammals as a previously unsuspected heme tolerance pathway.
Collapse
Affiliation(s)
- Rini H Pek
- Department of Animal and Avian SciencesUniversity of MarylandCollege ParkUnited States
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkUnited States
| | - Xiaojing Yuan
- Department of Animal and Avian SciencesUniversity of MarylandCollege ParkUnited States
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkUnited States
| | - Nicole Rietzschel
- Department of Animal and Avian SciencesUniversity of MarylandCollege ParkUnited States
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkUnited States
| | - Jianbing Zhang
- Department of Animal and Avian SciencesUniversity of MarylandCollege ParkUnited States
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkUnited States
| | - Laurie Jackson
- Department of MedicineUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - Eiji Nishibori
- Faculty of Pure and Applied SciencesUniversity of TsukubaTsukubaJapan
- Tsukuba Research Center for Energy Materials ScienceUniversity of TsukabaTsukabaJapan
| | - Ana Ribeiro
- Department of Animal and Avian SciencesUniversity of MarylandCollege ParkUnited States
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkUnited States
| | - William Simmons
- Genetics and Molecular Biology BranchNational Human Genome Research Institute, National Institutes of HealthBethesdaUnited States
| | - Jaya Jagadeesh
- Genetics and Molecular Biology BranchNational Human Genome Research Institute, National Institutes of HealthBethesdaUnited States
| | | | - Md Zahidul Alam
- Department of Pathology and Laboratory MedicinePerelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Lisa Garrett
- NHGRI Embryonic Stem Cell and Transgenic Mouse CoreNational Human Genome Research Institute, National Institutes of HealthBethesdaUnited States
| | - Malay Haldar
- Department of Pathology and Laboratory MedicinePerelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Martina Ralle
- Department of Molecular and Medical GeneticsOregon Health and Science UniversityPortlandUnited States
| | - John D Phillips
- Department of MedicineUniversity of Utah School of MedicineSalt Lake CityUnited States
| | - David M Bodine
- Genetics and Molecular Biology BranchNational Human Genome Research Institute, National Institutes of HealthBethesdaUnited States
| | - Iqbal Hamza
- Department of Animal and Avian SciencesUniversity of MarylandCollege ParkUnited States
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkUnited States
| |
Collapse
|
44
|
Abstract
Iron is an essential element that is indispensable for life. The delicate physiological body iron balance is maintained by both systemic and cellular regulatory mechanisms. The iron-regulatory hormone hepcidin assures maintenance of adequate systemic iron levels and is regulated by circulating and stored iron levels, inflammation and erythropoiesis. The kidney has an important role in preventing iron loss from the body by means of reabsorption. Cellular iron levels are dependent on iron import, storage, utilization and export, which are mainly regulated by the iron response element-iron regulatory protein (IRE-IRP) system. In the kidney, iron transport mechanisms independent of the IRE-IRP system have been identified, suggesting additional mechanisms for iron handling in this organ. Yet, knowledge gaps on renal iron handling remain in terms of redundancy in transport mechanisms, the roles of the different tubular segments and related regulatory processes. Disturbances in cellular and systemic iron balance are recognized as causes and consequences of kidney injury. Consequently, iron metabolism has become a focus for novel therapeutic interventions for acute kidney injury and chronic kidney disease, which has fuelled interest in the molecular mechanisms of renal iron handling and renal injury, as well as the complex dynamics between systemic and local cellular iron regulation.
Collapse
|
45
|
Hara M, Nakamura Y, Suzuki H, Asao R, Nakamura M, Nishida K, Kenmotsu S, Inagaki M, Tsuji M, Kiuchi Y, Ohsawa I, Goto Y, Gotoh H. Hepcidin‐25/erythroferrone ratio predicts improvement of anaemia in haemodialysis patients treated with ferric citrate hydrate. Nephrology (Carlton) 2019; 24:819-826. [DOI: 10.1111/nep.13495] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Masaki Hara
- Department of Internal MedicineSaiyu Soka Hospital Soka‐shi Japan
- Tokyo Dialysis Frontier Ikebukuro Station North Clinic Tokyo Japan
| | - Yuya Nakamura
- Department of Internal MedicineSaiyu Soka Hospital Soka‐shi Japan
- Department of PharmacologySchool of Medicine Tokyo Japan
| | - Hiroki Suzuki
- Department of Internal MedicineSaiyu Soka Hospital Soka‐shi Japan
| | - Rin Asao
- Department of Internal MedicineSaiyu Soka Hospital Soka‐shi Japan
| | | | - Kazumasa Nishida
- Department of Internal MedicineSaiyu Soka Hospital Soka‐shi Japan
| | - Sachiyo Kenmotsu
- Department of ChemistryCollege of Arts and Sciences, Showa University Tokyo Japan
| | - Masahiro Inagaki
- Department of ChemistryCollege of Arts and Sciences, Showa University Tokyo Japan
| | - Mayumi Tsuji
- Department of PharmacologySchool of Medicine Tokyo Japan
| | - Yuji Kiuchi
- Department of PharmacologySchool of Medicine Tokyo Japan
| | - Isao Ohsawa
- Department of Internal MedicineSaiyu Soka Hospital Soka‐shi Japan
| | - Yoshikazu Goto
- Department of Internal MedicineSaiyu Soka Hospital Soka‐shi Japan
| | - Hiromichi Gotoh
- Department of Internal MedicineSaiyu Soka Hospital Soka‐shi Japan
| |
Collapse
|
46
|
Dopsaj V, Topić A, Savković M, Milinković N, Novaković I, Ćujić D, Simić-Ogrizović S. Associations of Common Variants in HFE and TMPRSS6 Genes with Hepcidin-25 and Iron Status Parameters in Patients with End-Stage Renal Disease. DISEASE MARKERS 2019; 2019:4864370. [PMID: 30984307 PMCID: PMC6431474 DOI: 10.1155/2019/4864370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/28/2019] [Accepted: 02/05/2019] [Indexed: 02/02/2023]
Abstract
BACKGROUND Influence of TMPRSS6 A736V and HFE (C282Y and H63D) polymorphisms on serum hepcidin-25 levels and iron status parameters in end-stage renal disease (ESRD) patients stratified according to gender has not been previously investigated. In addition, we aimed to evaluate the diagnostic accuracy of the parameters to separate iron-deficiency anemia (IDA) from anemia of chronic disease. MATERIALS AND METHODS Iron status parameters and genetic analysis were performed in 126 ESRD patients and in 31 IDA patients as the control group. RESULTS ESRD patients had significantly higher ferritin and hepcidin-25 (<0.001) relative to IDA patients. Cut-off values with the best diagnostic accuracy were found for hepcidin ≥9.32 ng/mL, ferritin ≥48.2 μg/L, transferrin saturation ≥16.8%, and MCV ≥81 fL. Interaction between gender and HFE haplotypes for the hepcidin-25 and ferritin levels in ESRD patients (p = 0.005, partial eta squared = 0.09; p = 0.027, partial eta squared = 0.06, respectively) was found. Serum transferrin was influenced by the combined effect of gender and TMPRSS6 A736V polymorphism in ESRD patients (p = 0.002, partial eta squared = 0.07). CONCLUSION Our findings could contribute to the further investigation of mechanisms involved in the pathophysiology and important gender-related involvement of the TMPRSS6 and HFE polymorphisms on anemia in ESRD patients.
Collapse
Affiliation(s)
- Violeta Dopsaj
- Department of Medical Biochemistry, University of Belgrade - Faculty of Pharmacy, Belgrade 11221, Serbia
- Center of Medical Biochemistry, Clinical Center of Serbia, Belgrade 11000, Serbia
| | - Aleksandra Topić
- Department of Medical Biochemistry, University of Belgrade - Faculty of Pharmacy, Belgrade 11221, Serbia
| | - Miljan Savković
- Center of Medical Biochemistry, Clinical Center of Serbia, Belgrade 11000, Serbia
| | - Neda Milinković
- Department of Medical Biochemistry, University of Belgrade - Faculty of Pharmacy, Belgrade 11221, Serbia
| | - Ivana Novaković
- Institute of Human Genetics, Medical Faculty, University of Belgrade, Belgrade 11010, Serbia
| | - Danica Ćujić
- Department of Biology of Reproduction, Institute for the Application of Nuclear Energy (INEP), University of Belgrade, Belgrade 11080, Serbia
| | - Sanja Simić-Ogrizović
- Clinic of Nephrology, Clinical Center of Serbia, Belgrade 11000, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| |
Collapse
|
47
|
Zhang WZ, Butler JJ, Cloonan SM. Smoking-induced iron dysregulation in the lung. Free Radic Biol Med 2019; 133:238-247. [PMID: 30075191 PMCID: PMC6355389 DOI: 10.1016/j.freeradbiomed.2018.07.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 07/26/2018] [Accepted: 07/30/2018] [Indexed: 12/12/2022]
Abstract
Iron is one of the most abundant transition elements and is indispensable for almost all organisms. While the ability of iron to participate in redox chemistry is an essential requirement for participation in a range of vital enzymatic reactions, this same feature of iron also makes it dangerous in the generation of hydroxyl radicals and superoxide anions. Given the high local oxygen tensions in the lung, the regulation of iron acquisition, utilization, and storage therefore becomes vitally important, perhaps more so than in any other biological system. Iron plays a critical role in the biology of essentially every cell type in the lung, and in particular, changes in iron levels have important ramifications on immune function and the local lung microenvironment. There is substantial evidence that cigarette smoke causes iron dysregulation, with the implication that iron may be the link between smoking and smoking-related lung diseases. A better understanding of the connection between cigarette smoke, iron, and respiratory diseases will help to elucidate pathogenic mechanisms and aid in the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- William Z Zhang
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA; Department of Medicine, New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY 10021, USA
| | - James J Butler
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Suzanne M Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA.
| |
Collapse
|
48
|
Moretti D, Mettler S, Zeder C, Lundby C, Geurts-Moetspot A, Monnard A, Swinkels DW, Brittenham GM, Zimmermann MB. An intensified training schedule in recreational male runners is associated with increases in erythropoiesis and inflammation and a net reduction in plasma hepcidin. Am J Clin Nutr 2018; 108:1324-1333. [PMID: 30351387 DOI: 10.1093/ajcn/nqy247] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 08/21/2018] [Indexed: 01/24/2023] Open
Abstract
Background Iron status is a determinant of physical performance, but training may induce both low-grade inflammation and erythropoiesis, exerting opposing influences on hepcidin and iron metabolism. To our knowledge, the combined effects on iron absorption and utilization during training have not been examined directly in humans. Objective We hypothesized that 3 wk of exercise training in recreational male runners would decrease oral iron bioavailability by increasing inflammation and hepcidin concentrations. Design In a prospective intervention, nonanemic, iron-sufficient men (n = 10) completed a 34-d study consisting of a 16-d control phase and a 22-d exercise-training phase of 8 km running every second day. We measured oral iron absorption and erythroid iron utilization using oral 57Fe and intravenous 58Fe tracers administered before and during training. We measured hemoglobin mass (mHb) and total red blood cell volume (RCV) by carbon monoxide rebreathing. Iron status, interleukin-6 (IL-6), plasma hepcidin (PHep), erythropoietin (EPO), and erythroferrone were measured before, during, and after training. Results Exercise training induced inflammation, as indicated by an increased mean ± SD IL-6 (0.87 ± 1.1 to 5.17 ± 2.2 pg/mL; P < 0.01), while also enhancing erythropoiesis, as indicated by an increase in mean EPO (0.66 ± 0.42 to 2.06 ± 1.6 IU/L), mHb (10.5 ± 1.6 to 10.8 ± 1.8 g/kg body weight), and mean RCV (30.7 ± 4.3 to 32.7 ± 4.6 mL/kg) (all P < 0.05). Training tended to increase geometric mean iron absorption by 24% (P = 0.083), consistent with a decreased mean ± SD PHep (7.25 ± 2.14 to 5.17 ± 2.24 nM; P < 0.05). The increase in mHb and erythroid iron utilization were associated with the decrease in PHep (P < 0.05). Compartmental modeling indicated that iron for the increase in mHb was obtained predominantly (>80%) from stores mobilization rather than from increased dietary absorption. Conclusions In iron-sufficient men, mild intensification of exercise intensity increases both inflammation and erythropoiesis. The net effect is to decrease hepcidin concentrations and to tend to increase oral iron absorption. This trial was registered at clinicaltrials.gov as NCT01730521.
Collapse
Affiliation(s)
- Diego Moretti
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology, ETH Zürich, Zurich, Switzerland
| | - Samuel Mettler
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology, ETH Zürich, Zurich, Switzerland.,Swiss Federal Institute of Sports, Magglingen, Switzerland.,Department of Business, Health, and Social Work, Bern University of Applied Sciences, Bern, Switzerland
| | - Christophe Zeder
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology, ETH Zürich, Zurich, Switzerland
| | - Carsten Lundby
- Zurich Center for Integrative Human Physiology, Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Anneke Geurts-Moetspot
- Hepcidinanalysis.com and Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, Netherlands
| | - Arnaud Monnard
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology, ETH Zürich, Zurich, Switzerland
| | - Dorine W Swinkels
- Hepcidinanalysis.com and Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, Netherlands
| | - Gary M Brittenham
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology, ETH Zürich, Zurich, Switzerland
| | - Michael B Zimmermann
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology, ETH Zürich, Zurich, Switzerland
| |
Collapse
|
49
|
Deregulated iron metabolism in bone marrow from adenine-induced mouse model of chronic kidney disease. Int J Hematol 2018; 109:59-69. [DOI: 10.1007/s12185-018-2531-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 12/11/2022]
|
50
|
Adris N, Chua ACG, Knuiman MW, Divitini ML, Trinder D, Olynyk JK. A prospective cohort examination of haematological parameters in relation to cancer death and incidence: the Busselton Health Study. BMC Cancer 2018; 18:863. [PMID: 30176879 PMCID: PMC6122556 DOI: 10.1186/s12885-018-4775-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 08/23/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cancer risk is associated with serum iron levels. The aim of this study was to evaluate whether haematological parameters reflect serum iron levels and may also be associated with cancer risk. METHODS We studied 1564 men and 1769 women who were enrolled in the Busselton Health Study, Western Australia. Haematological parameters evaluated included haemoglobin (Hb), mean cell volume (MCV), mean cell haemoglobin (MCH) and mean cell haemoglobin concentration (MCHC) and red cell distribution width (RCDW). Statistical analyses included t-tests for quantitative variables, chi-square tests for categorical variables and Cox proportional hazards regression modelling for cancer incidence and death. RESULTS There was marginal evidence of an association between MCV (as a continuous variable) and non-skin cancer incidence in women (HR 1.15, 95% CI 1.013, 1.302; p = 0.030) but the hazard ratio was attenuated to non-significance after adjustment for serum ferritin (SF), iron and transferrin saturation (TS) (HR 1.11, 95% CI 0.972, 1.264; p = 0.126). There was strong evidence of an association between MCHC and prostate cancer incidence in men; the estimated hazard ratio for an increase of one SD (0.5) in MCHC was 1.27 (95% CI 1.064, 1.507; p = 0.008). These results remained significant after further adjustment for SF and iron; the estimated hazard ratio for an increase of one SD (0.5) in MCHC was 1.25 (p = 0.014, 95% CI 1.05 to 1.48). CONCLUSIONS The MCHC and MCV were associated with cancer incidence in a Western Australian population, although only MCHC remained associated with prostate cancer after adjusting with serum iron and TS (circulating iron) and SF (storage iron). Haematological parameters are thus of limited utility in population profiling for future cancer risk.
Collapse
Affiliation(s)
- Niwansa Adris
- Department of Gastroenterology and Hepatology, Fiona Stanley and Fremantle Hospital Group, Murdoch, WA 6150 Australia
| | - Anita Chai Geik Chua
- Medical School, The University of Western Australia, Fiona Stanley Hospital, Murdoch, WA 6150 Australia
- Harry Perkins Institute of Medical Research, Murdoch, WA 6150 Australia
| | - Matthew William Knuiman
- School of Population and Global Health, The University of Western Australia, Crawley, WA 6009 Australia
| | - Mark Laurence Divitini
- School of Population and Global Health, The University of Western Australia, Crawley, WA 6009 Australia
| | - Debbie Trinder
- Medical School, The University of Western Australia, Fiona Stanley Hospital, Murdoch, WA 6150 Australia
- Harry Perkins Institute of Medical Research, Murdoch, WA 6150 Australia
| | - John Kevin Olynyk
- Department of Gastroenterology and Hepatology, Fiona Stanley and Fremantle Hospital Group, Murdoch, WA 6150 Australia
- School of Health and Medical Sciences, Edith Cowan University, Joondalup, 6027 Western Australia
| |
Collapse
|