1
|
Hanafusa N, Henn L, Bieber B, Hasegawa T, Usui T, Robinson B, Karaboyas A, Nangaku M. Erythropoiesis-stimulating agent hyporesponsiveness was associated with worse survival of hemodialysis patients independent of the serum ferritin level. Ther Apher Dial 2025; 29:23-33. [PMID: 38962901 DOI: 10.1111/1744-9987.14169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 04/25/2024] [Accepted: 05/15/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION Ferritin level and erythropoiesis-stimulating agent (ESA) responsiveness are each associated with hemodialysis patient survival. We assessed interrelationships between these two vs. survival. METHODS Patients in the Japan Dialysis Outcomes and Practice Patterns Study Phases 4-6 (2009-2018) were included. All-cause mortality associations were assessed with progressive adjustment to evaluate covariate influence. RESULTS During follow-up (median 2.6 years), 773 of 5154 patients died. After covariate adjustment, the mortality hazard ratio (HR) was 0.99 (95% CI: 0.81, 1.20) for low serum ferritin and 1.12 (CI: 0.89, 1.41) for high serum ferritin. By contrast, mortality risk with elevated ESA resistance index (ERI) persisted after covariate adjustment (HR 1.44, CI [1.17-1.78]). The serum ferritin and ERI interaction was not significant; p > 0.96 across all models. CONCLUSIONS Japanese hemodialysis patients with high ERI experienced worse survival independent of serum ferritin levels, highlighting the importance of identifying and mitigating ESA hyporesponsiveness among dialysis patients.
Collapse
Affiliation(s)
- Norio Hanafusa
- Department of Blood Purification, Kidney Center, Tokyo Women's Medical University, Tokyo, Japan
| | - Lisa Henn
- Arbor Research Collaborative for Health, Ann Arbor, Michigan, USA
| | - Brian Bieber
- Arbor Research Collaborative for Health, Ann Arbor, Michigan, USA
| | - Takeshi Hasegawa
- Institute of Clinical Epidemiology (iCE), Department of Nephrology, Graduate School of Medicine, Showa University, Tokyo, Japan
| | | | - Bruce Robinson
- Arbor Research Collaborative for Health, Ann Arbor, Michigan, USA
| | - Angelo Karaboyas
- Arbor Research Collaborative for Health, Ann Arbor, Michigan, USA
| | - Masaomi Nangaku
- Institute of Clinical Epidemiology (iCE), Department of Nephrology, Graduate School of Medicine, Showa University, Tokyo, Japan
| |
Collapse
|
2
|
Hu R, Li G, Hu P, Niu H, Li W, Jiang S, Guan G, Xu Q, Liu M, Chen L. bmp10 maintains cardiac function by regulating iron homeostasis. J Genet Genomics 2024; 51:1459-1473. [PMID: 39414074 DOI: 10.1016/j.jgg.2024.10.003] [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: 06/23/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 10/18/2024]
Abstract
Heart disease remains the leading cause of death worldwide. Iron imbalance, whether deficiency or overload, contributes to heart failure. However, the molecular mechanisms governing iron homeostasis in the heart are poorly understood. Here, we demonstrate that mutation of bmp10, a heart-born morphogen crucial for embryonic heart development, results in severe anemia and cardiac hypertrophy in zebrafish. Initially, bmp10 deficiency causes cardiac iron deficiency, which later progresses to iron overload due to the dysregulated hepcidin/ferroportin axis in cardiac cells, leading to ferroptosis and heart failure. Early iron supplementation in bmp10-/- mutants rescues erythropoiesis, while iron chelation in juvenile fishes significantly alleviates cardiac hypertrophy. We further demonstrate that the interplay between HIF1α-driven hypoxic signaling and the IL6/p-STAT3 inflammatory pathways is critical for regulating cardiac iron metabolism. Our findings reveal BMP10 as a key regulator of iron homeostasis in the vertebrate heart and highlight the potential of targeting the BMP10-hepcidin-iron axis as a therapeutic strategy for iron-related cardiomyopathy.
Collapse
Affiliation(s)
- Ruiqin Hu
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Genfang Li
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Peng Hu
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Hongbo Niu
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Wenhao Li
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Shouwen Jiang
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Guijun Guan
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Qianghua Xu
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Sustainable Exploitation of Oceanic Fisheries Resources, College of Marine Science, Shanghai Ocean University, Shanghai 201306, China
| | - Mingli Liu
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China
| | - Liangbiao Chen
- International Research Center for Marine Bioscience (Ministry of Science and Technology), Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education) Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
3
|
Maltaneri RE, Chamorro ME, Gionco SE, Nesse AB, Vittori DC. Erythropoietin enhances iron bioavailability in HepG2 cells by downregulating hepcidin through mTOR, C/EBPα and HIF-1α. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119800. [PMID: 39047915 DOI: 10.1016/j.bbamcr.2024.119800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 06/26/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
The regulation of iron (Fe) levels is essential to maintain an adequate supply for erythropoiesis, among other processes, and to avoid possible toxicity. The liver-produced peptide hepcidin is regarded as the main regulator of Fe absorption in enterocytes and release from hepatocytes and macrophages, as it impairs Fe export through ferroportin. The glycoprotein erythropoietin (Epo) drives erythroid progenitor survival and differentiation in the bone marrow, and has been linked to the mobilization of Fe reserves necessary for hemoglobin production. Herein we show that Epo inhibits hepcidin expression directly in the HepG2 hepatic cell line, thus leading to a decrease in intracellular Fe levels. Such inhibition was dependent on the Epo receptor-associated kinase JAK2, as well as on the PI3K/AKT/mTOR pathway, which regulates nutrient homeostasis. Epo was also found to decrease binding of the C/EBP-α transcription factor to the hepcidin promoter, which could be attributed to an increased expression of its inhibitor CHOP. Epo did not only hinder the stimulating effect of C/EBP-α on hepcidin transcription, but also favored hepcidin inhibition by HIF-1α, by increasing is nuclear translocation as well as its protein levels. Moreover, in assays with the inhibitor genistein, this transcription factor was found necessary for Epo-induced hepcidin suppression. Our findings support the involvement of the PI3K/AKT/mTOR pathway in the regulation of Fe levels by Epo, and highlight the contrasting roles of the C/EBP-α and HIF-1α transcription factors as downstream effectors of the cytokine in this process.
Collapse
Affiliation(s)
- Romina Eugenia Maltaneri
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina.
| | - María Eugenia Chamorro
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
| | - Silvana Estela Gionco
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
| | - Alcira Beatriz Nesse
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
| | - Daniela Cecilia Vittori
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto del Departamento de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
| |
Collapse
|
4
|
Gugo K, Tandara L, Juricic G, Pavicic Ivelja M, Rumora L. Effects of Hypoxia and Inflammation on Hepcidin Concentration in Non-Anaemic COVID-19 Patients. J Clin Med 2024; 13:3201. [PMID: 38892911 PMCID: PMC11173117 DOI: 10.3390/jcm13113201] [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: 04/22/2024] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Background/Objectives: This study aimed to explore the influence of hypoxia, inflammation, and erythropoiesis on hepcidin and other iron status parameters in non-anaemic COVID-19 patients admitted to the emergency unit before the introduction of therapeutic interventions. Methods: Ninety-six COVID-19 patients and 47 healthy subjects were recruited. Patients were subdivided into hypoxic or normoxic groups and, after follow-up, into mild and moderate, severe or critical disease severity groups. Iron, unsaturated iron-binding capacity (UIBC), ferritin, C-reactive protein (CRP), and interleukin 6 (IL-6) were measured on automatic analysers. ELISA kits were used for hepcidin and erythropoietin (EPO) determination. We calculated total iron-binding capacity (TIBC) and ratios of hepcidin with parameters of iron metabolism (ferritin/hepcidin, hepcidin/iron), inflammation (hepcidin/CRP, hepcidin/IL-6), and erythropoietic activity (hepcidin/EPO). Results: Hepcidin, ferritin, EPO, CRP, IL-6, ferritin/hepcidin, and hepcidin/iron were increased, while UIBC, TIBC, hepcidin/CRP, and hepcidin/IL-6 were decreased in hypoxic compared to normoxic patients as well as in patients with severe or critical disease compared to those with mild and moderate COVID-19. Regarding predictive parameters of critical COVID-19 occurrence, in multivariable logistic regression analysis, a combination of EPO and ferritin/hepcidin showed very good diagnostic performances and correctly classified 88% of cases, with an AUC of 0.838 (0.749-0.906). Conclusions: The hypoxic signal in our group of patients was not strong enough to overcome the stimulating effect of inflammation on hepcidin expression. EPO and ferritin/hepcidin might help to identify on-admission COVID-19 patients at risk of developing a critical form of the disease.
Collapse
Affiliation(s)
- Katarina Gugo
- Medical Laboratory Diagnostic Division, University Hospital of Split, Soltanska 1, 21000 Split, Croatia;
- Department of Health Studies, University of Split, Rudera Boskovica 35, 21000 Split, Croatia;
| | - Leida Tandara
- Medical Laboratory Diagnostic Division, University Hospital of Split, Soltanska 1, 21000 Split, Croatia;
- Department of Medical Chemistry and Biochemistry, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia
| | - Gordana Juricic
- Department of Laboratory Diagnostics, General Hospital Pula, Santoriova 24a, 52100 Pula, Croatia;
| | - Mirela Pavicic Ivelja
- Department of Health Studies, University of Split, Rudera Boskovica 35, 21000 Split, Croatia;
- Department of Infectious Diseases, University Hospital of Split, Soltanska 1, 21000 Split, Croatia
| | - Lada Rumora
- Department of Medical Biochemistry and Haematology, University of Zagreb Faculty of Pharmacy and Biochemistry, A. Kovacica 1, 10000 Zagreb, Croatia;
| |
Collapse
|
5
|
Abba ML, Riabov V, Nowak D, Hofmann WK, Boch T. Understanding iron homeostasis in MDS: the role of erythroferrone. Front Oncol 2024; 14:1404817. [PMID: 38835379 PMCID: PMC11148345 DOI: 10.3389/fonc.2024.1404817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/07/2024] [Indexed: 06/06/2024] Open
Abstract
Myelodysplastic neoplasms (MDS) are a heterogenous group of clonal stem cell disorders characterized by dysplasia and cytopenia in one or more cell lineages. Anemia is a very common symptom that is often treated with blood transfusions and/or erythropoiesis stimulating factors. Iron overload results from a combination of these factors together with the disease-associated ineffective erythropoiesis, that is seen especially in MDS cases with SF3B1 mutations. A growing body of research has shown that erythroferrone is an important regulator of hepcidin, the master regulator of systemic iron homeostasis. Consequently, it is of interest to understand how this molecule contributes to regulating the iron balance in MDS patients. This short review evaluates our current understanding of erythroferrone in general, but more specifically in MDS and seeks to place in context how the current knowledge could be utilized for prognostication and therapy.
Collapse
Affiliation(s)
- Mohammed L Abba
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Vladimir Riabov
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Daniel Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| | - Tobias Boch
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Hematology and Oncology, University Hospital Mannheim, Mannheim, Germany
| |
Collapse
|
6
|
Drummer DJ, McNiff JL, Howard EE, Gwin JA, Carrigan CT, Murphy NE, Wilson MA, Michalak J, Ryan BJ, McClung JP, Pasiakos SM, Margolis LM. Exogenous erythropoietin increases hematological status, fat oxidation, and aerobic performance in males following prolonged strenuous training. Physiol Rep 2024; 12:e16038. [PMID: 38757249 PMCID: PMC11099744 DOI: 10.14814/phy2.16038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
This study investigated the effects of EPO on hemoglobin (Hgb) and hematocrit (Hct), time trial (TT) performance, substrate oxidation, and skeletal muscle phenotype throughout 28 days of strenuous exercise. Eight males completed this longitudinal controlled exercise and feeding study using EPO (50 IU/kg body mass) 3×/week for 28 days. Hgb, Hct, and TT performance were assessed PRE and on Days 7, 14, 21, and 27 of EPO. Rested/fasted muscle obtained PRE and POST EPO were analyzed for gene expression, protein signaling, fiber type, and capillarization. Substrate oxidation and glucose turnover were assessed during 90-min of treadmill load carriage (LC; 30% body mass; 55 ± 5% V̇O2peak) exercise using indirect calorimetry, and 6-6-[2H2]-glucose PRE and POST. Hgb and Hct increased, and TT performance improved on Days 21 and 27 compared to PRE (p < 0.05). Energy expenditure, fat oxidation, and metabolic clearance rate during LC increased (p < 0.05) from PRE to POST. Myofiber type, protein markers of mitochondrial biogenesis, and capillarization were unchanged PRE to POST. Transcriptional regulation of mitochondrial activity and fat metabolism increased from PRE to POST (p < 0.05). These data indicate EPO administration during 28 days of strenuous exercise can enhance aerobic performance through improved oxygen carrying capacity, whole-body and skeletal muscle fat metabolism.
Collapse
Affiliation(s)
- Devin J. Drummer
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
- Oak Ridge Institute for Science and EducationBelcampMarylandUSA
| | - Julie L. McNiff
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
- Combat Feeding DivisionU.S. Army Combat Capabilities Development Command (DEVCOM)NatickMassachusettsUSA
| | - Emily E. Howard
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| | - Jess A. Gwin
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| | - Christopher T. Carrigan
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| | - Nancy E. Murphy
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| | - Marques A. Wilson
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| | - Julia Michalak
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
- Oak Ridge Institute for Science and EducationBelcampMarylandUSA
| | - Benjamin J. Ryan
- Thermal and Mountain Medicine DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| | - James P. McClung
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| | - Stefan M. Pasiakos
- Office of Dietary Supplements, National Institutes of HealthU.S. Department of Health and Human ServicesBethesdaMarylandUSA
| | - Lee M. Margolis
- Military Nutrition DivisionU.S. Army Research Institute of Environmental MedicineNatickMassachusettsUSA
| |
Collapse
|
7
|
Yu Y, Su Y, Yang S, Liu Y, Lin Z, Das NK, Wu Q, Zhou J, Sun S, Li X, Yue W, Shah YM, Min J, Wang F. Activation of Intestinal HIF2α Ameliorates Iron-Refractory Anemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307022. [PMID: 38243847 DOI: 10.1002/advs.202307022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/06/2023] [Indexed: 01/22/2024]
Abstract
In clinics, hepcidin levels are elevated in various anemia-related conditions, particularly in iron-refractory anemia and in high inflammatory states that suppress iron absorption, which remains an urgent unmet medical need. To identify effective treatment options for various types of iron-refractory anemia, the potential effect of hypoxia and pharmacologically-mimetic drug FG-4592 (Roxadustat) are evaluated, a hypoxia-inducible factor (HIF)-prolyl hydroxylase (PHD) inhibitor, on mouse models of iron-refractory iron-deficiency anemia (IRIDA), anemia of inflammation and 5-fluorouracil-induced chemotherapy-related anemia. The potent protective effects of both hypoxia and FG-4592 on IRIDA as well as other 2 tested mouse cohorts are found. Mechanistically, it is demonstrated that hypoxia or FG-4592 could stabilize duodenal Hif2α, leading to the activation of Fpn transcription regardless of hepcidin levels, which in turn results in increased intestinal iron absorption and the amelioration of hepcidin-activated anemias. Moreover, duodenal Hif2α overexpression fully rescues phenotypes of Tmprss6 knockout mice, and Hif2α knockout in the gut significantly delays the recovery from 5-fluorouracil-induced anemia, which can not be rescued by FG-4592 treatment. Taken together, the findings of this study provide compelling evidence that targeting intestinal hypoxia-related pathways can serve as a potential therapeutic strategy for treating a broad spectrum of anemia, especially iron refractory anemia.
Collapse
Affiliation(s)
- Yingying Yu
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yunxing Su
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Sisi Yang
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yutong Liu
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zhiting Lin
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Nupur K Das
- Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Qian Wu
- International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Jiahui Zhou
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Shumin Sun
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xiaopeng Li
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Wuyang Yue
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yatrik M Shah
- Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Junxia Min
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fudi Wang
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
- The First Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, 421001, China
| |
Collapse
|
8
|
Olivera J, Zhang V, Nemeth E, Ganz T. Erythroferrone exacerbates iron overload and ineffective extramedullary erythropoiesis in a mouse model of β-thalassemia. Blood Adv 2023; 7:3339-3349. [PMID: 36995275 PMCID: PMC10345853 DOI: 10.1182/bloodadvances.2022009307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/08/2023] [Accepted: 03/29/2023] [Indexed: 03/31/2023] Open
Abstract
β-thalassemia is characterized by chronic hepcidin suppression and iron overload, even in patients who have not undergone transfusion. The HbbTh3/+ (Th3/+) mouse model of nontransfusion-dependent β-thalassemia (NTDBT) partially recapitulates the human phenotype but lacks chronic hepcidin suppression, progressive iron accumulation into adulthood, or the interindividual variation of the rate of iron loading observed in patients. Erythroferrone (ERFE) is an erythroid regulator that suppresses hepcidin during increased erythropoiesis. ERFE concentrations in the sera of patients with NTDBT correlate negatively with hepcidin levels but vary over a broad range, possibly explaining the variability of iron overload in patients. To analyze the effect of high ERFE concentrations on hepcidin and iron overload in NTDBT, we crossed Th3/+ mice with erythroid ERFE-overexpressing transgenic mice. Th3/ERFE-transgenic mice suffered high perinatal mortality, but embryos at E18.5 showed similar viability, appearance, and anemia effects as Th3/+ mice. Compared with Th3/+ littermates, adult Th3/ERFE mice had similarly severe anemia but manifested greater suppression of serum hepcidin and increased iron accumulation in the liver, kidney, and spleen. The Th3/ERFE mice had much higher concentrations of serum ERFE than either parental strain, a finding attributable to both a higher number of erythroblasts and higher production of ERFE by each erythroblast.Th3/+ and Th3/ERFE mice had similar red blood cell count and shortened erythrocyte lifespan, but Th3/ERFE mice had an increased number of erythroid precursors in their larger spleens, indicative of aggravated ineffective extramedullary erythropoiesis. Thus, high ERFE concentrations increase the severity of nontransfusional iron overload and ineffective erythropoiesis in thalassemic mice but do not substantially affect anemia or hemolysis.
Collapse
Affiliation(s)
- Joseph Olivera
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Vida Zhang
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Elizabeta Nemeth
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA
- Department of Pathology, David Geffen School of Medicine, UCLA, Los Angeles, CA
| |
Collapse
|
9
|
Ganz T, Nemeth E, Rivella S, Goldberg P, Dibble AR, McCaleb ML, Guo S, Monia BP, Barrett TD. TMPRSS6 as a Therapeutic Target for Disorders of Erythropoiesis and Iron Homeostasis. Adv Ther 2023; 40:1317-1333. [PMID: 36690839 PMCID: PMC10070284 DOI: 10.1007/s12325-022-02421-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 12/23/2022] [Indexed: 01/25/2023]
Abstract
TMPRSS6 is a serine protease highly expressed in the liver. Its role in iron regulation was first reported in 2008 when mutations in TMPRSS6 were shown to be the cause of iron-refractory iron deficiency anemia (IRIDA) in humans and in mouse models. TMPRSS6 functions as a negative regulator of the expression of the systemic iron-regulatory hormone hepcidin. Over the last decade and a half, growing understanding of TMPRSS6 biology and mechanism of action has enabled development of new therapeutic approaches for patients with diseases of erythropoiesis and iron homeostasis.ClinicalTrials.gov identifier NCT03165864.
Collapse
Affiliation(s)
- Tomas Ganz
- Department of Medicine and Pathology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
| | - Elizabeta Nemeth
- Department of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology, Children's Hospital of Philadelphia (CHOP), 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Cell and Molecular Biology Graduate Group (CAMB), University of Pennsylvania, Abramson Research Center, 3615 Civic Center Boulevard, Room 316B, Philadelphia, PA, 19104, USA
| | - Paul Goldberg
- Prilenia Therapeutics, Herzliya, Israel
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | | | | | - Shuling Guo
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | | | | |
Collapse
|
10
|
Abstract
Iron overload remains a lethal complication of β-thalassemia and other anemias caused by ineffective erythropoiesis. This review discusses the pathogenetic mechanisms of iron overload in thalassemia, at organismal, cellular, and molecular levels.
Collapse
Affiliation(s)
- Tomas Ganz
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA.
| | - Elizabeta Nemeth
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA
| |
Collapse
|
11
|
Yoshida S, Saito T, Shibagaki K, Hirao K, Yuza T, Tomosugi N, Honda H. Changes of biomarkers for erythropoiesis, iron metabolism, and FGF23 by supplementation with roxadustat in patients on hemodialysis. Sci Rep 2023; 13:3181. [PMID: 36823243 PMCID: PMC9950357 DOI: 10.1038/s41598-023-30331-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
This study aimed to confirm changes in biomarkers of erythropoiesis and iron metabolism and serum fibroblast growth factor 23 (FGF-23) during darbepoetin-α treatment and then switching to the hypoxia-inducible factor prolyl hydroxylase inhibitor roxadustat. A total of 28 patients on hemodialysis who received weekly doses of darbepoetin-α were switched to roxadustat. Biomarkers for erythropoiesis and iron metabolism and intact and C-terminal FGF-23 were measured in blood samples collected before the HD session on days - 7 (darbepoetin-α injection), - 4, and - 2, and days 0 (switch to roxadustat treatment, three times weekly), 3, 5, 7, 14, 21, and 28. Erythropoietin and erythroferrone levels were elevated on day - 4 by darbepoetin-α injection and decreased to baseline levels at day 0. Levels of erythropoietin were not significantly increased by roxadustat supplementation, but erythroferrone levels were continuously elevated, similar to darbepoetin-α treatment. Hepcidin-25 and total iron binding capacity were significantly decreased or increased in patients treated with roxadustat compared with darbepoetin-α. Changes of intact and C-terminal FGF-23 levels were parallel to changes of phosphate levels during roxadustat treatment. However, the actual and percentage changes of intact FGF-23 and C-terminal FGF-23 in patients with low ferritin levels were greater than those in patients with high ferritin levels. Roxadustat might stimulate erythropoiesis by increasing iron usage through hepcidin-25, which was suppressed by erythroferrone in the physiological erythropoietin condition. Changes of intact FGF-23 and C-terminal FGF-23 levels might be affected by roxadustat in patients on hemodialysis, especially those with a low-iron condition.
Collapse
Affiliation(s)
- Shunsuke Yoshida
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8666, Japan
| | - Tomohiro Saito
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8666, Japan
| | | | | | | | - Naohisa Tomosugi
- Division of Systems Bioscience for Drug Discovery, Medical Research Institute, Kanazawa Medical University, Kanazawa, Japan
| | - Hirokazu Honda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8666, Japan.
| |
Collapse
|
12
|
Arora EK, Sharma V. Iron metabolism: pathways and proteins in homeostasis. REV INORG CHEM 2022. [DOI: 10.1515/revic-2022-0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Iron is essential to human survival. The biological role and trafficking of this trace essential inorganic element which is also a potential toxin is constantly being researched and unfolded. Vital for oxygen transport, DNA synthesis, electron transport, neurotransmitter biosynthesis and present in numerous other heme and non-heme enzymes the physiological roles are immense. Understanding the molecules and pathways that regulate this essential element at systemic and cellular levels are of importance in improving therapeutic strategies for iron related disorders. This review highlights the progress in understanding the metabolism and trafficking of iron along with the pathophysiology of iron related disorders.
Collapse
Affiliation(s)
- Ekta Kundra Arora
- Chemistry Department, St. Stephen’s College , University of Delhi , Delhi 110007 , India
| | - Vibha Sharma
- Chemistry Department, St. Stephen’s College , University of Delhi , Delhi 110007 , India
| |
Collapse
|
13
|
Hildebrand S, Busbridge M, Duncan ND, Tam FWK, Ashby DR. Predictors of iron versus erythropoietin responsiveness in anemic hemodialysis patients. Hemodial Int 2022; 26:519-526. [PMID: 35833334 PMCID: PMC9796788 DOI: 10.1111/hdi.13030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 03/30/2022] [Accepted: 05/30/2022] [Indexed: 01/07/2023]
Abstract
Anemia protocols for hemodialysis patients usually titrate erythropoietin (ESA) according to hemoglobin and iron according to a threshold of ferritin, with variable response seen. A universally optimum threshold for ferritin may be incorrect, and another view is that ESA and iron are alternative anemia treatments, which should be selected based on the likely response to each. Hemodialysis patients developing moderate anemia were randomised to treatment with either an increase in ESA or a course of intravenous iron. Over 2423 patient-months in 197 patients, there were 133 anemia episodes with randomized treatment. Treatment failure was seen in 20/66 patients treated with ESA and 20/67 patients treated with iron (30.3 vs. 29.9%, p = 1.0). Successful ESA treatment was associated with lower C-reactive protein (13.5 vs. 28.6 mg/L, p = 0.038) and lower previous ESA dose (6621 vs. 9273 μg/week, p = 0.097). Successful iron treatment was associated with lower reticulocyte hemoglobin (33.8 vs. 35.5 pg, p = 0.047), lower hepcidin (91.4 vs. 131.0 μg/ml, p = 0.021), and higher C-reactive protein (29.5 vs. 12.6 mg/L, p = 0.085). A four-variable iron preference score was developed to indicate the more favorable treatment, which in a retrospective analysis reduced treatment failure to 17%. Increased ESA and iron are equally effective, though treatment failure occurs in almost 30%. Baseline variables including hepcidin can predict treatment response, and a four-variable score shows promise in allowing directed treatment with improved response rates.
Collapse
Affiliation(s)
- Sarah Hildebrand
- Hammersmith HospitalImperial College Healthcare NHS TrustLondonUK
| | - Mark Busbridge
- Hammersmith HospitalImperial College Healthcare NHS TrustLondonUK
| | - Neill D. Duncan
- Hammersmith HospitalImperial College Healthcare NHS TrustLondonUK
| | - Frederick W. K. Tam
- Hammersmith HospitalImperial College Healthcare NHS TrustLondonUK,Centre for Inflammatory Disease, Department of MedicineImperial College LondonLondonUK
| | - Damien R. Ashby
- Hammersmith HospitalImperial College Healthcare NHS TrustLondonUK
| |
Collapse
|
14
|
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: 2.3] [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
|
15
|
Joharapurkar AA, Patel VJ, Kshirsagar SG, Patel MS, Savsani HH, Kajavadara C, Valani D, Jain MR. Prolyl hydroxylase inhibitor desidustat improves anemia in erythropoietin hyporesponsive state. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100102. [PMID: 35570856 PMCID: PMC9096675 DOI: 10.1016/j.crphar.2022.100102] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/14/2022] [Accepted: 04/21/2022] [Indexed: 11/24/2022] Open
Abstract
Many anemic chronic kidney disease (CKD) patients are refractory to erythropoietin (EPO) effects due to inflammation, deranged iron utilization, and generation of EPO antibodies. This work assessed the effect of desidustat, an inhibitor of hypoxia inducible factor (HIF) prolyl hydroxylase (PHD), on EPO-refractory renal anemia. Sprague Dawley rats were made anemic by cisplatin (5 mg/kg, IP, single dose) and turpentine oil (5 mL/kg, SC, once a week). These rats were given recombinant human EPO (rhEPO, 1 μg/kg) and desidustat (15 or 30 mg/kg) for eight weeks. Separately, rhEPO (1-5 μg/kg) was given to anemic rats to sustain the normal hemoglobin levels and desidustat (15 mg/kg) for eight weeks. In another experiment, the anemic rats were treated rhEPO (5 μg/kg) for two weeks and then desidustat (15 mg/kg) for the next two weeks. Dosing of rhEPO was thrice a week, and for desidustat, it was on alternate days. Desidustat inhibited EPO-resistance caused by rhEPO treatment, decreased hepcidin, IL-6, IL-1β, and increased iron and liver ferroportin. Desidustat reduced EPO requirement and anti-EPO antibodies. Desidustat also maintained normal hemoglobin levels after cessation of rhEPO treatment. Thus, novel prolyl hydroxylase inhibitor desidustat can treat EPO resistance via improved iron utilization and decreased inflammation.
Collapse
Affiliation(s)
- Amit A. Joharapurkar
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| | - Vishal J. Patel
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| | - Samadhan G. Kshirsagar
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| | - Maulik S. Patel
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| | - Hardikkumar H. Savsani
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| | - Chetan Kajavadara
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| | - Darshan Valani
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| | - Mukul R. Jain
- Department of Pharmacology and Toxicology, Zydus Research Centre, Cadila Healthcare Limited, Sarkhej Bavla NH 8A, Moraiya, Ahmedabad, Gujarat, India
| |
Collapse
|
16
|
Jefferies R, Puttagunta H, Krishnan A, Irish A, Swaminathan R, Olynyk JK. mTOR Inhibitors Induce Erythropoietin Resistance in Renal Transplant Recipients. Front Med (Lausanne) 2022; 9:722058. [PMID: 35273970 PMCID: PMC8901721 DOI: 10.3389/fmed.2022.722058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 01/27/2022] [Indexed: 11/23/2022] Open
Abstract
Aim To elucidate the role of mTOR inhibitors on iron, hepcidin and erythropoietin-mediated regulation of hemopoiesis in stable renal transplant recipients (RTR). Background Impaired hemopoiesis is common following renal transplantation managed using mTOR inhibitors. The mechanisms responsible are uncertain but include direct effects on iron, hepcidin or erythropoietin-mediated hemopoiesis. Methods We conducted a single center prospective case-control study of 26 adult RTR with stable allograft function. RTR received stable mTOR dosing (cases, 11/26 [42%]) or stable tacrolimus dosing (controls, 15/26 [58%]). Baseline demographics, full blood count, renal function, iron studies, hepcidin-25, Interleukin-6 (IL-6) and erythropoietin (EPO) levels were determined. Results There were no differences in age, gender or allograft function. Mean daily sirolimus dose for cases was 1.72 mg, with mean trough level of 8.46 ng/mL. Mean daily tacrolimus dose for controls was 4.3 mg, with mean trough level of 5.8 ng/mL. There were no differences in mean hemoglobin (143 vs. 147 g/L; p = 0.59), MCV (88 vs. 90 fL; p = 0.35), serum ferritin (150 vs. 85.7 μg/L; p = 0.06), transferrin saturation (26 vs. 23.3%; p = 0.46), IL-6 (11 vs. 7.02 pg/ml; p = 0.14) or hepcidin-25 (3.62 vs. 3.26 nM; p = 0.76) between the groups. EPO levels were significantly higher in the group receiving mTOR therapy (16.8 vs. 8.49 IU/L; p = 0.028). On logistic regression analysis EPO level was the only variable that had a significant impact providing an odds ratio of 0.84 (95%CI 0.66–0.98). The area under the receiver operator characteristic curve (ROC) for the analysis was 0.77 (95%CI 0.54–0.94) with p = 0.04. Conclusion: Higher levels of EPO in the absence of deranged iron biochemistry or hepcidin-25 levels suggest that EPO resistance rather than impaired iron metabolism may contribute to the impaired hemopoiesis previously demonstrated in RTR on mTOR therapy.
Collapse
Affiliation(s)
- Reece Jefferies
- Department of Nephrology and Renal Transplant, Fiona Stanley Hospital, Perth, WA, Australia
| | - Harish Puttagunta
- Department of Nephrology and Renal Transplant, Fiona Stanley Hospital, Perth, WA, Australia
| | - Anoushka Krishnan
- Department of Nephrology and Renal Transplant, Fiona Stanley Hospital, Perth, WA, Australia
| | - Ashley Irish
- Department of Nephrology and Renal Transplant, Fiona Stanley Hospital, Perth, WA, Australia
| | - Ramyasuda Swaminathan
- Department of Nephrology and Renal Transplant, Fiona Stanley Hospital, Perth, WA, Australia
| | - John K Olynyk
- Department of Gastroenterology, Fiona Stanley Hospital, Perth, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| |
Collapse
|
17
|
Kaczorowska-Hac B, Luszczyk M, Wasilewska E, Antosiewicz J, Kaczor JJ. Erythropoietin Concentration in Boys With p.His63Asp Polymorphism of the HFE Gene. J Pediatr Hematol Oncol 2022; 44:e68-e73. [PMID: 33625078 DOI: 10.1097/mph.0000000000002126] [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: 10/09/2020] [Accepted: 01/24/2021] [Indexed: 10/22/2022]
Abstract
The molecular mechanism that regulates iron homeostasis is based on a network of signals, which reflect on the iron requirements of the body. HFE-related hemochromatosis is characterized by excessive intestinal absorption of dietary iron, in particular cases resulting in pathologically high iron storage in tissues and organs. During childhood, HFE gene homozygosity or heterozygosity manifests exclusively in the form of biochemical abnormalities. Because of their mutual link, bioavailable iron and endogenous erythropoietin (EPO) are indispensable for effective erythropoiesis. We analyzed the impact of p.(His63Asp) polymorphism of the HFE gene on erythropoiesis taking into consideration endogenous EPO production in the developmental age. In the study we performed, we observed a significant, strong and negative correlation between the concentration of EPO, hemoglobin, and red blood cell count. A negative trend was also noted on the impact of iron concentration and transferrin saturation on EPO production. In conclusion, this preliminary study demonstrates an impaired impact of endogenous EPO on erythropoiesis in the presence of increased iron content in carriers of p.(His63Asp) (heterozygotes) variant of the HFE gene in developmental age.
Collapse
Affiliation(s)
| | - Marcin Luszczyk
- Physiology and Biochemistry Gdansk University of Physical Education and Sport
| | | | - Jedrzej Antosiewicz
- Bioenergetics and Physiology of Exercise, Medical University of Gdansk, Gdansk, Poland
| | - Jan J Kaczor
- Physiology and Biochemistry Gdansk University of Physical Education and Sport
| |
Collapse
|
18
|
Pirotte M, Fillet M, Seidel L, Jaspers A, Baron F, Beguin Y. Erythroferrone and hepcidin as mediators between erythropoiesis and iron metabolism during allogeneic hematopoietic stem cell transplant. Am J Hematol 2021; 96:1275-1286. [PMID: 34310730 PMCID: PMC9291814 DOI: 10.1002/ajh.26300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/13/2021] [Accepted: 07/15/2021] [Indexed: 12/16/2022]
Abstract
Hematopoietic cell transplantation (HCT) brings important alterations in erythropoiesis and iron metabolism. Hepcidin, which regulates iron metabolism, increases in iron overload or inflammation and decreases with iron deficiency or activated erythropoiesis. Erythroferrone (ERFE) is the erythroid regulator of hepcidin. We investigated erythropoiesis and iron metabolism after allogeneic HCT in 70 patients randomized between erythropoietin (EPO) treatment or no EPO, by serially measuring hepcidin, ERFE, CRP (inflammation), soluble transferrin receptor (sTfR, erythropoiesis), serum iron and transferrin saturation (Tsat; iron for erythropoiesis) and ferritin (iron stores). We identified biological and clinical factors associated with serum hepcidin and ERFE levels. Serum ERFE correlated overall with sTfR and reticulocytes and inversely with hepcidin. Erythroferrone paralleled sTfR levels, dropping during conditioning and recovering with engraftment. Inversely, hepcidin peaked after conditioning and decreased during engraftment. Erythroferrone and hepcidin were not significantly different with or without EPO. Multivariate analyses showed that the major determinant of ERFE was erythropoiesis (sTfR, reticulocytes or serum Epo). Pretransplant hepcidin was associated with previous RBC transfusions and ferritin. After transplantation, the major determinants of hepcidin were iron status (ferritin at all time points and Tsat at day 56) and erythropoiesis (sTfR or reticulocytes or ERFE), while the impact of inflammation was less clear and clinical parameters had no detectable influence. Hepcidin remained significantly higher in patients with high compared to low pretransplant ferritin. After allogeneic HCT with or without EPO therapy, significant alterations of hepcidin occur between pretransplant and day 180, in correlation with iron status and inversely with erythroid ERFE.
Collapse
Affiliation(s)
- Michelle Pirotte
- Department of Hematology University Hospital of Liège and ULiege Liège Belgium
| | - Marianne Fillet
- Laboratory for the Analysis of Medicines CIRM, ULiege Liège Belgium
| | - Laurence Seidel
- Department of Biostatistics and Medico‐Economics University Hospital of Liège and ULiege Liège Belgium
| | - Aurélie Jaspers
- Department of Hematology University Hospital of Liège and ULiege Liège Belgium
| | - Fréderic Baron
- Department of Hematology University Hospital of Liège and ULiege Liège Belgium
| | - Yves Beguin
- Department of Hematology University Hospital of Liège and ULiege Liège Belgium
| |
Collapse
|
19
|
Effect of Perioperative Intravenous iron Supplementation for Complex Cardiac Surgery on Transfusion Requirements: A Randomized, Double-Blinded Placebo-Controlled Trial. Ann Surg 2021; 275:232-239. [PMID: 34171864 DOI: 10.1097/sla.0000000000005011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES We investigated whether routine perioperative intravenous iron replenishment reduces the requirement for packed erythrocytes (pRBC) transfusion. SUMMARY BACKGROUND DATA Patients undergoing complex cardiac surgery are at high risk of developing postoperative iron deficiency anemia, thus requiring transfusion, which is associated with adverse outcomes. METHODS Patients were randomized to receive either ferric derisomaltose 20 mg/kg (n = 103) or placebo (n = 101) twice during the perioperative period: 3 days before and after the surgery. The primary endpoint was the proportion of patients who received pRBC transfusion until postoperative day (POD) 10. Hemoglobin, reticulocyte count, serum iron profile, hepcidin, and erythropoietin were serially measured. RESULTS pRBC was transfused in 60.4% and 57.2% of patients in the control and iron group, respectively (P = 0.651). Hemoglobin concentration at 3 weeks postoperatively was higher in the iron group than in the control group (11.6 ± 1.5 g/dl vs. 10.9 ± 1.4 g/dl, P < 0.001). The iron group showed higher reticulocyte count (205 [150-267]×103/μl vs. 164 [122-207]×103/μl, P = 0.003) at POD 10. Transferrin saturation and serum ferritin were significantly increased in the iron group than in the control group (P < 0.001). Serum hepcidin was higher in the iron group than in the control group at POD 3 (106.3 [42.9-115.9] ng/ml vs. 39.3 [33.3-43.6] ng/ml, P < 0.001). Erythropoietin concentration increased postoperatively in both groups (P = 0.003), with no between-group difference. CONCLUSIONS Intravenous iron supplementation during index hospitalization for complex cardiac surgery did not minimize pRBC transfusion despite replenished iron store and augmented erythropoiesis, which may be attributed to enhanced hepcidin expression.
Collapse
|
20
|
Haase VH. Hypoxia-inducible factor-prolyl hydroxylase inhibitors in the treatment of anemia of chronic kidney disease. Kidney Int Suppl (2011) 2021; 11:8-25. [PMID: 33777492 PMCID: PMC7983025 DOI: 10.1016/j.kisu.2020.12.002] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/18/2020] [Accepted: 12/29/2020] [Indexed: 12/11/2022] Open
Abstract
Hypoxia-inducible factor-prolyl hydroxylase domain inhibitors (HIF-PHIs) are a promising new class of orally administered drugs currently in late-stage global clinical development for the treatment of anemia of chronic kidney disease (CKD). HIF-PHIs activate the HIF oxygen-sensing pathway and are efficacious in correcting and maintaining hemoglobin levels in patients with non-dialysis- and dialysis-dependent CKD. In addition to promoting erythropoiesis through the increase in endogenous erythropoietin production, HIF-PHIs reduce hepcidin levels and modulate iron metabolism, providing increases in total iron binding capacity and transferrin levels, and potentially reducing the need for i.v. iron supplementation. Furthermore, HIF-activating drugs are predicted to have effects that extend beyond erythropoiesis. This review summarizes clinical data from current HIF-PHI trials in patients with anemia of CKD, discusses mechanisms of action and pharmacologic properties of HIF-PHIs, and deliberates over safety concerns and potential impact on anemia management in patients with CKD.
Collapse
Affiliation(s)
- Volker H. Haase
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Molecular Physiology and Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
21
|
Robach P, Gammella E, Recalcati S, Girelli D, Castagna A, Roustit M, Lundby C, Lundby AK, Bouzat P, Vergès S, Séchaud G, Banco P, Uhr M, Cornu C, Sallet P, Cairo G. Induction of erythroferrone in healthy humans by micro-dose recombinant erythropoietin or high-altitude exposure. Haematologica 2021; 106:384-390. [PMID: 31919080 PMCID: PMC7849588 DOI: 10.3324/haematol.2019.233874] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/02/2020] [Indexed: 01/22/2023] Open
Abstract
The erythropoietin (Epo)-erythroferrone (ERFE)-hepcidin axis coordinates erythropoiesis and iron homeostasis. While mouse studies have established that Epo-induced ERFE production represses hepcidin synthesis by inhibiting hepatic BMP/SMAD signaling, evidence for the role of ERFE in humans is limited. To investigate the role of ERFE as a physiological erythroid regulator in humans, we conducted two studies. First, 24 males were given six injections of saline (placebo), recombinant Epo (rhEpo) at a dose of 20 IU/kg (micro-dose) or rhEpo at 50 IU/kg (low dose). Second, we quantified ERFE in 22 subjects exposed to high altitude (3800 m) for 15 h. In the first study, total hemoglobin mass (Hbmass) increased after low- but not after micro-dose injections, when compared to the mass after placebo injections. Serum ERFE levels were enhanced by rhEpo, remaining higher than after placebo for 48 h (micro-dose) or 72 h (low-dose) after injections. Conversely, hepcidin levels decreased when Epo and ERFE rose, before any changes in serum iron parameters occurred. In the second study, serum Epo and ERFE increased at high altitude. The present results demonstrate that in healthy humans ERFE responds to slightly increased Epo levels not associated with Hbmass expansion and downregulates hepcidin in an apparently iron-independent way. Notably, ERFE flags micro-dose Epo, thus holding promise as a novel biomarker of doping.
Collapse
Affiliation(s)
- Paul Robach
- National School for Mountains Sports, Chamonix, France
| | - Elena Gammella
- Dept Biomedical Sciences for Health, University of Milan, Italy
| | | | | | | | | | - Carsten Lundby
- Center for Physical Activity Research, University Hospital, Copenhagen, Denmark
| | | | - Pierre Bouzat
- Grenoble Alpes University Hospital, Grenoble, France
| | - Samuel Vergès
- HP2 Laboratory, U1042, Grenoble Alpes University, INSERM, Grenoble, France
| | | | | | - Mario Uhr
- Dept. Hematology Synlab-Suisse, Lugano, Switzerland
| | - Catherine Cornu
- Hospices Civils de Lyon INSERM CIC1407/UMR5558, Hôpital Louis Pradel, Bron, France
| | | | - Gaetano Cairo
- Dept Biomedical Sciences for Health, University of Milan, Italy
| |
Collapse
|
22
|
Yamaguchi Y, Zampino M, Tanaka T, Bandinelli S, Osawa Y, Ferrucci L, Semba RD. Elevated Plasma Growth and Differentiation Factor 15 Predicts Incident Anemia in Older Adults Aged 60 Years and Older. J Gerontol A Biol Sci Med Sci 2020; 76:1192-1197. [PMID: 33367500 DOI: 10.1093/gerona/glaa324] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Anemia is common in older adults and associated with greater morbidity and mortality. The causes of anemia in older adults have not been completely characterized. Although elevated circulating growth and differentiation factor 15 (GDF-15) has been associated with anemia in older adults, it is not known whether elevated GDF-15 predicts the development of anemia. We examined the relationship between plasma GDF-15 concentrations at baseline in 708 nonanemic adults, aged 60 years and older, with incident anemia during 15 years of follow-up among participants in the Invecchiare in Chianti (InCHIANTI) Study. During follow-up, 179 (25.3%) participants developed anemia. The proportion of participants who developed anemia from the lowest to highest quartile of plasma GDF-15 was 12.9%, 20.1%, 21.2%, and 45.8%, respectively. Adults in the highest quartile of plasma GDF-15 had an increased the risk of developing anemia (hazards ratio 1.15, 95% confidence interval 1.09, 1.21, p < .0001) compared to those in the lower 3 quartiles in a multivariable Cox proportional hazards model adjusting for age, sex, serum iron, soluble transferrin receptor, ferritin, vitamin B12, congestive heart failure, diabetes mellitus, and cancer. Circulating GDF-15 is an independent predictor for the development of anemia in older adults.
Collapse
Affiliation(s)
- Yuko Yamaguchi
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marta Zampino
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Toshiko Tanaka
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | | | - Yusuke Osawa
- Graduate School of Health Management, Keio University, Kanagawa, Japan
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Richard D Semba
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
23
|
Bondu S, Alary AS, Lefèvre C, Houy A, Jung G, Lefebvre T, Rombaut D, Boussaid I, Bousta A, Guillonneau F, Perrier P, Alsafadi S, Wassef M, Margueron R, Rousseau A, Droin N, Cagnard N, Kaltenbach S, Winter S, Kubasch AS, Bouscary D, Santini V, Toma A, Hunault M, Stamatoullas A, Gyan E, Cluzeau T, Platzbecker U, Adès L, Puy H, Stern MH, Karim Z, Mayeux P, Nemeth E, Park S, Ganz T, Kautz L, Kosmider O, Fontenay M. A variant erythroferrone disrupts iron homeostasis in SF3B1-mutated myelodysplastic syndrome. Sci Transl Med 2020; 11:11/500/eaav5467. [PMID: 31292266 DOI: 10.1126/scitranslmed.aav5467] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 03/19/2019] [Accepted: 06/12/2019] [Indexed: 12/21/2022]
Abstract
Myelodysplastic syndromes (MDS) with ring sideroblasts are hematopoietic stem cell disorders with erythroid dysplasia and mutations in the SF3B1 splicing factor gene. Patients with MDS with SF3B1 mutations often accumulate excessive tissue iron, even in the absence of transfusions, but the mechanisms that are responsible for their parenchymal iron overload are unknown. Body iron content, tissue distribution, and the supply of iron for erythropoiesis are controlled by the hormone hepcidin, which is regulated by erythroblasts through secretion of the erythroid hormone erythroferrone (ERFE). Here, we identified an alternative ERFE transcript in patients with MDS with the SF3B1 mutation. Induction of this ERFE transcript in primary SF3B1-mutated bone marrow erythroblasts generated a variant protein that maintained the capacity to suppress hepcidin transcription. Plasma concentrations of ERFE were higher in patients with MDS with an SF3B1 gene mutation than in patients with SF3B1 wild-type MDS. Thus, hepcidin suppression by a variant ERFE is likely responsible for the increased iron loading in patients with SF3B1-mutated MDS, suggesting that ERFE could be targeted to prevent iron-mediated toxicity. The expression of the variant ERFE transcript that was restricted to SF3B1-mutated erythroblasts decreased in lenalidomide-responsive anemic patients, identifying variant ERFE as a specific biomarker of clonal erythropoiesis.
Collapse
Affiliation(s)
- Sabrina Bondu
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France
| | - Anne-Sophie Alary
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France.,Service d'hématologie biologique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Paris Centre-Cochin, Paris 75014, France
| | - Carine Lefèvre
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France.,Laboratoire d'excellence du Globule Rouge GR-Ex, Paris 75015, France
| | - Alexandre Houy
- Institut Curie, PSL Research University, Human Genetics and Oncogenesis, Paris 75005, France
| | - Grace Jung
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Thibaud Lefebvre
- Université de Paris, Paris 75006, France.,Laboratoire d'excellence du Globule Rouge GR-Ex, Paris 75015, France.,INSERM, UMR 1149/ERL CNRS 8252, Centre de Recherches sur l'inflammation, Université de Paris, Paris 75018, France
| | - David Rombaut
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France
| | - Ismael Boussaid
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France
| | - Abderrahmane Bousta
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France
| | - François Guillonneau
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France.,Proteomic platform 3P5, Université de Paris, Paris 75014, France
| | - Prunelle Perrier
- Institut de Recherche en Santé Digestive (IRSD), Université de Toulouse, INSERM U1220, Institut National de la Recherche Agronomique U1416, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse 31024, France
| | - Samar Alsafadi
- Institut Curie, PSL Research University, Department of Translational Research, Paris 75005, France
| | - Michel Wassef
- Institut Curie, PSL Research University, INSERM 934/UMR 3215, Genetics and biology of Development, Paris 75005 France
| | - Raphaël Margueron
- Institut Curie, PSL Research University, INSERM 934/UMR 3215, Genetics and biology of Development, Paris 75005 France
| | - Alice Rousseau
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France
| | - Nathalie Droin
- Institut Gustave Roussy, Genomic platform, Villejuif 94805, France
| | - Nicolas Cagnard
- Université de Paris, Paris 75006, France.,Platform Bioinformatics, Université de Paris, Paris 75015, France
| | - Sophie Kaltenbach
- Université de Paris, Paris 75006, France.,Laboratoire de Génétique, AP-HP, Hôpital Necker, Paris 75015, France
| | - Susann Winter
- Medical Clinic und Policlinic 1, Technische Universität Dresden, Dresden 01307, Germany
| | - Anne-Sophie Kubasch
- Medical Clinic und Policlinic 1, Hematology and Cellular Therapy, University Hospital, Leipzig 04103, Germany
| | - Didier Bouscary
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France.,Service d'Hématologie clinique, AP-HP, Hôpitaux Universitaires Paris Centre-Cochin, Paris 75014, France
| | - Valeria Santini
- MDS unit, Hematology, AOU Careggi, University of Florence, Florence 50134, Italy
| | - Andrea Toma
- Département d'Hématologie, AP-HP, Hôpital Henri-Mondor, Université Paris 12, Créteil 94000, France
| | - Mathilde Hunault
- Service des Maladies du Sang, Centre hospitalo-universitaire, Angers 49100, France
| | | | - Emmanuel Gyan
- Service d'hématologie et thérapie cellulaire, Centre hospitalo-universitaire, CNRS ERL 7001 LNOx, Université de Tours, Tours 37044, France
| | - Thomas Cluzeau
- Côte d'Azur University, CHU of Nice, Hematology department and INSERM U1065, Mediterranean Center of Molecular Medecine, Nice 06204, France
| | - Uwe Platzbecker
- Medical Clinic und Policlinic 1, Hematology and Cellular Therapy, University Hospital, Leipzig 04103, Germany
| | - Lionel Adès
- Université de Paris, Paris 75006, France.,Service d'Hématologie Senior, AP-HP, Hôpital Saint-Louis, Paris 75010, France
| | - Hervé Puy
- Université de Paris, Paris 75006, France.,Laboratoire d'excellence du Globule Rouge GR-Ex, Paris 75015, France.,INSERM, UMR 1149/ERL CNRS 8252, Centre de Recherches sur l'inflammation, Université de Paris, Paris 75018, France
| | - Marc-Henri Stern
- Institut Curie, PSL Research University, INSERM U830, Genetics and biology of cancers, DNA repair and uveal melanoma (D.R.U.M.), Équipe labellisée par la Ligue nationale contre le cancer, Paris 75005, France
| | - Zoubida Karim
- Université de Paris, Paris 75006, France.,Laboratoire d'excellence du Globule Rouge GR-Ex, Paris 75015, France.,INSERM, UMR 1149/ERL CNRS 8252, Centre de Recherches sur l'inflammation, Université de Paris, Paris 75018, France
| | - Patrick Mayeux
- Université de Paris, Paris 75006, France.,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France.,Laboratoire d'excellence du Globule Rouge GR-Ex, Paris 75015, France.,Proteomic platform 3P5, Université de Paris, Paris 75014, France
| | - Elizabeta Nemeth
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Sophie Park
- Département d'Hématologie, Centre Hospitalier Universitaire, Université de Grenoble Alpes, La Tronche 38700, France
| | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Léon Kautz
- Institut de Recherche en Santé Digestive (IRSD), Université de Toulouse, INSERM U1220, Institut National de la Recherche Agronomique U1416, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse 31024, France
| | - Olivier Kosmider
- Université de Paris, Paris 75006, France. .,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France.,Service d'hématologie biologique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Paris Centre-Cochin, Paris 75014, France.,Laboratoire d'excellence du Globule Rouge GR-Ex, Paris 75015, France
| | - Michaëla Fontenay
- Université de Paris, Paris 75006, France. .,Institut Cochin, Département Développement, Reproduction, Cancer, Paris 75014, France.,Institut National de la Santé et de la Recherche médicale (INSERM) U1016, Paris 75014, France.,Centre National de la Recherche Scientifique (CNRS) Unité Mixte de recherche (UMR) 8104, Paris 75014, France.,Service d'hématologie biologique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Paris Centre-Cochin, Paris 75014, France.,Laboratoire d'excellence du Globule Rouge GR-Ex, Paris 75015, France
| |
Collapse
|
24
|
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: 4.0] [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
|
25
|
Cammalleri V, Muscoli S, Versaci F, Romeo F. Periprocedural anemia management in severe aortic stenosis patients undergoing transcatheter aortic valve implantation. J Cardiol 2020; 75:117-123. [DOI: 10.1016/j.jjcc.2019.08.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/29/2019] [Accepted: 08/13/2019] [Indexed: 12/17/2022]
|
26
|
The Influence of Inflammation on Anemia in CKD Patients. Int J Mol Sci 2020; 21:ijms21030725. [PMID: 31979104 PMCID: PMC7036805 DOI: 10.3390/ijms21030725] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/15/2020] [Accepted: 01/19/2020] [Indexed: 02/06/2023] Open
Abstract
Anemia is frequently observed in the course of chronic kidney disease (CKD) and it is associated with diminishing the quality of a patient’s life. It also enhances morbidity and mortality and hastens the CKD progression rate. Patients with CKD frequently suffer from a chronic inflammatory state which is related to a vast range of underlying factors. The results of studies have demonstrated that persistent inflammation may contribute to the variability in Hb levels and hyporesponsiveness to erythropoietin stimulating agents (ESA), which are frequently observed in CKD patients. The understanding of the impact of inflammatory cytokines on erythropoietin production and hepcidin synthesis will enable one to unravel the net of interactions of multiple factors involved in the pathogenesis of the anemia of chronic disease. It seems that anti-cytokine and anti-oxidative treatment strategies may be the future of pharmacological interventions aiming at the treatment of inflammation-associated hyporesponsiveness to ESA. The discovery of new therapeutic approaches towards the treatment of anemia in CKD patients has become highly awaited. The treatment of anemia with erythropoietin (EPO) was associated with great benefits for some patients but not all.
Collapse
|
27
|
Iron Overload in Renal Transplant Patients: The Role of Hepcidin and Erythropoietin. Transplant Proc 2020; 52:169-174. [DOI: 10.1016/j.transproceed.2019.10.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 10/18/2019] [Indexed: 12/29/2022]
|
28
|
Korlesky C, Kling PJ, Pham DQD, Ovasapyan AA, Leyns CA, Weber MB, Coe CL. Cord Blood Erythropoietin and Hepcidin Reflect Lower Newborn Iron Stores due to Maternal Obesity during Pregnancy. Am J Perinatol 2019; 36:511-516. [PMID: 30193381 PMCID: PMC6431561 DOI: 10.1055/s-0038-1669444] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
OBJECTIVE Obesity during pregnancy impedes fetal iron endowment. In adults, both iron depletion and hypoxia stimulate erythropoietin (Epo) production, while hepcidin, the primary iron regulator, is inhibited by Epo and stimulated by obesity. To understand this relationship in fetuses, we investigated obesity, inflammation, and fetal iron status on fetal Epo and hepcidin levels. STUDY DESIGN Epo, hepcidin, C-reactive protein (CRP), and ferritin levels were measured in 201 newborns of 35 to 40 weeks' gestation with historical risk factors for a low fetal iron endowment, including half with maternal obesity. RESULTS Epo was unrelated to fetal size, but Epo was directly related to maternal body mass index (BMI; kg/m2) (p < 0.03) and CRP (p < 0.0005) at delivery. Epo levels were twice as likely to be elevated (≥50 IU/L) while comparing the lowest quartile of ferritin with the upper three quartiles (p < 0.01). Hepcidin was directly related to ferritin (p < 0.001) and indirectly related to maternal BMI (p < 0.015), but BMI became nonsignificant when undergoing multivariate analysis. Hepcidin was unrelated to Epo. CONCLUSION Although some of the fetal responses involving Epo were similar to adults, we did not find a hepcidin-Epo relationship like that of adults, where fetal liver is the site of both hepcidin and Epo production.
Collapse
Affiliation(s)
- Colin Korlesky
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI,University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Pamela J. Kling
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI,University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Daphne Q.-D. Pham
- Department of Biological Sciences, University of Wisconsin-Parkside, Kenosha, WI
| | - Albina A. Ovasapyan
- Department of Biological Sciences, University of Wisconsin-Parkside, Kenosha, WI,,North Central Health Care, Medical College of Wisconsin, Milwaukee, WI
| | - Cheryl A. Leyns
- Department of Biological Sciences, University of Wisconsin-Parkside, Kenosha, WI,,Department of Neurology, Washington University School of Medicine in St. Louis
| | - Morgan B. Weber
- University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI,Department of Orthopedics, Case Western Reserve, Cleveland, OH
| | - Christopher L. Coe
- Department of Psychology, Harlow Center for Biological Psychology and University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
29
|
Abstract
Erythropoiesis is the predominant consumer of iron in humans and other vertebrates. By decreasing the transcription of the gene encoding the iron-regulatory hormone hepcidin, erythropoietic activity stimulates iron absorption, as well as the release of iron from recycling macrophages and from stores in hepatocytes. The main erythroid regulator of hepcidin is erythroferrone (ERFE), synthesized and secreted by erythroblasts in the marrow and extramedullary sites. The production of ERFE is induced by erythropoietin (EPO) and is also proportional to the total number of responsive erythroblasts. ERFE acts on hepatocytes to suppress the production of hepcidin, through an as yet unknown mechanism that involves the bone morphogenetic protein pathway. By suppressing hepcidin, ERFE facilitates iron delivery during stress erythropoiesis but also contributes to iron overload in anemias with ineffective erythropoiesis. Although most of these mechanisms have been defined in mouse models, studies to date indicate that the pathophysiology of ERFE is similar in humans. ERFE antagonists and mimics may prove useful for the prevention and treatment of iron disorders.
Collapse
Affiliation(s)
- Tomas Ganz
- Departments of Medicine and Pathology, David Geffen School of Medicine, UCLA, Los Angeles, USA.
| |
Collapse
|
30
|
Abstract
Hepcidin, the main regulator of iron metabolism, is synthesized and released by hepatocytes in response to increased body iron concentration and inflammation. Deregulation of hepcidin expression is a common feature of genetic and acquired iron disorders: in Hereditary Hemochromatosis (HH) and iron-loading anemias low hepcidin causes iron overload, while in Iron Refractory Iron Deficiency Anemia (IRIDA) and anemia of inflammation (AI), high hepcidin levels induce iron-restricted erythropoiesis. Hepcidin expression in the liver is mainly controlled by the BMP-SMAD pathway, activated in a paracrine manner by BMP2 and BMP6 produced by liver sinusoidal endothelial cells. The BMP type I receptors ALK2 and ALK3 are responsible for iron-dependent hepcidin upregulation and basal hepcidin expression, respectively. Characterization of animal models with genetic inactivation of the key components of the pathway has suggested the existence of two BMP/SMAD pathway branches: the first ALK3 and HH proteins dependent, responsive to BMP2 for basal hepcidin activation, and the second ALK2 dependent, activated by BMP6 in response to increased tissue iron. The erythroid inhibitor of hepcidin Erythroferrone also impacts on the liver BMP-SMAD pathway although its effect is blunted by pathway hyper-activation. The liver BMP-SMAD pathway is required also in inflammation to cooperate with JAK2/STAT3 signaling for full hepcidin activation. Pharmacologic targeting of BMP-SMAD pathway components or regulators may improve the outcome of both genetic and acquired disorders of iron overload and deficiency by increasing or inhibiting hepcidin expression.
Collapse
|
31
|
Santos-Silva A, Ribeiro S, Reis F, Belo L. Hepcidin in chronic kidney disease anemia. VITAMINS AND HORMONES 2019; 110:243-264. [PMID: 30798815 DOI: 10.1016/bs.vh.2019.01.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD) is associated with several complications that worsen with progression of disease; anemia, disturbances in iron metabolism and inflammation are common features. Inflammatory response starts early, releasing pro-inflammatory cytokines, acute phase reactants and hepcidin. Hepcidin production is modulated by several factors, as hypoxia/anemia, erythropoietin and erythropoiesis products, transferrin saturation (TSAT) and liver iron levels, which are altered in CKD. Treatment of CKD anemia is based on pharmaceutical intervention, with erythropoietic stimulating agents and/or iron supplementation; however, in spite of the erythropoietic benefits, this therapy, on a regular basis, involves risks, namely iron overload. To overcome these risks, some therapeutic approaches are under study to target CKD anemia. Considering the actual alerts about risk of iron overload in dialysis patients, inhibition of hepcidin, the central key player in iron homeostasis, could be a pivotal strategy in the management of CKD anemia.
Collapse
Affiliation(s)
- Alice Santos-Silva
- UCIBIO\REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
| | - Sandra Ribeiro
- UCIBIO\REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Flávio Reis
- Institute of Pharmacology & Experimental Therapeutics, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, CNC.IBILI Consortium & CIBB Consortium, University of Coimbra, Coimbra, Portugal
| | - Luís Belo
- UCIBIO\REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
32
|
Abstract
Since its discovery in 2001, there have been a number of important discoveries and findings that have increased our knowledge about the functioning of hepcidin. Hepcidin, the master iron regulator has been shown to be regulated by a number of physiological stimuli and their associated signaling pathways. This chapter will summarize our current understanding of how these physiological stimuli and downstream signaling molecules are involved in hepcidin modulation and ultimately contribute to the regulation of systemic or local iron homeostasis. The signaling pathways and molecules described here have been shown to primarily affect hepcidin at a transcriptional level, but these transcriptional changes correlate with changes in systemic iron levels as well, supporting the functional effects of hepcidin regulation by these signaling pathways.
Collapse
Affiliation(s)
- Gautam Rishi
- The Liver Disease and Iron Disorders Research Group, Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - V Nathan Subramaniam
- The Liver Disease and Iron Disorders Research Group, Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia.
| |
Collapse
|
33
|
Impact of serum hepcidin and inflammatory markers on resistance to erythropoiesis-stimulating therapy in haemodialysis patients. Int Urol Nephrol 2019; 51:325-334. [DOI: 10.1007/s11255-018-2062-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 12/12/2018] [Indexed: 12/19/2022]
|
34
|
Petzer V, Theurl I, Weiss G. Established and Emerging Concepts to Treat Imbalances of Iron Homeostasis in Inflammatory Diseases. Pharmaceuticals (Basel) 2018; 11:E135. [PMID: 30544952 PMCID: PMC6315795 DOI: 10.3390/ph11040135] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 02/06/2023] Open
Abstract
Inflammation, being a hallmark of many chronic diseases, including cancer, inflammatory bowel disease, rheumatoid arthritis, and chronic kidney disease, negatively affects iron homeostasis, leading to iron retention in macrophages of the mononuclear phagocyte system. Functional iron deficiency is the consequence, leading to anemia of inflammation (AI). Iron deficiency, regardless of anemia, has a detrimental impact on quality of life so that treatment is warranted. Therapeutic strategies include (1) resolution of the underlying disease, (2) iron supplementation, and (3) iron redistribution strategies. Deeper insights into the pathophysiology of AI has led to the development of new therapeutics targeting inflammatory cytokines and the introduction of new iron formulations. Moreover, the discovery that the hormone, hepcidin, plays a key regulatory role in AI has stimulated the development of several therapeutic approaches targeting the function of this peptide. Hence, inflammation-driven hepcidin elevation causes iron retention in cells and tissues. Besides pathophysiological concepts and diagnostic approaches for AI, this review discusses current guidelines for iron replacement therapies with special emphasis on benefits, limitations, and unresolved questions concerning oral versus parenteral iron supplementation in chronic inflammatory diseases. Furthermore, the review explores how therapies aiming at curing the disease underlying AI can also affect anemia and discusses emerging hepcidin antagonizing drugs, which are currently under preclinical or clinical investigation.
Collapse
Affiliation(s)
- Verena Petzer
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Igor Theurl
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria.
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
35
|
Vazenmiller D, Ponamaryova O, Muravlyova L, Molotov-Luchanskiy V, Klyuyev D, Bakirova R, Amirbekova Z. The Levels of Hepcidin and Erythropoietin in Pregnant Women with Anemia of Various Geneses. Open Access Maced J Med Sci 2018; 6:2111-2114. [PMID: 30559870 PMCID: PMC6290438 DOI: 10.3889/oamjms.2018.471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/02/2018] [Accepted: 11/03/2018] [Indexed: 11/15/2022] Open
Abstract
AIM: The purpose of the present research was to study the content of erythropoietin and hepcidin in serum in pregnant women with iron deficiency anaemia and anaemia of chronic inflammation. METHODS: The authors examined 98 pregnant women who were observed in LLP (Regional obstetric-gynaecological centre) in Karaganda. The including criteria for pregnant women in the study was the informed consent of the woman to participate in the study. Exclusion criteria were oncological diseases, HIV-infection, tuberculosis, severe somatic pathology, mental illness, drug addiction. The design of the study was by the legislation of the Republic of Kazakhstan, international ethical norms and normative documents of research organisations, approved by the ethics committee of the Karaganda State Medical University. RESULTS: As a result of the study, it was determined that the content of erythropoietin and hepcidin in pregnant women with anemias of different genesis varies ambiguously. In the main group of pregnant women with IDA, the erythropoietin content rises, and the hepcidin level decreases. In pregnant women with ACI, on the contrary, the level of hepcidin increases, and in one subgroup it is significant. However, in pregnant women and with IDA and anemia of chronic inflammation, there is a subgroup of women in whom erythropoietin is either comparable with hepcidin, or their changes are of opposite nature. CONCLUSION: The authors concluded that the obtained data indicate ambiguous changes in the level of erythropoietin and hepcidin in pregnant women with anaemias of various origins. In all likelihood, there are still unaccounted factors affecting the content of these protein-regulators of iron metabolism, which require further definition and interpretation in anaemia of pregnant women.
Collapse
|
36
|
Erythroferrone inhibits the induction of hepcidin by BMP6. Blood 2018; 132:1473-1477. [PMID: 30097509 DOI: 10.1182/blood-2018-06-857995] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/02/2018] [Indexed: 02/06/2023] Open
Abstract
Decreased hepcidin mobilizes iron, which facilitates erythropoiesis, but excess iron is pathogenic in β-thalassemia. Erythropoietin (EPO) enhances erythroferrone (ERFE) synthesis by erythroblasts, and ERFE suppresses hepatic hepcidin production through an unknown mechanism. The BMP/SMAD pathway in the liver is critical for hepcidin control, and we show that EPO suppressed hepcidin and other BMP target genes in vivo in a partially ERFE-dependent manner. Furthermore, recombinant ERFE suppressed the hepatic BMP/SMAD pathway independently of changes in serum and liver iron. In vitro, ERFE decreased SMAD1, SMAD5, and SMAD8 phosphorylation and inhibited expression of BMP target genes. ERFE specifically abrogated the induction of hepcidin by BMP5, BMP6, and BMP7 but had little or no effect on hepcidin induction by BMP2, BMP4, BMP9, or activin B. A neutralizing anti-ERFE antibody prevented ERFE from inhibiting hepcidin induction by BMP5, BMP6, and BMP7. Cell-free homogeneous time-resolved fluorescence assays showed that BMP5, BMP6, and BMP7 competed with anti-ERFE for binding to ERFE. We conclude that ERFE suppresses hepcidin by inhibiting hepatic BMP/SMAD signaling via preferentially impairing an evolutionarily closely related BMP subgroup of BMP5, BMP6, and BMP7. ERFE can act as a natural ligand trap generated by stimulated erythropoiesis to regulate the availability of iron.
Collapse
|
37
|
Yin X, Wu Q, Monga J, Xie E, Wang H, Wang S, Zhang H, Wang ZY, Zhou T, Shi Y, Rogers J, Lin H, Min J, Wang F. HDAC1 Governs Iron Homeostasis Independent of Histone Deacetylation in Iron-Overload Murine Models. Antioxid Redox Signal 2018; 28:1224-1237. [PMID: 29113455 DOI: 10.1089/ars.2017.7161] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
AIMS Iron-overload disorders are common and could lead to significant morbidity and mortality worldwide. Due to limited treatment options, there is a great need to develop novel strategies to remove the excess body iron. To discover potential epigenetic modulator in hepcidin upregulation and subsequently decreasing iron burden, we performed an epigenetic screen. The in vivo effects of the identified compounds were further tested in iron-overload mouse models, including Hfe-/-, Hjv-/-, and hepatocyte-specific Smad4 knockout (Smad4fl/fl;Alb-Cre+) mice. RESULTS Entinostat (MS-275), the clinical used histone deacetylase 1 (HDAC1) inhibitor, was identified the most potent hepcidin agonist. Consistently, Hdac1-deficient mice also presented higher hepcidin levels than wild-type controls. Notably, the long-term treatment with entinostat in Hfe-/- mice significantly alleviated iron overload through upregulating hepcidin transcription. In contrast, entinostat showed no effect on hepcidin expression and iron levels in Smad4fl/fl;Alb-Cre+ mice. Further mechanistic studies revealed that HDAC1 suppressed expression of hepcidin through interacting with SMAD4 rather than deacetylation of SMAD4 or histone-H3 on the hepcidin promoter. INNOVATION The findings uncovered HDAC1 as a novel hepcidin suppressor through complexing with SMAD4 but not deacetylation of either histone 3 or SMAD4. In addition, our study suggested a novel implication of entinostat in treating iron-overload disorders. CONCLUSIONS Based on our results, we conclude that entinostat strongly activated hepcidin in vivo and in vitro. HDAC1 could serve as a novel hepcidin suppressor by binding to SMAD4, effect of which is independent of BMP/SMAD1/5/8 signaling. Antioxid. Redox Signal. 28, 1224-1237.
Collapse
Affiliation(s)
- Xiangju Yin
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
- 2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China
- 3 Institute of Resources and Environment, Henan Polytechnic University , Jiaozuo, China
| | - Qian Wu
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
- 2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China
| | - Jitender Monga
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Enjun Xie
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Hao Wang
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
- 2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China
| | - Shufen Wang
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Huizhen Zhang
- 2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China
| | - Zhan-You Wang
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Tianhua Zhou
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Yujun Shi
- 4 Laboratory of Pathology, West China Hospital, Sichuan University , Chengdu, China
| | - Jack Rogers
- 5 Neurochemistry Laboratory, Departments of Psychiatry and Pediatrics, Massachusetts General Hospital and Harvard Medical School , Charlestown, Massachusetts
| | - Hening Lin
- 6 Department of Chemistry and Chemical Biology, Howard Hughes Medical Institute, Cornell University , Ithaca, New York
| | - Junxia Min
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Fudi Wang
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
- 2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China
| |
Collapse
|
38
|
Karkouti K, Yip P, Chan C, Chawla L, Rao V. Pre-operative anaemia, intra-operative hepcidin concentration and acute kidney injury after cardiac surgery: a retrospective observational study. Anaesthesia 2018. [PMID: 29529338 DOI: 10.1111/anae.14274] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Acute kidney after cardiac surgery is more common in anaemic patients, whereas haemolysis during cardiopulmonary bypass may lead to iron-induced renal injury. Hepcidin promotes iron sequestration by macrophages: hepcidin concentration is reduced by anaemia and increased by inflammation. We analysed the associations in 525 patients between pre-operative anaemia (haemoglobin < 130 g.l-1 in men and < 120 g.l-1 in women), intra-operative hepcidin concentration and acute kidney injury (dialysis or > 26.4 μmol.l-1 or > 50% creatinine increase during the first two days after cardiac surgery. Rates of pre-operative anaemia and postoperative kidney injury were 109/525 (21%) and 36/525 (7%), respectively. The median (IQR [range]) intra-operative hepcidin concentration was 20 (10-33 [0-125]) μg.l-1 and was lower in anaemic patients than those who were not: 15 (4-28 [0-125]) μg.l-1 vs. 21 (12-33 [0-125]) μg.l-1 , respectively, p = 0.002. Four variables were independently associated with postoperative kidney injury, for which the beta-coefficients (SE) were: minutes on cardiopulmonary bypass, 0.016 (0.004), p < 0.001; intra-operative hepcidin concentration, 0.032 (0.008), p < 0.001; pre-operative anaemia, 1.97 (0.56), p < 0.001; and Cleveland clinic risk score, 0.88 (0.35), p = 0.005. Contrary to generally increased rates of kidney injury in patients with higher hepcidin concentrations, rates of kidney injury in anaemic patients were lower in patients with higher hepcidin concentrations, beta-coefficient (SE) -0.037 (0.01), p = 0.007. In cardiac surgical patients the rate of postoperative acute kidney injury predicted by the Cleveland risk score might be adjusted for pre-operative anaemia and intra-operative cardiopulmonary bypass time and hepcidin concentration. Pre-operative correction of anaemia, reduction in intra-operative bypass time and modification of iron homeostasis and hepcidin concentration might reduce acute kidney injury.
Collapse
Affiliation(s)
- K Karkouti
- Department of Anesthesia and Pain Management, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, University of Toronto, ON, Canada
| | - P Yip
- Department of Clinical Biochemistry, Toronto General Hospital, University Health Network, University of Toronto, ON, Canada
| | - C Chan
- Division of Nephrology, Department of Medicine, Toronto General Hospital, University Health Network, University of Toronto, ON, Canada
| | - L Chawla
- Department of Anesthesiology and Critical Care Medicine, The George Washington University, Washington, DC, USA.,La Jolla Pharmaceutical Company, San Diego, CA, USA
| | - V Rao
- Division of Cardiac Surgery, Department of Surgery, Toronto General Hospital, University Health Network, University of Toronto, ON, Canada
| |
Collapse
|
39
|
Abstract
The hepatic iron-regulatory hormone hepcidin and its receptor, the cellular iron exporter ferroportin, constitute a feedback-regulated mechanism that maintains adequate plasma concentrations of iron-transferrin for erythropoiesis and other functions, ensures sufficient iron stores, and avoids iron toxicity and iron-dependent microbial pathogenesis. In chronic kidney disease, inflammation and impaired renal clearance increase plasma hepcidin, inhibiting duodenal iron absorption and sequestering iron in macrophages. These effects of hepcidin can cause systemic iron deficiency, decreased availability of iron for erythropoiesis, and resistance to endogenous and exogenous erythropoietin. Together with impaired renal production of erythropoietin, hepcidin-mediated iron restriction contributes to anemia of chronic kidney disease.
Collapse
Affiliation(s)
- Tomas Ganz
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA.
| | - Elizabeta Nemeth
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
40
|
Collins JF, Flores SR, Wang X, Anderson GJ. Mechanisms and Regulation of Intestinal Iron Transport. PHYSIOLOGY OF THE GASTROINTESTINAL TRACT 2018:1451-1483. [DOI: 10.1016/b978-0-12-809954-4.00060-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
41
|
|
42
|
Fischli S, von Wyl V, Trummler M, Konrad D, Wueest S, Ruefer A, Heering K, Streuli R, Steuer C, Bernasconi L, Recher M, Henzen C. Iron metabolism in patients with Graves' hyperthyroidism. Clin Endocrinol (Oxf) 2017; 87:609-616. [PMID: 28833367 DOI: 10.1111/cen.13450] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/14/2017] [Accepted: 08/13/2017] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Graves' hyperthyroidism (GH) interferes with iron metabolism and elevates ferritin. The precise mechanisms remain unclear. The influence of thyroid hormones on the synthesis/regulation of hepcidin, an important regulator of iron metabolism, remains uncharacterized. DESIGN Prospective observational study. PATIENTS We included patients (n = 31) with new-onset and untreated GH. MEASUREMENTS Laboratory parameters indicative of iron metabolism (ferritin, transferrin, hepcidin), inflammatory markers/cytokines and smoking status were assessed at the diagnosis of GH (T0) and at euthyroidism (T1) in the same patients using multivariable analyses. Hepcidin was measured by mass spectrometry (hepcidinMS ) and ELISA (hepcidinEL ). The impact of T3 on hepatic hepcidin expression was studied in a cell culture model using HepG2 cells. RESULTS Median ferritin levels were significantly lower and transferrin significantly higher at T1 than at T0. HepcidinMS levels were lower in males and females at T1 (statistically significant in males only). No statistically significant difference in hepcidinEL was detected between T0 and T1. Plasma levels of inflammatory markers (high-sensitive CRP, procalcitonin) and cytokines (interleukin 6, interleukin 1ß, tumour necrosis factor α) were not different between T0 and T1. Smokers tended to have lower fT3 and fT4 at T0 than nonsmoking GH patients. T3 significantly induced hepcidin mRNA expression in HepG2 cells. CONCLUSIONS Iron metabolism in patients with GH undergoes dynamic changes in patients with GH that resemble an acute-phase reaction. Inflammatory parameters and cytokines were unaffected by thyroid status. Gender and smoking status had an impact on ferritin, hepcidin and thyroid hormones.
Collapse
Affiliation(s)
- Stefan Fischli
- Division of Endocrinology and Diabetes, Luzerner Kantonsspital, Luzern, Switzerland
| | - Viktor von Wyl
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Michael Trummler
- Department of Clinical Chemistry, Luzerner Kantonsspital, Luzern, Switzerland
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Axel Ruefer
- Division of Hematology, Luzerner Kantonsspital, Luzern, Switzerland
| | - Kerstin Heering
- Division of Endocrinology and Diabetes, Luzerner Kantonsspital, Luzern, Switzerland
| | - Regina Streuli
- Division of Endocrinology and Diabetes, Luzerner Kantonsspital, Luzern, Switzerland
| | - Christian Steuer
- Institute of Laboratory Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Luca Bernasconi
- Institute of Laboratory Medicine, Kantonsspital Aarau, Aarau, Switzerland
| | - Mike Recher
- Immunodeficiency Clinic, Medical Outpatient Clinic and Department of Biomedicine, University Hospital Basel and University Basel, Basel, Switzerland
| | - Christoph Henzen
- Division of Endocrinology and Diabetes, Luzerner Kantonsspital, Luzern, Switzerland
| |
Collapse
|
43
|
Biggar P, Kim GH. Treatment of renal anemia: Erythropoiesis stimulating agents and beyond. Kidney Res Clin Pract 2017; 36:209-223. [PMID: 28904872 PMCID: PMC5592888 DOI: 10.23876/j.krcp.2017.36.3.209] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/02/2017] [Accepted: 07/12/2017] [Indexed: 12/18/2022] Open
Abstract
Anemia, complicating the course of chronic kidney disease, is a significant parameter, whether interpreted as subjective impairment or an objective prognostic marker. Renal anemia is predominantly due to relative erythropoietin (EPO) deficiency. EPO inhibits apoptosis of erythrocyte precursors. Studies using EPO substitution have shown that increasing hemoglobin (Hb) levels up to 10–11 g/dL is associated with clinical improvement. However, it has not been unequivocally proven that further intensification of erythropoiesis stimulating agent (ESA) therapy actually leads to a comprehensive benefit for the patient, especially as ESAs are potentially associated with increased cerebro-cardiovascular events. Recently, new developments offer interesting options not only via stimulating erythropoeisis but also by employing additional mechanisms. The inhibition of activin, a member of the transforming growth factor superfamily, has the potential to correct anemia by stimulating liberation of mature erythrocyte forms and also to mitigate disturbed mineral and bone metabolism as well. Hypoxia-inducible factor prolyl hydroxylase inhibitors also show pleiotropic effects, which are at the focus of present research and have the potential of reducing mortality. However, conventional ESAs offer an extensive body of safety evidence, against which the newer substances should be measured. Carbamylated EPO is devoid of Hb augmenting effects whilst exerting promising tissue protective properties. Additionally, the role of hepcidin antagonists is discussed. An innovative new hemodialysis blood tube system, reducing blood contact with air, conveys a totally different and innocuous option to improve renal anemia by reducing mechanical hemolysis.
Collapse
Affiliation(s)
- Patrick Biggar
- Department of Nephrology, Klinikum Coburg, GmbH, Coburg, Germany.,KfH Kidney Centre, Coburg, Germany
| | - Gheun-Ho Kim
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
44
|
Goto K, Kasai N, Kojima C, Ishibashi A. Postexercise serum hepcidin response to repeated sprint exercise under normoxic and hypoxic conditions. Appl Physiol Nutr Metab 2017; 43:221-226. [PMID: 28961407 DOI: 10.1139/apnm-2017-0418] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We determined the effects of repeated sprint exercise under normoxic and hypoxic conditions on serum hepcidin levels. Ten male athletes (age: 20.9 ± 0.3 years; height: 175.7 ± 6.0 cm; weight: 67.3 ± 6.3 kg) performed 2 exercise trials under normoxic (NOR; fraction of inspiratory oxygen (FiO2): 20.9%) or hypoxic conditions (HYPO; FiO2: 14.5%). The exercise consisted of 3 sets of 5 × 6 s of maximal pedaling (30-s rest periods between sprints, 10-min rest periods between sets). Blood samples were collected before exercise, immediately after exercise, and 1 and 3 h after exercise. Serum hepcidin levels were significantly elevated after exercise in both trials (both P < 0.01), with no significant difference between the trials. The postexercise blood lactate levels were significantly higher in the HYPO than the NOR (P < 0.05). Both trials caused similar increases in plasma interleukin-6 and serum iron levels (P < 0.001), with no significant difference between the trials. A significant interaction (trial × time) was apparent in terms of serum erythropoietin (EPO) levels (P = 0.003). The EPO level was significantly higher in the HYPO than the NOR at 3 h after exercise (P < 0.05). In conclusion, repeated sprint exercise significantly increased serum hepcidin levels to similar extent in 2 trials, despite differences in the inspired oxygen concentrations during both the exercise and the 3-h postexercise period.
Collapse
Affiliation(s)
- Kazushige Goto
- a Graduate School of Sports and Health Science, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Nobukazu Kasai
- a Graduate School of Sports and Health Science, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Chihiro Kojima
- a Graduate School of Sports and Health Science, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Aya Ishibashi
- a Graduate School of Sports and Health Science, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan.,b Department of Sports Science, Japan Institute of Sports Science, Kitaku, Tokyo, 525-8577, Japan
| |
Collapse
|
45
|
[The role of erythroferrone in iron metabolism: From experimental results to pathogenesis]. Rev Med Interne 2017; 39:178-184. [PMID: 28666715 DOI: 10.1016/j.revmed.2017.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/19/2017] [Accepted: 05/30/2017] [Indexed: 11/22/2022]
Abstract
In case of erythropoiesis, body iron needs to increase to enable the production of new red blood cells. In the 1950s, the observation of an increased digestive iron absorption in the case of phlebotomies had led to propose the existence of an "erythroid factor", which regulate the availability of iron for erythropoiesis in this situation. The factor regulating iron stores has been identified in 2000 to be hepcidin. Recently, in 2014, a new factor was discovered, which regulates iron metabolism, independently of iron stores and responds to the increased requirements for iron after stimulation of erythropoiesis by erythropoietin. This factor has been referred to as erythroferrone. Thus, the regulation of iron stores depends on hepcidin, while the adaptation mechanisms of iron availability in case of anemia, are mediated by an erythroid factor that could be erythroferrone. This review summarizes the current knowledge on the role of erythroferrone in iron metabolism, starting from experimental results, obtained mainly on mouse models, and related to iron overload in β-thalassemia, iron disturbances during anemia of chronic diseases and chronic renal failure. These results will have to be compared with those obtained in humans, as soon as a reliable assay for human erythroferrone is available. From a clinical point of view, erythroferrone could become a useful biological marker of iron metabolism and a therapeutic target.
Collapse
|
46
|
Papanikolaou G, Pantopoulos K. Systemic iron homeostasis and erythropoiesis. IUBMB Life 2017; 69:399-413. [DOI: 10.1002/iub.1629] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/16/2017] [Indexed: 01/01/2023]
Affiliation(s)
- George Papanikolaou
- Department of Nutrition and DieteticsSchool of Health Science and Education, Harokopion UniversityAthens Greece
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research and Department of MedicineMcGill UniversityMontreal Quebec Canada
| |
Collapse
|
47
|
Abstract
Anemia is one of the main comorbidities related to chronic kidney disease (CKD). Until the advent of erythropoiesis stimulating agents (ESA), endogenous erythropoietin deficiency has been thought to be the main culprit of anemia in CKD patients. The use of ESAs has shed new light on the physiology of CKD anemia, where iron homeostasis plays an increasingly important role. Disorders of iron homeostasis occurring in CKD turn the anemia management in those patients into a complex multifactorial therapeutic task, where ESA and Iron dose must be properly balanced to achieve the desired outcome without exposing the patients to the risk of serious adverse events. This review covers diagnostic markers traditionally used for quantifying iron status in CKD patients, such as serum ferritin and transferrin saturation, new ones, such as reticulocyte hemoglobin content and percent hypochromic red cells (HRC), as well as experimental ones, such as hepcidin and soluble transferrin receptor (sTfR). Each marker is presented in terms of their diagnostic performance, followed by biological and analytical variability data. Advantages and disadvantages of each marker are briefly discussed. Although serum ferritin and transferrin saturation are easily available, they exhibit large biological variability and require caution when used for diagnosing iron status in CKD patients. Reticulocyte hemoglobin content and the percentage of HRC are more powerful, but their widespread use is hampered by the issue of sample stability in storage. sTfR and hepcidin show promise, but require further investigation as well as the development of standardized, low-cost assay platforms.
Collapse
Affiliation(s)
- Adam E Gaweda
- Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
48
|
|
49
|
Ichinomiya K, Maruyama K, Inoue T, Koizumi A, Inoue F, Fukuda K, Yamazaki Y, Arakawa H. Perinatal Factors Affecting Serum Hepcidin Levels in Low-Birth-Weight Infants. Neonatology 2017; 112:180-186. [PMID: 28601871 DOI: 10.1159/000473871] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/30/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND Hepcidin, an iron-regulatory hormone, plays a key role in preventing iron overload. Few studies have investigated the regulation of hepcidin in low-birth-weight (LBW) infants who are vulnerable to iron imbalance. OBJECTIVES To identify perinatal factors associated with serum hepcidin levels in LBW infants. METHODS Ninety-two LBW infants with a median gestational age (GA) of 32.6 weeks and birth weight of 1,587 g were prospectively enrolled. Serum hepcidin-25 (Hep25) levels were measured from umbilical cord blood using liquid chromatography-tandem mass spectrometry. The relationship between Hep25 levels and prematurity or other possible hepcidin-regulatory factors was evaluated. RESULTS The median Hep25 level was 7.3 ng/mL (interquartile range: 2.85-16.38). log(Hep25) correlated with birth weight (r = 0.229, p = 0.028), log(interleukin-6 [IL-6]) (r = 0.408, p < 0.001), log(erythropoietin) (r = -0.302, p = 0.004), transferrin saturation (r = 0.29, p = 0.005), soluble transferrin receptor (r = -0.500, p < 0.001), and log(ferritin) (r = 0.696, p < 0.001). Serum iron and hemoglobin levels did not correlate with log(Hep25). Hep25 levels were higher among infants with chorioamnionitis and infants born vaginally and lower among infants born to mothers with pregnancy-induced hypertension than among infants without the respective characteristics. Stepwise multiple linear regression analysis confirmed the significant association of log(Hep25) with GA, log(IL-6), log(erythropoietin), and soluble transferrin receptor. CONCLUSIONS Among LBW infants, GA, IL-6, erythropoietin, and soluble transferrin receptor were associated with Hep25 levels. Therefore, prematurity, inflammation, hypoxia, and erythropoietic activity may be important perinatal factors that affect hepcidin levels.
Collapse
Affiliation(s)
- Kenji Ichinomiya
- Department of Neonatology, Gunma Children's Medical Center, Gunma, Japan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Gunes T, Yildirim S, Ozdemir A, Gokahmetoglu S, Korkut S, Ozturk MA, Kurtoglu S. Maternal and cord blood hepcidin levels based on gestational weeks in term and preterm infants. PEDIATRIC HEMATOLOGY ONCOLOGY JOURNAL 2016. [DOI: 10.1016/j.phoj.2016.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|