1
|
Medjbeur T, Personnaz J, Kautz L. [FGL1 : a new target for the treatment of anemia ?]. Med Sci (Paris) 2024; 40:722-724. [PMID: 39450956 DOI: 10.1051/medsci/2024123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024] Open
Affiliation(s)
- Thanina Medjbeur
- IRSD, Université de Toulouse, Inserm, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS) Toulouse France
| | - Jean Personnaz
- IRSD, Université de Toulouse, Inserm, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS) Toulouse France
| | - Léon Kautz
- IRSD, Université de Toulouse, Inserm, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS) Toulouse France
| |
Collapse
|
2
|
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
|
3
|
Mendoza E, Duque X, Reyes-Maldonado E, Hernández-Franco JI, Martínez-Andrade G, Vilchis-Gil J, Martinez H, Morán S. Serum hepcidin recalibrated values in Mexican schoolchildren by demographic characteristics, nutritional and infection/inflammation status. Ann Hematol 2024; 103:3979-3986. [PMID: 39039174 DOI: 10.1007/s00277-024-05889-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Hepcidin production is regulated by iron concentration, erythropoietic activity, and inflammation. There is no reference method for determining its levels, but results obtained through various methods strongly correlate and can be compared using recalibration equations. OBJECTIVE To describe recalibrated serum hepcidin values at different percentiles in schoolchildren, considering age, sex, inflammatory processes, H. pylori infection, and iron status. METHODS Secondary analysis of data incorporating information on inflammation, H. pylori infection, and iron status of 349 schoolchildren. Hepcidin analysis was performed using a competitive ELISA, and recalibrated hepcidin values were calculated using the inverse of the linear regression model equation obtained by van der Vorm et al. Results: Recalibrated hepcidin values were lower than non-calibrated values. In schoolchildren without infection/inflammation and without iron deficiency, recalibrated values at the 50th percentile (25th-75th) were 4.89 ng/mL (2.68-8.42). For schoolchildren without infection/inflammation but with iron deficiency, recalibrated values were 2.34 ng/mL (1.10-6.58), the lowest hepcidin values observed. The highest values were found in the group with infection/inflammation, regardless of iron deficiency status. CONCLUSIONS Recalibrated hepcidin values were lower than non-calibrated values. The highest values were observed in schoolchildren with infectious or inflammatory processes, and the lowest values were observed in schoolchildren with iron deficiency but only in the absence of infectious or inflammatory processes. Using recalibrated hepcidin values allows comparison between data obtained using different analytical methods.
Collapse
Affiliation(s)
- Eugenia Mendoza
- Infectious Diseases Research Unit, Mexican Social Security Institute, Av. Cuauhtemoc No. 330, Col. Doctores, Del. Cuauhtemoc, Mexico City, CP 06720, Mexico
| | - Ximena Duque
- Infectious Diseases Research Unit, Mexican Social Security Institute, Av. Cuauhtemoc No. 330, Col. Doctores, Del. Cuauhtemoc, Mexico City, CP 06720, Mexico.
| | - Elba Reyes-Maldonado
- Department of Hematopathology, National Polytechnic Institute, National School of Biological Sciences, Mexico City, 01135, Mexico
| | | | - Gloria Martínez-Andrade
- Academic Area of Nutrition, Institute of Health Sciences, Autonomous University of the State of Hidalgo, Pachuca Hidalgo, 42039, Mexico
| | - Jenny Vilchis-Gil
- Hospital Infantil de México "Federico Gomez", Mexico City, 06720, Mexico
| | - Homero Martinez
- Hospital Infantil de México "Federico Gomez", Mexico City, 06720, Mexico
- Global Technical Services-NTEAM, Nutrition International, Ottawa, ON, K2P 2K3, Canada
| | - Segundo Morán
- Gastroenterology Research Laboratory, Mexican Social Security Institute, Mexico City, 06720, Mexico
| |
Collapse
|
4
|
Babar S, Saboor M. Erythroferrone in focus: emerging perspectives in iron metabolism and hematopathologies. BLOOD SCIENCE 2024; 6:e00198. [PMID: 39027903 PMCID: PMC11254117 DOI: 10.1097/bs9.0000000000000198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024] Open
Abstract
Beyond its core role in iron metabolism, erythroferrone (ERFE) has emerged as a key player with far-reaching implications in various hematologic disorders. Its regulatory effect on hepcidin underlines its significance in conditions characterized by disrupted iron homeostasis. In β-thalassemia and myelodysplastic syndromes, its dysregulation intricately contributes to the clinical challenges of anemia and iron overload which highlights its potential as a therapeutic target. In anemia of chronic disease and iron deficiency anemia, ERFE presents a unique profile. In chronic kidney disease (CKD), the intricate interplay between ERFE, erythropoietin, and hepcidin undergoes dysregulation, contributing to the complex iron imbalance characteristic of this condition. Recent research suggests that ERFE plays a multifaceted role in restoring iron balance in CKD, beyond simply suppressing hepcidin production. The potential to modulate ERFE activity offers a novel approach to treating a spectrum of disorders associated with iron dysregulation. As our understanding of ERFE continues to evolve, it is poised to become a key focus in the development of targeted treatments, making it an exciting and dynamic area of ongoing research. Modulating ERFE activity presents a groundbreaking approach to treat iron dysregulation in conditions like iron deficiency anemia, thalassemia, and hemochromatosis. As new research unveils its intricate roles, ERFE has rapidly emerged as a key target for developing targeted therapies like ERFE agonists and antagonists. With promising studies underway, this dynamic field holds immense potential to improve patient outcomes, reduce complications, and offer personalized treatment options in hematology research. This comprehensive overview of ERFE's role across various conditions underscores its pivotal function in iron metabolism and associated pathologies.
Collapse
Affiliation(s)
- Sadia Babar
- Baqai Institute of Hematology, Baqai Medical University, Karachi, Pakistan
- Baqai Institute of Medical Technology, Baqai Medical University, Karachi, Pakistan
| | - Muhammad Saboor
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
5
|
Boucher AA, Dayton VJ, Pratt AR, Nassar NN, Elgammal Y, Kalfa TA. Three-generation female cohort with macrocytic anemia and iron overload. Am J Hematol 2024. [PMID: 39329459 DOI: 10.1002/ajh.27489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Affiliation(s)
- Alexander A Boucher
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Vanessa J Dayton
- Laboratory Medicine and Pathology, Hennepin County Medical Center, Hennepin Healthcare Research Institute, Minneapolis, Minnesota, USA
| | - Annaliisa R Pratt
- Laboratory Medicine and Pathology, Hennepin County Medical Center, Hennepin Healthcare Research Institute, Minneapolis, Minnesota, USA
| | - Nicolas N Nassar
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati Medical School, Cincinnati, Ohio, USA
| | - Yasmin Elgammal
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Theodosia A Kalfa
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati Medical School, Cincinnati, Ohio, USA
| |
Collapse
|
6
|
Pilo F, Angelucci E. Vamifeport: Monography of the First Oral Ferroportin Inhibitor. J Clin Med 2024; 13:5524. [PMID: 39337010 PMCID: PMC11432582 DOI: 10.3390/jcm13185524] [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: 08/15/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Over the last few years, several mechanisms that are involved in congenital diseases characterized by ineffective erythropoiesis have been described. Therefore, multiple new target drugs are being developed in preclinical models against the main regulators of normal erythropoiesis. Above all, the key mechanism that regulates systemic iron homeostasis, represented by the hepcidin-ferroportin axis, is considered to be the target for new therapies. The main hypothesis is that iron restriction, through blocking ferroportin (the unique iron transporter in mammals) in such diseases, ameliorates erythropoiesis. The action of vamifeport is different from the currently approved drugs in this setting since it acts straight on the ferroportin-hepcidin axis. The data presented in the sickle cell disease (SCD) Townes mouse model showed a preclinical proof-of-concept for the efficacy of oral ferroportin inhibitor. Vamifeport reduced hemoglobin concentration in red blood cells (RBCs) and diminished intravascular hemolysis and inflammation, improving hemodynamics and preventing vascular occlusive crises. On this basis, clinical trials were commenced in patients with SCD, non-transfusion-dependent (NTD) thalassemia and transfusion-dependent (TD) thalassemia. Preliminary data in NTD thalassemic patients also confirm the safety and efficacy in decreasing iron level. In conclusion, vamifeport represents a new option in the panorama of drugs targeting the hepcidin-ferroportin axis, but its efficacy is still under investigation as a single agent.
Collapse
Affiliation(s)
- Federica Pilo
- Hematology and Transplant Center, Azienda Ospedaliera Brotzu, 09121 Cagliari, Italy
| | - Emanuele Angelucci
- Hematology and Cellular Therapy Center, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy;
| |
Collapse
|
7
|
Nazarov K, Perik-Zavodskii R, Perik-Zavodskaia O, Alrhmoun S, Volynets M, Shevchenko J, Sennikov S. Acute blood loss in mice forces differentiation of both CD45-positive and CD45-negative erythroid cells and leads to a decreased CCL3 chemokine production by bone marrow erythroid cells. PLoS One 2024; 19:e0309455. [PMID: 39231178 PMCID: PMC11373861 DOI: 10.1371/journal.pone.0309455] [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: 05/20/2024] [Accepted: 08/09/2024] [Indexed: 09/06/2024] Open
Abstract
Hemorrhage, a condition that accompanies most physical trauma cases, remains an important field of study, a field that has been extensively studied in the immunological context for myeloid and lymphoid cells, but not as much for erythroid cells. In this study, we studied the immunological response of murine erythroid cells to acute blood loss using flow cytometry, NanoString immune transcriptome profiling, and BioPlex cytokine secretome profiling. We observed that acute blood loss forces the differentiation of murine erythroid cells in both bone marrow and spleen and that there was an up-regulation of several immune response genes, in particular pathogen-associated molecular pattern sensing gene Clec5a in post-acute blood loss murine bone marrow erythroid cells. We believe that the up-regulation of the Clec5a gene in bone marrow erythroid cells could help bone marrow erythroid cells detect and eliminate pathogens with the help of reactive oxygen species and antimicrobial proteins calprotectin and cathelicidin, the genes of which (S100a8, S100a9, and Camp) dominate the expression in bone marrow erythroid cells of mice.
Collapse
Affiliation(s)
- Kirill Nazarov
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Roman Perik-Zavodskii
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Olga Perik-Zavodskaia
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Saleh Alrhmoun
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Marina Volynets
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Julia Shevchenko
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Sergey Sennikov
- Laboratory of molecular immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
- Department of Immunology, Zelman Institute for Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
8
|
Musallam KM, Sheth S, Cappellini MD, Forni GL, Maggio A, Taher AT. Anemia and iron overload as prognostic markers of outcomes in β-thalassemia. Expert Rev Hematol 2024; 17:631-642. [PMID: 39037857 DOI: 10.1080/17474086.2024.2383420] [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: 06/04/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Ineffective erythropoiesis and subsequent anemia as well as primary and secondary (transfusional) iron overload are key drivers for morbidity and mortality outcomes in patients with β-thalassemia. AREAS COVERED In this review, we highlight evidence from observational studies evaluating the association between measures of anemia and iron overload versus outcomes in both non-transfusion-dependent and transfusion-dependent forms of β-thalassemia. EXPERT OPINION Several prognostic thresholds have been identified with implications for patient management. These have also formed the basis for the design of novel therapy clinical trials by informing eligibility and target endpoints. Still, several data gaps persist in view of the challenge of assessing prospective long-term outcomes in a chronic disease. Pooling insights on the prognostic value of different measures of disease mechanism will be key to design future scoring systems that can help optimize patient management.
Collapse
Affiliation(s)
- Khaled M Musallam
- Center for Research on Rare Blood Disorders (CR-RBD), Burjeel Medical City, Abu Dhabi, United Arab Emirates
- Division of Hematology/Oncology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Sujit Sheth
- Division of Hematology/Oncology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Maria Domenica Cappellini
- Department of Clinical Sciences and Community, University of Milan, Ca' Granda Foundation IRCCS Maggiore Policlinico Hospital, Milan, Italy
| | | | - Aurelio Maggio
- Campus of Haematology Franco and Piera Cutino, AOOR Villa Sofia-V. Cervello, Palermo, Italy
| | - Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
9
|
Pinto VM, Mazzi F, De Franceschi L. Novel therapeutic approaches in thalassemias, sickle cell disease, and other red cell disorders. Blood 2024; 144:853-866. [PMID: 38820588 DOI: 10.1182/blood.2023022193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/16/2024] [Accepted: 05/19/2024] [Indexed: 06/02/2024] Open
Abstract
ABSTRACT In this last decade, a deeper understanding of the pathophysiology of hereditary red cell disorders and the development of novel classes of pharmacologic agents have provided novel therapeutic approaches to thalassemias, sickle cell disease (SCD), and other red cell disorders. Here, we analyze and discuss the novel therapeutic options according to their targets, taking into consideration the complex process of erythroid differentiation, maturation, and survival of erythrocytes in the peripheral circulation. We focus on active clinical exploratory and confirmatory trials on thalassemias, SCD, and other red cell disorders. Beside β-thalassemia and SCD, we found that the development of new therapeutic strategies has allowed for the design of clinic studies for hereditary red cell disorders still lacking valuable therapeutic alternative such as α-thalassemias, congenital dyserythropoietic anemia, or Diamond-Blackfan anemia. In addition, reduction of heme synthesis, which can be achieved by the repurposed antipsychotic drug bitopertin, might affect not only hematological disorders but multiorgan diseases such as erythropoietic protoporphyria. Finally, our review highlights the current state of therapeutic scenarios, in which multiple indications targeting different red cell disorders are being considered for a single agent. This is a welcome change that will hopefully expand therapeutic option for patients affected by thalassemias, SCD, and other red cell disorders.
Collapse
Affiliation(s)
- Valeria Maria Pinto
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Centro della Microcitemia, Anemie Congenite e Dismetabolismo del Ferro, Ente Ospedaliero Ospedali Galliera, Genoa, Italy
| | - Filippo Mazzi
- Department of Medicine, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Lucia De Franceschi
- Department of Medicine, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy
- Department of Engineering for Innovative Medicine, University of Verona, Verona, Italy
| |
Collapse
|
10
|
Zhao Y, Zhu L, Shi D, Gao J, Fan M. Key Genes FECH and ALAS2 under Acute High-Altitude Exposure: A Gene Expression and Network Analysis Based on Expression Profile Data. Genes (Basel) 2024; 15:1075. [PMID: 39202434 PMCID: PMC11353374 DOI: 10.3390/genes15081075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/08/2024] [Accepted: 08/11/2024] [Indexed: 09/03/2024] Open
Abstract
High-altitude acclimatization refers to the physiological adjustments and adaptation processes by which the human body gradually adapts to the hypoxic conditions of high altitudes after entering such environments. This study analyzed three mRNA expression profile datasets from the GEO database, focusing on 93 healthy residents from low altitudes (≤1400 m). Peripheral blood samples were collected for analysis on the third day after these individuals rapidly ascended to higher altitudes (3000-5300 m). The analysis identified significant differential expression in 382 genes, with 361 genes upregulated and 21 downregulated. Further, gene ontology (GO) annotation analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis indicated that the top-ranked enriched pathways are upregulated, involving blood gas transport, erythrocyte development and differentiation, and heme biosynthetic process. Network analysis highlighted ten key genes, namely, SLC4A1, FECH, EPB42, SNCA, GATA1, KLF1, GYPB, ALAS2, DMTN, and GYPA. Analysis revealed that two of these key genes, FECH and ALAS2, play a critical role in the heme biosynthetic process, which is pivotal in the development and maturation of red blood cells. These findings provide new insights into the key gene mechanisms of high-altitude acclimatization and identify potential biomarkers and targets for personalized acclimatization strategies.
Collapse
Affiliation(s)
- Yifan Zhao
- School of Information Science and Engineering, Lanzhou University, Lanzhou 730000, China;
| | - Lingling Zhu
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China;
| | - Dawei Shi
- School of Automation, Beijing Institute of Technology, Beijing 100850, China;
| | - Jiayue Gao
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China;
| | - Ming Fan
- School of Information Science and Engineering, Lanzhou University, Lanzhou 730000, China;
| |
Collapse
|
11
|
Zhang H, Liu R, Fang Z, Nie L, Ma Y, Sun F, Mei J, Song Z, Ginzburg YZ, Liu J, Chen H. Mitoxantrone ameliorates ineffective erythropoiesis in a β-thalassemia intermedia mouse model. Blood Adv 2024; 8:4017-4024. [PMID: 38861356 PMCID: PMC11339037 DOI: 10.1182/bloodadvances.2024012679] [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: 01/17/2024] [Revised: 05/15/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024] Open
Abstract
ABSTRACT β-thalassemia is a condition characterized by reduced or absent synthesis of β-globin resulting from genetic mutations, leading to expanded and ineffective erythropoiesis. Mitoxantrone has been widely used clinically as an antitumor agent considering its ability to inhibit cell proliferation. However, its therapeutic effect on expanded and ineffective erythropoiesis in β-thalassemia is untested. We found that mitoxantrone decreased α-globin precipitates and ameliorated anemia, splenomegaly, and ineffective erythropoiesis in the HbbTh3/+ mouse model of β-thalassemia intermedia. The partially reversed ineffective erythropoiesis is a consequence of effects on autophagy as mitochondrial retention and protein levels of mTOR, P62, and LC3 in reticulocytes decreased in mitoxantrone-treated HbbTh3/+ mice. These data provide significant preclinical evidence for targeting autophagy as a novel therapeutic approach for β-thalassemia.
Collapse
Affiliation(s)
- Haihang Zhang
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Rui Liu
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Zheng Fang
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Ling Nie
- Xiangya Hospital, Central South University, Changsha, China
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, the First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
| | - Fei Sun
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, the First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
| | - Jingjing Mei
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, the First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
| | - Zhiyin Song
- College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yelena Z. Ginzburg
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jing Liu
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Huiyong Chen
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| |
Collapse
|
12
|
Xu P, Wong RSM, Yan X. Early erythroferrone levels can predict the long-term haemoglobin responses to erythropoiesis-stimulating agents. Br J Pharmacol 2024; 181:2833-2850. [PMID: 38653449 DOI: 10.1111/bph.16396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND AND PURPOSE Our previous study reported that erythroferrone (ERFE), a newly identified hormone produced by erythroblasts, responded to recombinant human erythropoietin (rHuEPO) sensitively but its dynamics was complicated by double peaks and circadian rhythm. This study intends to elucidate the underlying mechanisms for the double peaks of ERFE dynamics and further determine whether early ERFE measurements can predict haemoglobin responses to rHuEPO. EXPERIMENTAL APPROACH By using the purified recombinant rat ERFE protein and investigating its deposition in rats, the production of ERFE was deconvoluted. To explore the role of iron in ERFE production, we monitored short-term changes of iron status after injection of rHuEPO or deferiprone. Pharmacokinetic/pharmacodynamic (PK/PD) modelling was used to confirm the mechanisms and examine the predictive ability of ERFE for long-term haemoglobin responses. KEY RESULTS The rRatERFE protein was successfully purified. The production of ERFE was deconvoluted and showed two independent peaks (2 and 8 h). Transient iron decrease was observed at 4 h after rHuEPO injection and deferiprone induced significant increases of ERFE. Based on this mechanism, the PK/PD model could characterize the complex dynamics of ERFE. In addition, the model predictions further revealed a stronger correlation between ERFE and haemoglobin peak values than that for observed values. CONCLUSIONS AND IMPLICATIONS The complex dynamics of ERFE should be composited by an immediate release and transient iron deficiency-mediated secondary production of ERFE. The early peak values of ERFE, which occur within a few hours, can predict haemoglobin responses several weeks after ESA treatment.
Collapse
Affiliation(s)
- Peng Xu
- School of Pharmacy, The Chinese University of Hong Kong, HKSAR, China
- Phase I Clinical Trial Center, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Raymond S M Wong
- Division of Hematology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoyu Yan
- School of Pharmacy, The Chinese University of Hong Kong, HKSAR, China
| |
Collapse
|
13
|
Stankiewicz B, Mieszkowski J, Kochanowicz A, Brzezińska P, Niespodziński B, Kowalik T, Waldziński T, Kowalski K, Borkowska A, Reczkowicz J, Daniłowicz-Szymanowicz L, Antosiewicz J. Effect of Single High-Dose Vitamin D 3 Supplementation on Post-Ultra Mountain Running Heart Damage and Iron Metabolism Changes: A Double-Blind Randomized Controlled Trial. Nutrients 2024; 16:2479. [PMID: 39125358 PMCID: PMC11313756 DOI: 10.3390/nu16152479] [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: 07/10/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Exercise-induced inflammation can influence iron metabolism. Conversely, the effects of vitamin D3, which possesses anti-inflammatory properties, on ultramarathon-induced heart damage and changes in iron metabolism have not been investigated. Thirty-five healthy long-distance semi-amateur runners were divided into two groups: one group received 150,000 IU of vitamin D3 24 h prior to a race (n = 16), while the other group received a placebo (n = 19). Serum iron, hepcidin (HPC), ferritin (FER), erythroferrone (ERFE), erythropoietin (EPO), neopterin (NPT), and cardiac troponin T (cTnT) levels were assessed. A considerable effect of ultramarathon running on all examined biochemical markers was observed, with a significant rise in serum levels of ERFE, EPO, HPC, NPT, and cTnT detected immediately post-race, irrespective of the group factor. Vitamin D3 supplementation showed a notable interaction with the UM, specifically in EPO and cTnT, with no other additional changes in the other analysed markers. In addition to the correlation between baseline FER and post-run ERFE, HPC was modified by vitamin D. The ultramarathon significantly influenced the EPO/ERFE/HPC axis; however, a single substantial dose of vitamin D3 had an effect only on EPO, which was associated with the lower heart damage marker cTnT after the run.
Collapse
Affiliation(s)
- Błażej Stankiewicz
- Department of Theory and Methodology of Physical Education and Sport, Faculty of Health Sciences and Physical Education, Kazimierz Wielki University, 85-064 Bydgoszcz, Poland; (B.S.); (T.K.)
| | - Jan Mieszkowski
- Department of Gymnastics and Dance, Gdańsk University of Physical Education and Sport, 80-336 Gdańsk, Poland; (A.K.); (P.B.)
- Faculty of Physical Education and Sport, Charles University, 16-252 Prague, Czech Republic
| | - Andrzej Kochanowicz
- Department of Gymnastics and Dance, Gdańsk University of Physical Education and Sport, 80-336 Gdańsk, Poland; (A.K.); (P.B.)
| | - Paulina Brzezińska
- Department of Gymnastics and Dance, Gdańsk University of Physical Education and Sport, 80-336 Gdańsk, Poland; (A.K.); (P.B.)
| | - Bartłomiej Niespodziński
- Department of Biological Foundations of Physical Education, Faculty of Health Sciences and Physical Education, Kazimierz Wielki University, 85-064 Bydgoszcz, Poland;
| | - Tomasz Kowalik
- Department of Theory and Methodology of Physical Education and Sport, Faculty of Health Sciences and Physical Education, Kazimierz Wielki University, 85-064 Bydgoszcz, Poland; (B.S.); (T.K.)
| | - Tomasz Waldziński
- Faculty of Health Sciences, University of Lomza, 18-400 Łomża, Poland;
| | - Konrad Kowalski
- Department of Bioenergetics and Physiology of Exercise, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (K.K.); (A.B.); (J.R.)
| | - Andżelika Borkowska
- Department of Bioenergetics and Physiology of Exercise, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (K.K.); (A.B.); (J.R.)
| | - Joanna Reczkowicz
- Department of Bioenergetics and Physiology of Exercise, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (K.K.); (A.B.); (J.R.)
| | | | - Jędrzej Antosiewicz
- Department of Bioenergetics and Physiology of Exercise, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (K.K.); (A.B.); (J.R.)
| |
Collapse
|
14
|
Glenthøj A, van Beers EJ, van Wijk R, Rab MAE, Groot E, Vejlstrup N, Toft N, Bendtsen SK, Petersen J, Helby J, Chermat F, Fenaux P, Kuo KHM. Designing a single-arm phase 2 clinical trial of mitapivat for adult patients with erythrocyte membranopathies (SATISFY): a framework for interventional trials in rare anaemias - pilot study protocol. BMJ Open 2024; 14:e083691. [PMID: 39079928 PMCID: PMC11293418 DOI: 10.1136/bmjopen-2023-083691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 07/12/2024] [Indexed: 08/03/2024] Open
Abstract
INTRODUCTION Membranopathies encompass haemolytic disorders arising from genetic variants in erythrocyte membrane proteins, including hereditary spherocytosis and stomatocytosis. Congenital dyserythropoietic anaemia type II (CDA II) is associated with the SEC23B gene and can exhibit phenotypic similarities to membranopathies. Current treatment options for these conditions, apart from splenectomy, are primarily supportive. Mitapivat, a novel pyruvate kinase (PK) activator, has demonstrated efficacy in increasing haemoglobin levels and reducing haemolysis in patients with PK deficiency, thalassemia, sickle cell disease and a mouse model of hereditary spherocytosis. METHODS AND ANALYSES Safety and efficacy of mitapivat sulfate in adult patients with erythrocyte membranopathies (SATISFY) is a prospective, multicentre, single-arm phase two trial involving approximately 25 adult patients (≥18 years) diagnosed with a membranopathy or CDA II. During the 8-week dose escalation period, subjects will receive an initial dose of 50 mg mitapivat two times per day and may increase to 100 mg two times per day at week 4 based on the safety and changes in haemoglobin levels. Patients tolerating mitapivat well may be eligible to continue in two consecutive 24-week fixed dose periods.The primary objective of this study is to evaluate the safety of mitapivat, assessed through the occurrence of treatment-emergent adverse events. Secondary objectives include assessing the effects of mitapivat on haemoglobin levels, haemolysis, erythropoiesis, patient-reported outcome measures and spleen size.SATISFY aims to assess the safety and efficacy of mitapivat in adult patients with red blood cell membranopathies and CDA II, with the aim of establishing proof-of-concept in patients living with these rare conditions. ETHICS AND DISSEMINATION NCT05935202/CTIS:2023-503271-24-01. Findings will be published in peer-reviewed journals. TRIAL REGISTRATION NUMBER Clinicaltrials.gov, NCT05935202. CTIS:2023-503271-24-01. Registered 07-July-2023. Protocol number: 2.1. https://clinicaltrials.gov/study/NCT05935202.
Collapse
Affiliation(s)
- Andreas Glenthøj
- Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Eduard J van Beers
- Benign Hematology Center, Van Creveldkliniek, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Richard van Wijk
- Central Diagnostic Laboratory - Research, Division of Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Urecht, Netherlands
| | - Minke A E Rab
- Central Diagnostic Laboratory - Research, Division of Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Urecht, Netherlands
| | - Evelyn Groot
- Benign Hematology Center, Van Creveldkliniek, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Niels Vejlstrup
- Department of Cardiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Nina Toft
- Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Selma Kofoed Bendtsen
- Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Jesper Petersen
- Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Jens Helby
- Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Fatiha Chermat
- EuroBloodNet Association, Université Paris Cité Faculté de Santé, Paris, France
| | - Pierre Fenaux
- EuroBloodNet Association, Université Paris Cité Faculté de Santé, Paris, France
| | - Kevin H M Kuo
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Division of Medical Oncology and Hematology, Department of Medicine, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Zeng H, Zeng D, Yin X, Zhang W, Wu M, Chen Z. Research progress on high-concentration oxygen therapy after cerebral hemorrhage. Front Neurol 2024; 15:1410525. [PMID: 39139771 PMCID: PMC11320605 DOI: 10.3389/fneur.2024.1410525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/18/2024] [Indexed: 08/15/2024] Open
Abstract
Recently, the role of high-concentration oxygen therapy in cerebral hemorrhage has been extensively discussed. This review describes the research progress in high-concentration oxygen therapy after cerebral hemorrhage. High-concentration oxygen therapy can be classified into two treatment methods: hyperbaric and normobaric high-concentration oxygen therapy. Several studies have reported that high-concentration oxygen therapy uses the pathological mechanisms of secondary ischemia and hypoxia after cerebral hemorrhage as an entry point to improve cerebral oxygenation, metabolic rate, cerebral edema, intracranial pressure, and oxidative stress. We also elucidate the mechanisms by which molecules such as Hypoxia-inducible factor 1-alpha (HIF-1α), vascular endothelial growth factor, and erythropoietin (EPO) may play a role in oxygen therapy. Although people are concerned about the toxicity of hyperoxia, combined with relevant literature, the evidence discussed in this article suggests that as long as the duration, concentration, pressure, and treatment interval of patients with cerebral hemorrhage are properly understood and oxygen is administered within the treatment window, it can be effective to avoid hyperoxic oxygen toxicity. Combined with the latest research, we believe that high-concentration oxygen therapy plays an important positive role in injuries and outcomes after cerebral hemorrhage, and we recommend expanding the use of normal-pressure high-concentration oxygen therapy for cerebral hemorrhage.
Collapse
Affiliation(s)
- He Zeng
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Dakai Zeng
- Department of Anorectal Surgery, Third Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xiaoping Yin
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Wumiao Zhang
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Moxin Wu
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Zhiying Chen
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| |
Collapse
|
16
|
Grander M, Haschka D, Indelicato E, Kremser C, Amprosi M, Nachbauer W, Henninger B, Stefani A, Högl B, Fischer C, Seifert M, Kiechl S, Weiss G, Boesch S. Genetic Determined Iron Starvation Signature in Friedreich's Ataxia. Mov Disord 2024; 39:1088-1098. [PMID: 38686449 DOI: 10.1002/mds.29819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Early studies in cellular models suggested an iron accumulation in Friedreich's ataxia (FA), yet findings from patients are lacking. OBJECTIVES The objective is to characterize systemic iron metabolism, body iron storages, and intracellular iron regulation in FA patients. METHODS In FA patients and matched healthy controls, we assessed serum iron parameters, regulatory hormones as well as the expression of regulatory proteins and iron distribution in peripheral blood mononuclear cells (PBMCs). We applied magnetic resonance imaging with R2*-relaxometry to quantify iron storages in the liver, spleen, and pancreas. Across all evaluations, we assessed the influence of the genetic severity as expressed by the length of the shorter GAA-expansion (GAA1). RESULTS We recruited 40 FA patients (19 women). Compared to controls, FA patients displayed lower serum iron and transferrin saturation. Serum ferritin, hepcidin, mean corpuscular hemoglobin and mean corpuscular volume in FA inversely correlated with the GAA1-repeat length, indicating iron deficiency and restricted availability for erythropoiesis with increasing genetic severity. R2*-relaxometry revealed a reduction of splenic and hepatic iron stores in FA. Liver and spleen R2* values inversely correlated with the GAA1-repeat length. FA PBMCs displayed downregulation of ferritin and upregulation of transferrin receptor and divalent metal transporter-1 mRNA, particularly in patients with >500 GAA1-repeats. In FA PBMCs, intracellular iron was not increased, but shifted toward mitochondria. CONCLUSIONS We provide evidence for a previously unrecognized iron starvation signature at systemic and cellular levels in FA patients, which is related to the underlying genetic severity. These findings challenge the use of systemic iron lowering therapies in FA. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Manuel Grander
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - David Haschka
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Elisabetta Indelicato
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Kremser
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Matthias Amprosi
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Wolfgang Nachbauer
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Benjamin Henninger
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ambra Stefani
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Birgit Högl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christine Fischer
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Kiechl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- VASCage, Centre on Clinical Stroke Research, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Sylvia Boesch
- Center for Rare Movement Disorders Innsbruck, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
17
|
Personnaz J, Guillou H, Kautz L. Fibrinogen-like 1: A hepatokine linking liver physiology to hematology. Hemasphere 2024; 8:e115. [PMID: 38966209 PMCID: PMC11223652 DOI: 10.1002/hem3.115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 07/06/2024] Open
Abstract
A recent study identified the critical contribution of the hepatokine FGL1 to the regulation of iron metabolism during the recovery from anemia. FGL1 is secreted by hepatocytes in response to hypoxia to sequester BMP ligands and repress the transcription of the iron-regulatory hormone hepcidin. This process ensures the proper supply of iron to the bone marrow for new red blood cell synthesis and the restoration of physiological oxygen levels. FGL1 may therefore contribute to the recovery from common anemias and cause iron overload in chronic anemias with ineffective erythropoiesis, such as ß-thalassemia, dyserythropoietic anemia, and myelodysplastic syndromes. However, FGL1 has also been described as a regulator of hepatocyte proliferation, glucose homeostasis, and insulin signaling, as well as a mediator of liver steatosis and immune evasion. Chronic exposure to elevated levels of FGL1 during anemia may therefore have systemic metabolic effects besides iron regulation and erythropoiesis. Here, we are providing an overview of the proposed functions of FGL1 in physiology and pathophysiology.
Collapse
Affiliation(s)
- Jean Personnaz
- IRSD, Université de ToulouseINSERM, INRAE, ENVT, Univ Toulouse III‐Paul Sabatier (UPS)ToulouseFrance
| | - Hervé Guillou
- Toxalim (Research Center in Food Toxicology)INRAE, ENVT, INP‐PURPAN, UMR 1331, UPS, Université de ToulouseToulouseFrance
| | - Léon Kautz
- IRSD, Université de ToulouseINSERM, INRAE, ENVT, Univ Toulouse III‐Paul Sabatier (UPS)ToulouseFrance
| |
Collapse
|
18
|
Guerra A, Hamilton N, Rivera A, Demsko P, Guo S, Rivella S. Combination of a TGF-β ligand trap (RAP-GRL) and TMPRSS6-ASO is superior for correcting β-thalassemia. Am J Hematol 2024; 99:1300-1312. [PMID: 38659383 PMCID: PMC11166515 DOI: 10.1002/ajh.27332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/11/2024] [Accepted: 03/22/2024] [Indexed: 04/26/2024]
Abstract
A recently approved drug that induces erythroid cell maturation (luspatercept) has been shown to improve anemia and reduce the need for blood transfusion in non-transfusion-dependent as well as transfusion-dependent β-thalassemia (BT) patients. Although these results were predominantly positive, not all the patients showed the expected increase in hemoglobin (Hb) levels or transfusion burden reduction. Additional studies indicated that administration of luspatercept in transfusion-dependent BT was associated with increased erythropoietic markers, decreased hepcidin levels, and increased liver iron content. Altogether, these studies suggest that luspatercept may necessitate additional drugs for improved erythroid and iron management. As luspatercept does not appear to directly affect iron metabolism, we hypothesized that TMPRSS6-ASO could improve iron parameters and iron overload when co-administered with luspatercept. We used an agent analogous to murine luspatercept (RAP-GRL) and another novel therapeutic, IONIS TMPRSS6-LRx (TMPRSS6-ASO), a hepcidin inducer, to treat non-transfusion-dependent BT-intermedia mice. Our study shows that RAP-GRL alone improved red blood cell (RBC) production, with no or limited effect on splenomegaly and iron parameters. In contrast, TMPRSS6-ASO improved RBC measurements, ameliorated splenomegaly, and improved iron overload most effectively. Our results provide pre-clinical support for combining TMPRSS6-ASO and luspatercept in treating BT, as these drugs together show potential for simultaneously improving both erythroid and iron parameters in BT patients.
Collapse
Affiliation(s)
- Amaliris Guerra
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Nolan Hamilton
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Ariel Rivera
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Perry Demsko
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Cell and Molecular Biology affinity group (CAMB), University of Pennsylvania, Philadelphia, PA, USA
| | - Shuling Guo
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Cell and Molecular Biology affinity group (CAMB), University of Pennsylvania, Philadelphia, PA, USA
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics-CHOP
- Penn Center for Musculoskeletal Disorders, CHOP, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
19
|
Chu J, Wang K, Lu L, Zhao H, Hu J, Xiao W, Wu Q. Advances of Iron and Ferroptosis in Diabetic Kidney Disease. Kidney Int Rep 2024; 9:1972-1985. [PMID: 39081773 PMCID: PMC11284386 DOI: 10.1016/j.ekir.2024.04.012] [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: 01/21/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 08/02/2024] Open
Abstract
Diabetes mellitus presents a significant threat to human health because it disrupts energy metabolism and gives rise to various complications, including diabetic kidney disease (DKD). Metabolic adaptations occurring in the kidney in response to diabetes contribute to the pathogenesis of DKD. Iron metabolism and ferroptosis, a recently defined form of cell death resulting from iron-dependent excessive accumulation of lipid peroxides, have emerged as crucial players in the progression of DKD. In this comprehensive review, we highlight the profound impact of adaptive and maladaptive responses regulating iron metabolism on the progression of kidney damage in diabetes. We summarize the current understanding of iron homeostasis and ferroptosis in DKD. Finally, we propose that precise manipulation of iron metabolism and ferroptosis may serve as potential strategies for kidney management in diabetes.
Collapse
Affiliation(s)
- Jiayi Chu
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Kewu Wang
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Lulu Lu
- Department of Nutrition and Toxicology, Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines of Zhejiang Province, School of Public Health, Hangzhou Normal University, Hangzhou, China
| | - Hui Zhao
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Jibo Hu
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| | - Wenbo Xiao
- Department of Radiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China
| | - Qian Wu
- Department of Radiology, Center of Regenerative and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang, China
| |
Collapse
|
20
|
Tanzi E, Di Modica SM, Bordini J, Olivari V, Pagani A, Furiosi V, Silvestri L, Campanella A, Nai A. Bone marrow Tfr2 deletion improves the therapeutic efficacy of the activin-receptor ligand trap RAP-536 in β-thalassemic mice. Am J Hematol 2024; 99:1313-1325. [PMID: 38629683 DOI: 10.1002/ajh.27336] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/02/2024] [Accepted: 04/06/2024] [Indexed: 06/12/2024]
Abstract
β-thalassemia is a disorder characterized by anemia, ineffective erythropoiesis (IE), and iron overload, whose treatment still requires improvement. The activin receptor-ligand trap Luspatercept, a novel therapeutic option for β-thalassemia, stimulates erythroid differentiation inhibiting the transforming growth factor β pathway. However, its exact mechanism of action and the possible connection with erythropoietin (Epo), the erythropoiesis governing cytokine, remain to be clarified. Moreover, Luspatercept does not correct all the features of the disease, calling for the identification of strategies that enhance its efficacy. Transferrin receptor 2 (TFR2) regulates systemic iron homeostasis in the liver and modulates the response to Epo of erythroid cells, thus balancing red blood cells production with iron availability. Stimulating Epo signaling, hematopoietic Tfr2 deletion ameliorates anemia and IE in Hbbth3/+ thalassemic mice. To investigate whether hematopoietic Tfr2 inactivation improves the efficacy of Luspatercept, we treated Hbbth3/+ mice with or without hematopoietic Tfr2 (Tfr2BMKO/Hbbth3/+) with RAP-536, the murine analog of Luspatercept. As expected, both hematopoietic Tfr2 deletion and RAP-536 significantly ameliorate IE and anemia, and the combined approach has an additive effect. Since RAP-536 has comparable efficacy in both Hbbth3/+ and Tfr2BMKO/Hbbth3/+ animals, we propose that the drug promotes erythroid differentiation independently of TFR2 and EPO stimulation. Notably, the lack of Tfr2, but not RAP-536, can also attenuate iron-overload and related complications. Overall, our results shed further light on the mechanism of action of Luspatercept and suggest that strategies aimed at inhibiting hematopoietic TFR2 might improve the therapeutic efficacy of activin receptor-ligand traps.
Collapse
Affiliation(s)
- Emanuele Tanzi
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Simona Maria Di Modica
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Jessica Bordini
- Vita-Salute San Raffaele University, Milan, Italy
- B-cell Neoplasia Unit, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Violante Olivari
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Alessia Pagani
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Valeria Furiosi
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Laura Silvestri
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Alessandro Campanella
- Vita-Salute San Raffaele University, Milan, Italy
- B-cell Neoplasia Unit, Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Antonella Nai
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
21
|
Lakhal-Littleton S, Cleland JGF. Iron deficiency and supplementation in heart failure. Nat Rev Cardiol 2024; 21:463-486. [PMID: 38326440 DOI: 10.1038/s41569-024-00988-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/09/2024]
Abstract
Non-anaemic iron deficiency (NAID) is a strategic target in cardiovascular medicine because of its association with a range of adverse effects in various conditions. Endeavours to tackle NAID in heart failure have yielded mixed results, exposing knowledge gaps in how best to define 'iron deficiency' and the handling of iron therapies by the body. To address these gaps, we harness the latest understanding of the mechanisms of iron homeostasis outside the erythron and integrate clinical and preclinical lines of evidence. The emerging picture is that current definitions of iron deficiency do not assimilate the multiple influences at play in patients with heart failure and, consequently, fail to identify those with a truly unmet need for iron. Additionally, current iron supplementation therapies benefit only certain patients with heart failure, reflecting differences in the nature of the unmet need for iron and the modifying effects of anaemia and inflammation on the handling of iron therapies by the body. Building on these insights, we identify untapped opportunities in the management of NAID, including the refinement of current approaches and the development of novel strategies. Lessons learned from NAID in cardiovascular disease could ultimately translate into benefits for patients with other chronic conditions such as chronic kidney disease, chronic obstructive pulmonary disease and cancer.
Collapse
Affiliation(s)
| | - John G F Cleland
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| |
Collapse
|
22
|
Nakai T, Saigusa D, Kato K, Fukuuchi T, Koshiba S, Yamamoto M, Suzuki N. The drug-specific properties of hypoxia-inducible factor-prolyl hydroxylase inhibitors in mice reveal a significant contribution of the kidney compared to the liver to erythropoietin induction. Life Sci 2024; 346:122641. [PMID: 38614299 DOI: 10.1016/j.lfs.2024.122641] [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: 02/08/2024] [Revised: 03/13/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
AIMS Kidney disease often leads to anemia due to a defect in the renal production of the erythroid growth factor erythropoietin (EPO), which is produced under the positive regulation of hypoxia-inducible transcription factors (HIFs). Chemical compounds that inhibit HIF-prolyl hydroxylases (HIF-PHs), which suppress HIFs, have been developed to reactivate renal EPO production in renal anemia patients. Currently, multiple HIF-PH inhibitors, in addition to conventional recombinant EPO reagents, are used for renal anemia treatment. This study aimed to elucidate the therapeutic mechanisms and drug-specific properties of HIF-PH inhibitors. METHODS AND KEY FINDINGS Gene expression analyses and mass spectrometry revealed that HIF-PH inhibitors (daprodustat, enarodustat, molidustat, and vadadustat) alter Epo gene expression levels in the kidney and liver in a drug-specific manner, with different pharmacokinetics in the plasma and urine after oral administration to mice. The drug specificity revealed the dominant contribution of EPO induction in the kidneys rather than in the liver to plasma EPO levels after HIF-PH inhibitor administration. We also found that several HIF-PH inhibitors directly induce duodenal gene expression related to iron intake, while these drugs indirectly suppress hepatic hepcidin expression to mobilize stored iron for hemoglobin synthesis through induction of the EPO-erythroferrone axis. SIGNIFICANCE Renal EPO induction is the major target of HIF-PH inhibitors for their therapeutic effects on erythropoiesis. Additionally, the drug-specific properties of HIF-PH inhibitors in EPO induction and iron metabolism have been shown in mice, providing useful information for selecting the proper HIF-PH inhibitor for each renal anemia patient.
Collapse
Affiliation(s)
- Taku Nakai
- Applied Oxygen Physiology Project, New Industry Creation Hatchery Center, Tohoku University, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan; Division of Oxygen Biology, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Daisuke Saigusa
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan
| | - Koichiro Kato
- Applied Oxygen Physiology Project, New Industry Creation Hatchery Center, Tohoku University, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan; Division of Oxygen Biology, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Tomoko Fukuuchi
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan
| | - Seizo Koshiba
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan; The Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, 2-1, Seiryo-machi, Aoba-ku, Sendai 980-8573, Japan
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Norio Suzuki
- Applied Oxygen Physiology Project, New Industry Creation Hatchery Center, Tohoku University, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan; Division of Oxygen Biology, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan.
| |
Collapse
|
23
|
van Beers EJ, Al-Samkari H, Grace RF, Barcellini W, Glenthøj A, DiBacco M, Wind-Rotolo M, Xu R, Beynon V, Patel P, Porter JB, Kuo KHM. Mitapivat improves ineffective erythropoiesis and iron overload in adult patients with pyruvate kinase deficiency. Blood Adv 2024; 8:2433-2441. [PMID: 38330179 PMCID: PMC11112604 DOI: 10.1182/bloodadvances.2023011743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/20/2023] [Accepted: 01/06/2024] [Indexed: 02/10/2024] Open
Abstract
ABSTRACT Pyruvate kinase (PK) deficiency is a rare, hereditary disease characterized by chronic hemolytic anemia. Iron overload is a common complication regardless of age, genotype, or transfusion history. Mitapivat, an oral, allosteric PK activator, improves anemia and hemolysis in adult patients with PK deficiency. Mitapivat's impact on iron overload and ineffective erythropoiesis was evaluated in adults with PK deficiency who were not regularly transfused in the phase 3 ACTIVATE trial and long-term extension (LTE) (#NCT03548220/#NCT03853798). Patients in the LTE received mitapivat throughout ACTIVATE/LTE (baseline to week 96; mitapivat-to-mitapivat [M/M] arm) or switched from placebo (baseline to week 24) to mitapivat (week 24 to week 96; placebo-to-mitapivat [P/M] arm). Changes from baseline in markers of iron overload and erythropoiesis were assessed to week 96. Improvements in hepcidin (mean, 4770.0 ng/L; 95% confidence interval [CI], -1532.3 to 11 072.3), erythroferrone (mean, -9834.9 ng/L; 95% CI, -14 328.4 to -5341.3), soluble transferrin receptor (mean, -56.0 nmol/L; 95% CI, -84.8 to -27.2), and erythropoietin (mean, -32.85 IU/L; 95% CI, -54.65 to -11.06) were observed in the M/M arm (n = 40) from baseline to week 24, sustained to week 96. No improvements were observed in the P/M arm (n = 40) to week 24; however, upon transitioning to mitapivat, improvements similar to those observed in the M/M arm were seen. Mean changes from baseline in liver iron concentration by magnetic resonance imaging at week 96 in the M/M arm and the P/M arm were -2.0 mg Fe/g dry weight (dw; 95% CI, -4.8 to -0.8) and -1.8 mg Fe/g dw (95% CI, -4.4 to 0.80), respectively. Mitapivat is the first disease-modifying pharmacotherapy shown to have beneficial effects on iron overload and ineffective erythropoiesis in patients with PK deficiency. This trial was registered at www.ClinicalTrials.gov as #NCT03548220 (ACTIVATE) and #NCT03853798 (LTE).
Collapse
Affiliation(s)
- Eduard J. van Beers
- Center for Benign Haematology, Thrombosis and Haemostasis, Van Creveldkliniek, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Hanny Al-Samkari
- Division of Hematology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Rachael F. Grace
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Wilma Barcellini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andreas Glenthøj
- Danish Red Blood Cell Center, Department of Haematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | | | - Rengyi Xu
- Agios Pharmaceuticals, Inc, Cambridge, MA
| | | | | | - John B. Porter
- Haematology Department, University College London Hospitals, London, United Kingdom
| | - Kevin H. M. Kuo
- Division of Hematology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
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
|
25
|
Ma JK, Su LD, Feng LL, Li JL, Pan L, Danzeng Q, Li Y, Shang T, Zhan XL, Chen SY, Ying S, Hu JR, Chen XQ, Zhang Q, Liang T, Lu XJ. TFPI from erythroblasts drives heme production in central macrophages promoting erythropoiesis in polycythemia. Nat Commun 2024; 15:3976. [PMID: 38729948 PMCID: PMC11087540 DOI: 10.1038/s41467-024-48328-8] [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: 07/25/2023] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
Bleeding and thrombosis are known as common complications of polycythemia for a long time. However, the role of coagulation system in erythropoiesis is unclear. Here, we discover that an anticoagulant protein tissue factor pathway inhibitor (TFPI) plays an essential role in erythropoiesis via the control of heme biosynthesis in central macrophages. TFPI levels are elevated in erythroblasts of human erythroblastic islands with JAK2V617F mutation and hypoxia condition. Erythroid lineage-specific knockout TFPI results in impaired erythropoiesis through decreasing ferrochelatase expression and heme biosynthesis in central macrophages. Mechanistically, the TFPI interacts with thrombomodulin to promote the downstream ERK1/2-GATA1 signaling pathway to induce heme biosynthesis in central macrophages. Furthermore, TFPI blockade impairs human erythropoiesis in vitro, and normalizes the erythroid compartment in mice with polycythemia. These results show that erythroblast-derived TFPI plays an important role in the regulation of erythropoiesis and reveal an interplay between erythroblasts and central macrophages.
Collapse
Affiliation(s)
- Jun-Kai Ma
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Lin-Lin Feng
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China
| | - Jing-Lin Li
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Li Pan
- The General Hospital of Tibet Military Area Command, Lhasa, China
| | - Qupei Danzeng
- Department of Tibetan Medicine; University of Tibetan Medicine, Lhasa, 540100, China
| | - Yanwei Li
- Core Facilities, Zhejiang University School of Medicine, Hangzhou, China
| | - Tongyao Shang
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xiao-Lin Zhan
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China
| | - Si-Ying Chen
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China
| | - Shibo Ying
- School of Public Health, Hangzhou Medical College, Hangzhou, 310013, China
| | - Jian-Rao Hu
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Xue Qun Chen
- Zhejiang University, School of Brain Science and Brain Medicine, Hangzhou, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Xin-Jiang Lu
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
26
|
Suzuki N, Iwamura Y, Kato K, Ishioka H, Konta Y, Sato K, Uchida N, Koida N, Sekine H, Tanaka T, Kumagai N, Nakai T. Crosstalk between oxygen signaling and iron metabolism in renal interstitial fibroblasts. J Clin Biochem Nutr 2024; 74:179-184. [PMID: 38799135 PMCID: PMC11111471 DOI: 10.3164/jcbn.24-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/23/2024] [Indexed: 05/29/2024] Open
Abstract
To maintain the oxygen supply, the production of red blood cells (erythrocytes) is promoted under low-oxygen conditions (hypoxia). Oxygen is carried by hemoglobin in erythrocytes, in which the majority of the essential element iron in the body is contained. Because iron metabolism is strictly controlled in a semi-closed recycling system to protect cells from oxidative stress caused by iron, hypoxia-inducible erythropoiesis is closely coordinated by regulatory systems that mobilize stored iron for hemoglobin synthesis. The erythroid growth factor erythropoietin (EPO) is mainly secreted by interstitial fibroblasts in the renal cortex, which are known as renal EPO-producing (REP) cells, and promotes erythropoiesis and iron mobilization. Intriguingly, EPO production is strongly induced by hypoxia through iron-dependent pathways in REP cells. Here, we summarize recent studies on the network mechanisms linking hypoxia-inducible EPO production, erythropoiesis and iron metabolism. Additionally, we introduce disease mechanisms related to disorders in the network mediated by REP cell functions. Furthermore, we propose future studies regarding the application of renal cells derived from the urine of kidney disease patients to investigate the molecular pathology of chronic kidney disease and develop precise and personalized medicine for kidney disease.
Collapse
Affiliation(s)
- Norio Suzuki
- Applied Oxygen Physiology Project, New Industry Creation Hatchery Center, Tohoku University, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Division of Oxygen Biology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Yuma Iwamura
- Division of Oxygen Biology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Koichiro Kato
- Applied Oxygen Physiology Project, New Industry Creation Hatchery Center, Tohoku University, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Division of Oxygen Biology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Hirotaka Ishioka
- Division of Oxygen Biology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Department of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Yusuke Konta
- Division of Oxygen Biology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Department of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Koji Sato
- Division of Oxygen Biology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Department of Nephrology, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Nao Uchida
- Department of Pediatrics, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Noa Koida
- Division of Oxygen Biology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Hiroki Sekine
- Division of Oxygen Biology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Tetsuhiro Tanaka
- Department of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Naonori Kumagai
- Department of Pediatrics, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Taku Nakai
- Division of Oxygen Biology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| |
Collapse
|
27
|
de Jong MF, Nemeth E, Rawee P, Bramham K, Eisenga MF. Anemia in Pregnancy With CKD. Kidney Int Rep 2024; 9:1183-1197. [PMID: 38707831 PMCID: PMC11069017 DOI: 10.1016/j.ekir.2024.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 12/05/2023] [Accepted: 01/08/2024] [Indexed: 05/07/2024] Open
Abstract
Chronic kidney disease (CKD), anemia, and iron deficiency are global health issues affecting individuals in both high-income and low-income countries. In pregnancy, both CKD and iron deficiency anemia increase the risk of adverse maternal and neonatal outcomes, including increased maternal morbidity and mortality, stillbirth, perinatal death, preterm birth, and low birthweight. However, it is unknown to which extent iron deficiency anemia contributes to adverse outcomes in CKD pregnancy. Furthermore, little is known regarding the prevalence, pathophysiology, and treatment of iron deficiency and anemia in pregnant women with CKD. Therefore, there are many unanswered questions regarding optimal management with oral or i.v. iron and recombinant human erythropoietin (rhEPO) in these women. In this review, we present a short overview of the (patho)physiology of anemia in healthy pregnancy and in people living with CKD. We present an evaluation of the literature on iron deficiency, anemia, and nutritional deficits in pregnant women with CKD; and we evaluate current knowledge gaps. Finally, we propose research priorities regarding anemia in pregnant women with CKD.
Collapse
Affiliation(s)
- Margriet F.C. de Jong
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, The Netherlands
| | - Elizabeta Nemeth
- Department of Medicine, University of California, Los Angeles, California, USA
| | - Pien Rawee
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, The Netherlands
| | - Kate Bramham
- Department of Women and Children’s Health, King’s College London, London, UK
| | - Michele F. Eisenga
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, The Netherlands
| |
Collapse
|
28
|
Martínez-Heredia L, Canelo-Moreno JM, García-Fontana B, Muñoz-Torres M. Non-Classical Effects of FGF23: Molecular and Clinical Features. Int J Mol Sci 2024; 25:4875. [PMID: 38732094 PMCID: PMC11084844 DOI: 10.3390/ijms25094875] [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: 04/08/2024] [Revised: 04/21/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
This article reviews the role of fibroblast growth factor 23 (FGF23) protein in phosphate metabolism, highlighting its regulation of vitamin D, parathyroid hormone, and bone metabolism. Although it was traditionally thought that phosphate-calcium homeostasis was controlled exclusively by parathyroid hormone (PTH) and calcitriol, pathophysiological studies revealed the influence of FGF23. This protein, expressed mainly in bone, inhibits the renal reabsorption of phosphate and calcitriol formation, mediated by the α-klotho co-receptor. In addition to its role in phosphate metabolism, FGF23 exhibits pleiotropic effects in non-renal systems such as the cardiovascular, immune, and metabolic systems, including the regulation of gene expression and cardiac fibrosis. Although it has been proposed as a biomarker and therapeutic target, the inhibition of FGF23 poses challenges due to its potential side effects. However, the approval of drugs such as burosumab represents a milestone in the treatment of FGF23-related diseases.
Collapse
Affiliation(s)
- Luis Martínez-Heredia
- Instituto de Investigación Biosanitaria de Granada, 18014 Granada, Spain;
- Biomedical Research Network in Fragility and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Beatriz García-Fontana
- Instituto de Investigación Biosanitaria de Granada, 18014 Granada, Spain;
- Biomedical Research Network in Fragility and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- Department of Cell Biology, University of Granada, 18016 Granada, Spain
| | - Manuel Muñoz-Torres
- Instituto de Investigación Biosanitaria de Granada, 18014 Granada, Spain;
- Biomedical Research Network in Fragility and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Endocrinology and Nutrition Unit, University Hospital Clínico San Cecilio, 18016 Granada, Spain
- Department of Medicine, University of Granada, 18016 Granada, Spain
| |
Collapse
|
29
|
Tatsumi Y, Yano M, Wakusawa S, Miyajima H, Ishikawa T, Imashuku S, Takano A, Nihei W, Kato A, Kato K, Hayashi H, Yoshioka K, Hayashi K. A Revised Classification of Primary Iron Overload Syndromes. J Clin Transl Hepatol 2024; 12:346-356. [PMID: 38638373 PMCID: PMC11022062 DOI: 10.14218/jcth.2023.00290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 11/02/2023] [Accepted: 12/11/2023] [Indexed: 04/20/2024] Open
Abstract
Background and Aims The clinical introduction of hepcidin25 (Hep25) has led to a more detailed understanding of its relationship with ferroportin (FP) and divalent metal transporter1 in primary iron overload syndromes (PIOSs). In 2012, we proposed a classification of PIOSs based on the Hep25/FP system, which consists of prehepatic aceruloplasminemia, hepatic hemochromatosis (HC), and posthepatic FP disease (FP-D). However, in consideration of accumulated evidence on PIOSs, we aimed to renew the classification. Methods We reviewed the 2012 classification and retrospectively renewed it according to new information on PIOSs. Results Iron-loading anemia was included in PIOSs as a prehepatic form because of the newly discovered erythroferrone-induced suppression of Hep25, and the state of traditional FP-D was remodeled as the BIOIRON proposal. The key molecules responsible for prehepatic PIOSs are low transferrin saturation in aceruloplasminemia and increased erythroferrone production by erythroblasts in iron-loading anemia. Hepatic PIOSs comprise four genotypes of HC, in each of which the synthesis of Hep25 is inappropriately reduced in the liver. Hepatic Hep25 synthesis is adequate in posthepatic PIOSs; however, two mutant FP molecules may resist Hep25 differently, resulting in SLC40A1-HC and FP-D, respectively. PIOS phenotypes are diagnosed using laboratory tests, including circulating Hep25, followed by suitable treatments. Direct sequencing of the candidate genes may be outsourced to gene centers when needed. Laboratory kits for the prevalent mutations, such as C282Y, may be the first choice for a genetic analysis of HC in Caucasians. Conclusions The revised classification may be useful worldwide.
Collapse
Affiliation(s)
- Yasuaki Tatsumi
- Department of Medical Biochemistry, Faculty of Pharmaceutical Sciences, Toho University, Funabashi, Japan
| | - Motoyoshi Yano
- Department of Gastroenterology, Yokkaichi Municipal Hospital, Yokkaichi, Japan
| | - Shinya Wakusawa
- Department of Medical Technology, Shubun University, Ichinomiya, Japan
| | - Hiroaki Miyajima
- Department of Medicine and Neurology, Tenryu Kohseikai Clinic, Hamamatsu, Japan
| | - Tetsuya Ishikawa
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinsaku Imashuku
- Department of Laboratory Medicine, Uji-Tokushukai Medical Center, Uji, Japan
| | - Atsuko Takano
- Department of Medicine, Saiseikai Takaoka Hospital, Takaoka, Japan
| | - Wataru Nihei
- Department of Medicine, Aichi-Gakuin University School of Pharmacy, Nagoya, Japan
| | - Ayako Kato
- Department of Medicine, Aichi-Gakuin University School of Pharmacy, Nagoya, Japan
| | - Koichi Kato
- Department of Medicine, Aichi-Gakuin University School of Pharmacy, Nagoya, Japan
| | - Hisao Hayashi
- Department of Medicine, Aichi-Gakuin University School of Pharmacy, Nagoya, Japan
| | - Kentaro Yoshioka
- Department of Gastroenterology, FNPS Meijo Hospital, Nagoya, Japan
| | - Kazuhiko Hayashi
- Department of Gastroenterology, FNPS Meijo Hospital, Nagoya, Japan
| |
Collapse
|
30
|
Wake M, Palin A, Belot A, Berger M, Lorgouilloux M, Bichon M, Papworth J, Bayliss L, Grimshaw B, Rynkiewicz N, Paterson J, Poindron A, Spearing E, Carter E, Hudson R, Campbell M, Petzer V, Besson-Fournier C, Latour C, Largounez A, Gourbeyre O, Fay A, Coppin H, Roth MP, Theurl I, Germaschewski V, Meynard D. A human anti-matriptase-2 antibody limits iron overload, α-globin aggregates, and splenomegaly in β-thalassemic mice. Blood Adv 2024; 8:1898-1907. [PMID: 38241484 PMCID: PMC11021894 DOI: 10.1182/bloodadvances.2023012010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/21/2024] Open
Abstract
ABSTRACT Iron plays a major role in the deterioration of β-thalassemia. Indeed, the high levels of transferrin saturation and iron delivered to erythroid progenitors are associated with production of α-globin precipitates that negatively affect erythropoiesis. Matriptase-2/TMPRSS6, a membrane-bound serine protease expressed in hepatocytes, negatively modulates hepcidin production and thus is a key target to prevent iron overload in β-thalassemia. To address safety concerns raised by the suppression of Tmprss6 by antisense oligonucleotides or small interfering RNA, we tested a fully human anti-matriptase-2 antibody, RLYB331, which blocks the protease activity of matriptase-2. When administered weekly to Hbbth3/+ mice, RLYB331 induced hepcidin expression, reduced iron loading, prevented the formation of toxic α-chain/heme aggregates, reduced ros oxygen species formation, and improved reticulocytosis and splenomegaly. To increase the effectiveness of RLYB331 in β-thalassemia treatment even further, we administered RLYB331 in combination with RAP-536L, a ligand-trapping protein that contains the extracellular domain of activin receptor type IIB and alleviates anemia by promoting differentiation of late-stage erythroid precursors. RAP-536L alone did not prevent iron overload but significantly reduced apoptosis in the erythroid populations of the bone marrow, normalized red blood cell counts, and improved hemoglobin and hematocrit levels. Interestingly, the association of RLYB331 with RAP-536L entirely reversed the β-thalassemia phenotype in Hbbth3/+ mice and simultaneously corrected iron overload, ineffective erythropoiesis, splenomegaly, and hematological parameters, suggesting that a multifunctional molecule consisting of the fusion of RLYB331 with luspatercept (human version of RAP-536L) would allow administration of a single medication addressing simultaneously the different pathophysiological aspects of β-thalassemia.
Collapse
Affiliation(s)
- Matthew Wake
- Kymab Ltd, Babraham Research Campus, Cambridge, United Kingdom
| | - Anaïs Palin
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, École Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Audrey Belot
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, École Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Mathieu Berger
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, École Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Megane Lorgouilloux
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, École Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Margot Bichon
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, École Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France
| | | | - Luke Bayliss
- Kymab Ltd, Babraham Research Campus, Cambridge, United Kingdom
| | | | | | - Jemima Paterson
- Kymab Ltd, Babraham Research Campus, Cambridge, United Kingdom
| | - Alicia Poindron
- Kymab Ltd, Babraham Research Campus, Cambridge, United Kingdom
| | - Erin Spearing
- Kymab Ltd, Babraham Research Campus, Cambridge, United Kingdom
| | - Emily Carter
- Kymab Ltd, Babraham Research Campus, Cambridge, United Kingdom
| | - Robyne Hudson
- Kymab Ltd, Babraham Research Campus, Cambridge, United Kingdom
| | - Millie Campbell
- Kymab Ltd, Babraham Research Campus, Cambridge, United Kingdom
| | - Verena Petzer
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Céline Besson-Fournier
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, École Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Chloé Latour
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, École Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Amélie Largounez
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, École Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Ophélie Gourbeyre
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, École Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Alexis Fay
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, École Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Hélène Coppin
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, École Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Marie-Paule Roth
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, École Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Igor Theurl
- Kymab Ltd, Babraham Research Campus, Cambridge, United Kingdom
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Delphine Meynard
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, Institut National de Recherche pour l'Agriculture, l'alimentation et l'Environnement, École Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
31
|
Correnti M, Gammella E, Cairo G, Recalcati S. Iron Absorption: Molecular and Pathophysiological Aspects. Metabolites 2024; 14:228. [PMID: 38668356 PMCID: PMC11052485 DOI: 10.3390/metabo14040228] [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: 03/22/2024] [Revised: 04/12/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Iron is an essential nutrient for growth among all branches of life, but while iron is among the most common elements, bioavailable iron is a relatively scarce nutrient. Since iron is fundamental for several biological processes, iron deficiency can be deleterious. On the other hand, excess iron may lead to cell and tissue damage. Consequently, iron balance is strictly regulated. As iron excretion is not physiologically controlled, systemic iron homeostasis is maintained at the level of absorption, which is mainly influenced by the amount of iron stores and the level of erythropoietic activity, the major iron consumer. Here, we outline recent advances that increased our understanding of the molecular aspects of iron absorption. Moreover, we examine the impact of these recent insights on dietary strategies for maintaining iron balance.
Collapse
Affiliation(s)
| | | | - Gaetano Cairo
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy; (M.C.); (E.G.); (S.R.)
| | | |
Collapse
|
32
|
Mbiandjeu SCT, Siciliano A, Mattè A, Federti E, Perduca M, Melisi D, Andolfo I, Amoresano A, Iolascon A, Valenti MT, Turrini F, Bovi M, Pisani A, Recchiuti A, Mattoscio D, Riccardi V, Dalle Carbonare L, Brugnara C, Mohandas N, De Franceschi L. Nrf2 Plays a Key Role in Erythropoiesis during Aging. Antioxidants (Basel) 2024; 13:454. [PMID: 38671902 PMCID: PMC11047311 DOI: 10.3390/antiox13040454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/06/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Aging is characterized by increased oxidation and reduced efficiency of cytoprotective mechanisms. Nuclear factor erythroid-2-related factor (Nrf2) is a key transcription factor, controlling the expression of multiple antioxidant proteins. Here, we show that Nrf2-/- mice displayed an age-dependent anemia, due to the combined contributions of reduced red cell lifespan and ineffective erythropoiesis, suggesting a role of Nrf2 in erythroid biology during aging. Mechanistically, we found that the expression of antioxidants during aging is mediated by activation of Nrf2 function by peroxiredoxin-2. The absence of Nrf2 resulted in persistent oxidation and overactivation of adaptive systems such as the unfolded protein response (UPR) system and autophagy in Nrf2-/- mouse erythroblasts. As Nrf2 is involved in the expression of autophagy-related proteins such as autophagy-related protein (Atg) 4-5 and p62, we found impairment of late phase of autophagy in Nrf2-/- mouse erythroblasts. The overactivation of the UPR system and impaired autophagy drove apoptosis of Nrf2-/- mouse erythroblasts via caspase-3 activation. As a proof of concept for the role of oxidation, we treated Nrf2-/- mice with astaxanthin, an antioxidant, in the form of poly (lactic-co-glycolic acid) (PLGA)-loaded nanoparticles (ATS-NPs) to improve its bioavailability. ATS-NPs ameliorated the age-dependent anemia and decreased ineffective erythropoiesis in Nrf2-/- mice. In summary, we propose that Nrf2 plays a key role in limiting age-related oxidation, ensuring erythroid maturation and growth during aging.
Collapse
Affiliation(s)
| | - Angela Siciliano
- Dipartimento Ingegneria per la Medicina di Innovazione—DIMI, University of Verona, 37134 Verona, Italy; (A.S.); (E.F.); (V.R.); (L.D.C.)
- Department of Medicine, AOUI Verona, 37134 Verona, Italy
| | - Alessandro Mattè
- Department of Medicine, University of Verona, 37134 Verona, Italy; (S.C.T.M.); (A.M.); (D.M.)
| | - Enrica Federti
- Dipartimento Ingegneria per la Medicina di Innovazione—DIMI, University of Verona, 37134 Verona, Italy; (A.S.); (E.F.); (V.R.); (L.D.C.)
- Department of Medicine, AOUI Verona, 37134 Verona, Italy
| | - Massimiliano Perduca
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (M.P.); (M.B.); (A.P.)
| | - Davide Melisi
- Department of Medicine, University of Verona, 37134 Verona, Italy; (S.C.T.M.); (A.M.); (D.M.)
| | - Immacolata Andolfo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (I.A.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80131 Naples, Italy
| | - Angela Amoresano
- Department of Chimical Sciences, University Federico II, 80138 Naples, Italy;
| | - Achille Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Naples, Italy; (I.A.); (A.I.)
- CEINGE Biotecnologie Avanzate, 80131 Naples, Italy
| | | | | | - Michele Bovi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (M.P.); (M.B.); (A.P.)
| | - Arianna Pisani
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (M.P.); (M.B.); (A.P.)
| | - Antonio Recchiuti
- Department of Medical, Oral, and Biotechnology Science, “G. d’Annunzio” University Chieti–Pescara, 66013 Chieti, Italy; (A.R.); (D.M.)
| | - Domenico Mattoscio
- Department of Medical, Oral, and Biotechnology Science, “G. d’Annunzio” University Chieti–Pescara, 66013 Chieti, Italy; (A.R.); (D.M.)
| | - Veronica Riccardi
- Dipartimento Ingegneria per la Medicina di Innovazione—DIMI, University of Verona, 37134 Verona, Italy; (A.S.); (E.F.); (V.R.); (L.D.C.)
| | - Luca Dalle Carbonare
- Dipartimento Ingegneria per la Medicina di Innovazione—DIMI, University of Verona, 37134 Verona, Italy; (A.S.); (E.F.); (V.R.); (L.D.C.)
- Department of Medicine, AOUI Verona, 37134 Verona, Italy
| | - Carlo Brugnara
- Department of Laboratory Medicine, Boston Children’s Hospital, Boston, MA 02114, USA;
- Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Narla Mohandas
- New York Blood Center Enterprises, New York, NY 10065, USA;
| | - Lucia De Franceschi
- Dipartimento Ingegneria per la Medicina di Innovazione—DIMI, University of Verona, 37134 Verona, Italy; (A.S.); (E.F.); (V.R.); (L.D.C.)
- Department of Medicine, AOUI Verona, 37134 Verona, Italy
| |
Collapse
|
33
|
Dulkadir R, Turna Saltoğlu G, Güneş A. Erythroferrone and hepcidin levels in children with iron deficiency anemia. BMC Pediatr 2024; 24:240. [PMID: 38575873 PMCID: PMC10993451 DOI: 10.1186/s12887-024-04594-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 01/25/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Iron deficiency anemia remains a significant public health issue in developing countries. The regulation of iron metabolism is primarily controlled by hepcidin, a key regulatory protein. During erythropoiesis, erythroferrone (ERFE), a hormone produced by erythroblasts in response to erythropoietin (EPO) synthesis, mediates the suppression of hepcidin. In this study, it was aimed to determine the correlation between erythroferrone (ERFE) and hepcidin levels in children with iron deficiency anemia. METHODS This is a case-control study conducted at Kırşehir Ahi Evran University Training and Research Hospital Pediatrics Clinic between 1 and 31 September 2020. The study included 26 healthy children and 26 children with iron deficiency anemia. In order to evaluate iron status,whole blood count, serum iron, total iron binding capacity (TIBC), and ferritin levels were analyzed. The study measured the levels of hepcidin and erythroferrone in the serum of children diagnosed with iron deficiency before and after one month of iron treatment, as well as in a control group, using the ELISA method. Correlation between whole blood count, initial ferritin, hepcidin, ERFE and ferritin in the iron deficiency group was evaluated. RESULTS Compared with healthy controls, the iron-deficient group had significantly lower haemoglobin (p < 0.001), MCV (p = 0.001), MCH (p < 0.001), MCHC (p < 0.001), iron (p < 0.001), ferritin (p < 0.001) and hepcidin (p = 0.001). Ferritin and hepcidin levels increased while erythroferrone levels remained unchanged after iron deficiency treatment. There was no correlation between hepcidin and ferritin levels in treatment group. CONCLUSIONS The study found a strong and positive correlation between ferritin and hepcidin levels in iron-deficient children, but not between ERFE levels, suggesting that hepcidin is largely regulated by iron deposition levels. In addition, there was an increase in ferritin and hepcidin levels after iron treatment. The study found no significant difference in erythroferrone levels between the iron-deficient group and the control group. It is thought that this may be due to the short duration of iron treatment given to the patients with iron deficiency anemia included in the study.
Collapse
Affiliation(s)
- Ramazan Dulkadir
- Ahi Evran Training and Research Hospital Pediatrics Clinic Bagbasi, Kırşehir Ahi Evran University, Kirsehir, Turkey.
| | - Gamze Turna Saltoğlu
- Ahi Evran Training and Research Hospital Pediatrics Clinic Bagbasi, Kırşehir Ahi Evran University, Kirsehir, Turkey
| | - Ali Güneş
- Ahi Evran Training and Research Hospital Pediatrics Clinic Bagbasi, Kırşehir Ahi Evran University, Kirsehir, Turkey
| |
Collapse
|
34
|
Terzi EM, Possemato R. Iron, Copper, and Selenium: Cancer's Thing for Redox Bling. Cold Spring Harb Perspect Med 2024; 14:a041545. [PMID: 37932129 PMCID: PMC10982729 DOI: 10.1101/cshperspect.a041545] [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] [Indexed: 11/08/2023]
Abstract
Cells require micronutrients for numerous basic functions. Among these, iron, copper, and selenium are particularly critical for redox metabolism, and their importance is heightened during oncogene-driven perturbations in cancer. In this review, which particularly focuses on iron, we describe how these micronutrients are carefully chaperoned about the body and made available to tissues, a process that is designed to limit the toxicity of free iron and copper or by-products of selenium metabolism. We delineate perturbations in iron metabolism and iron-dependent proteins that are observed in cancer, and describe the current approaches being used to target iron metabolism and iron-dependent processes.
Collapse
Affiliation(s)
- Erdem M Terzi
- Department of Pathology, New York University Grossman School of Medicine, New York, New York 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York, New York 10016, USA
| | - Richard Possemato
- Department of Pathology, New York University Grossman School of Medicine, New York, New York 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York, New York 10016, USA
| |
Collapse
|
35
|
Kawabata H, Miyazawa N, Matsuda Y, Satobe M, Mizogami Y, Kuriyama Y, Sakai T, Mori M, Sasa M. Measurement of serum hepcidin-25 by latex agglutination in healthy volunteers and patients with hematologic disorders. Int J Hematol 2024; 119:392-398. [PMID: 38372875 DOI: 10.1007/s12185-024-03720-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/20/2024]
Abstract
Iron is an essential trace metal, vital for various physiologic processes, but excess levels can harm health. Maintaining iron homeostasis is critical, with hepcidin playing a key role. The isoform hepcidin-25 exerts the most significant influence on iron metabolism, making its serum levels a valuable diagnostic tool. However, mass-spectrometry and other conventional measurement methods can be difficult to perform, and some immunoassays lack reliability. In this study, we employed a recently developed latex agglutination method integrated with a readily available automated analyzer to quantify serum hepcidin-25 levels in both volunteers recruited from personnel of our hospital (n = 93) and patients with various hematological disorders (n = 112). Our findings unveiled a robust positive correlation between serum hepcidin-25 and ferritin, as well as C-reactive protein levels, in both volunteers and patients. Among the patients with hematological disorders, there was a noteworthy negative correlation between hepcidin-25 levels and hemoglobin concentrations, as well as reticulocyte counts. Interestingly, the hepcidin-25/ferritin ratio was remarkably low in patients with hemolytic anemia and myelodysplastic syndromes with ring sideroblasts. Our findings suggest that quantifying serum hepcidin-25 and the hepcidin-25/ferritin ratio using this method may be valuable for screening of hematopoietic diseases and other iron metabolism disorders.
Collapse
Affiliation(s)
- Hiroshi Kawabata
- Department of Hematology, NHO Kyoto Medical Center, 1-1 Fukakusa Mukaihata-Cho, Fushimi-Ku, Kyoto, 612-8555, Japan.
- Department of Clinical Laboratory, NHO Kyoto Medical Center, Kyoto, Japan.
| | - Naoki Miyazawa
- Department of Clinical Laboratory, NHO Kyoto Medical Center, Kyoto, Japan
| | - Yumi Matsuda
- Department of Clinical Laboratory, NHO Kyoto Medical Center, Kyoto, Japan
| | - Misaki Satobe
- Department of Clinical Laboratory, NHO Kyoto Medical Center, Kyoto, Japan
| | - Yasushi Mizogami
- Department of Clinical Laboratory, NHO Kyoto Medical Center, Kyoto, Japan
| | - Yoko Kuriyama
- Department of Clinical Laboratory, NHO Kyoto Medical Center, Kyoto, Japan
| | - Tomomi Sakai
- Department of Hematology, NHO Kyoto Medical Center, 1-1 Fukakusa Mukaihata-Cho, Fushimi-Ku, Kyoto, 612-8555, Japan
| | - Minako Mori
- Department of Hematology, NHO Kyoto Medical Center, 1-1 Fukakusa Mukaihata-Cho, Fushimi-Ku, Kyoto, 612-8555, Japan
| | - Michio Sasa
- Department of Clinical Laboratory, NHO Kyoto Medical Center, Kyoto, Japan
| |
Collapse
|
36
|
Sardo U, Perrier P, Cormier K, Sotin M, Personnaz J, Medjbeur T, Desquesnes A, Cannizzo L, Ruiz-Martinez M, Thevenin J, Billoré B, Jung G, Abboud E, Peyssonnaux C, Nemeth E, Ginzburg YZ, Ganz T, Kautz L. The hepatokine FGL1 regulates hepcidin and iron metabolism during anemia in mice by antagonizing BMP signaling. Blood 2024; 143:1282-1292. [PMID: 38232308 PMCID: PMC11103088 DOI: 10.1182/blood.2023022724] [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: 09/27/2023] [Revised: 11/20/2023] [Accepted: 12/13/2023] [Indexed: 01/19/2024] Open
Abstract
ABSTRACT As a functional component of erythrocyte hemoglobin, iron is essential for oxygen delivery to all tissues in the body. The liver-derived peptide hepcidin is the master regulator of iron homeostasis. During anemia, the erythroid hormone erythroferrone regulates hepcidin synthesis to ensure the adequate supply of iron to the bone marrow for red blood cell production. However, mounting evidence suggested that another factor may exert a similar function. We identified the hepatokine fibrinogen-like 1 (FGL1) as a previously undescribed suppressor of hepcidin that is induced in the liver in response to hypoxia during the recovery from anemia, and in thalassemic mice. We demonstrated that FGL1 is a potent suppressor of hepcidin in vitro and in vivo. Deletion of Fgl1 in mice results in higher hepcidin levels at baseline and after bleeding. FGL1 exerts its activity by directly binding to bone morphogenetic protein 6 (BMP6), thereby inhibiting the canonical BMP-SMAD signaling cascade that controls hepcidin transcription.
Collapse
Affiliation(s)
- Ugo Sardo
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Prunelle Perrier
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Kevin Cormier
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Manon Sotin
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Jean Personnaz
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Thanina Medjbeur
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Aurore Desquesnes
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Lisa Cannizzo
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | | | - Julie Thevenin
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Benjamin Billoré
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Grace Jung
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Elise Abboud
- Institut Cochin, INSERM, Centre National de la Recherche Scientifique, Université de Paris, Paris, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Carole Peyssonnaux
- Institut Cochin, INSERM, Centre National de la Recherche Scientifique, Université de Paris, Paris, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Elizabeta Nemeth
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | | | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Department of Pathology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Léon Kautz
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III Paul Sabatier, Toulouse, France
| |
Collapse
|
37
|
Affiliation(s)
- Laura Silvestri
- Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele and Vita-Salute San Raffaele University
| |
Collapse
|
38
|
Matsuoka T, Abe M, Kobayashi H. Iron Metabolism and Inflammatory Mediators in Patients with Renal Dysfunction. Int J Mol Sci 2024; 25:3745. [PMID: 38612557 PMCID: PMC11012052 DOI: 10.3390/ijms25073745] [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: 01/31/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Chronic kidney disease (CKD) affects around 850 million people worldwide, posing significant challenges in healthcare due to complications like renal anemia, end-stage kidney disease, and cardiovascular diseases. This review focuses on the intricate interplay between iron metabolism, inflammation, and renal dysfunction in CKD. Renal anemia, prevalent in CKD, arises primarily from diminished erythropoietin (EPO) production and iron dysregulation, which worsens with disease progression. Functional and absolute iron deficiencies due to impaired absorption and chronic inflammation are key factors exacerbating erythropoiesis. A notable aspect of CKD is the accumulation of uremic toxins, such as indoxyl sulfate (IS), which hinder iron metabolism and worsen anemia. These toxins directly affect renal EPO synthesis and contribute to renal hypoxia, thus playing a critical role in the pathophysiology of renal anemia. Inflammatory cytokines, especially TNF-α and IL-6, further exacerbate CKD progression and disrupt iron homeostasis, thereby influencing anemia severity. Treatment approaches have evolved to address both iron and EPO deficiencies, with emerging therapies targeting hepcidin and employing hypoxia-inducible factor (HIF) stabilizers showing potential. This review underscores the importance of integrated treatment strategies in CKD, focusing on the complex relationship between iron metabolism, inflammation, and renal dysfunction to improve patient outcomes.
Collapse
Affiliation(s)
| | | | - Hiroki Kobayashi
- Division of Nephrology, Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine, Tokyo 173-8610, Japan
| |
Collapse
|
39
|
Rivella S. Optimizing lentiviral genomic integrations to cure beta-thalassemia: The least required for success? Mol Ther Methods Clin Dev 2024; 32:101222. [PMID: 38463140 PMCID: PMC10924054 DOI: 10.1016/j.omtm.2024.101222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Affiliation(s)
- Stefano Rivella
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Cell and Molecular Biology Affinity Group (CAMB), University of Pennsylvania, Philadelphia, PA, USA
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, CHOP, Philadelphia, PA, USA
- Penn Center for Musculoskeletal Disorders, CHOP, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- RNA Institute, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
40
|
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
|
41
|
Sun Y, Ren Y, Song LY, Wang YY, Li TG, Wu YL, Li L, Yang ZS. Targeting iron-metabolism:a potential therapeutic strategy for pulmonary fibrosis. Biomed Pharmacother 2024; 172:116270. [PMID: 38364737 DOI: 10.1016/j.biopha.2024.116270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/23/2024] [Accepted: 02/07/2024] [Indexed: 02/18/2024] Open
Abstract
Iron homeostasisis is integral to normal physiological and biochemical processes of lungs. The maintenance of iron homeostasis involves the process of intake, storage and output, dependening on iron-regulated protein/iron response element system to operate tightly metabolism-related genes, including TFR1, DMT1, Fth, and FPN. Dysregulation of iron can lead to iron overload, which increases the virulence of microbial colonisers and the occurrence of oxidative stress, causing alveolar epithelial cells to undergo necrosis and apoptosis, and form extracellular matrix. Accumulated iron drive iron-dependent ferroptosis to exacerbated pulmonary fibrosis. Notably, the iron chelator deferoxamine and the lipophilic antioxidant ferritin-1 have been shown to attenuate ferroptosis and inhibit lipid peroxidation in pulmonary fibrosis. The paper summarises the regulatory mechanisms of dysregulated iron metabolism and ferroptosis in the development of pulmonary fibrosis. Targeting iron metabolism may be a potential therapeutic strategy for the prevention and treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Yi Sun
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming, 650500, Yunnan, China
| | - Yu Ren
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming, 650500, Yunnan, China
| | - Li-Yun Song
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming, 650500, Yunnan, China
| | - Yin-Ying Wang
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, 1076 Yuhua Road Kunming, Yunnan 650500, China
| | - Tian-Gang Li
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming, 650500, Yunnan, China
| | - Ying-Li Wu
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming, 650500, Yunnan, China
| | - Li Li
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming, 650500, Yunnan, China.
| | - Zhong-Shan Yang
- Yunnan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, School of Basic Medical Sciences, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming, 650500, Yunnan, China.
| |
Collapse
|
42
|
Ebea PO, Vidyasagar S, Connor JR, Frazer DM, Knutson MD, Collins JF. Oral iron therapy: Current concepts and future prospects for improving efficacy and outcomes. Br J Haematol 2024; 204:759-773. [PMID: 38253961 PMCID: PMC10939879 DOI: 10.1111/bjh.19268] [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: 05/24/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 01/24/2024]
Abstract
Iron deficiency (ID) and iron-deficiency anaemia (IDA) are global public health concerns, most commonly afflicting children, pregnant women and women of childbearing age. Pathological outcomes of ID include delayed cognitive development in children, adverse pregnancy outcomes and decreased work capacity in adults. IDA is usually treated by oral iron supplementation, typically using iron salts (e.g. FeSO4 ); however, dosing at several-fold above the RDA may be required due to less efficient absorption. Excess enteral iron causes adverse gastrointestinal side effects, thus reducing compliance, and negatively impacts the gut microbiome. Recent research has sought to identify new iron formulations with better absorption so that lower effective dosing can be utilized. This article outlines emerging research on oral iron supplementation and focuses on molecular mechanisms by which different supplemental forms of iron are transported across the intestinal epithelium and whether these transport pathways are subject to regulation by the iron-regulatory hormone hepcidin.
Collapse
Affiliation(s)
- Pearl O. Ebea
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | | | - James R. Connor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
| | - David M. Frazer
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Mitchell D. Knutson
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, USA
| | - James F. Collins
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL, USA
| |
Collapse
|
43
|
Garbowski MW, Ugidos M, Risueño A, Shetty JK, Schwickart M, Hermine O, Porter JB, Thakurta A, Vodala S. Luspatercept stimulates erythropoiesis, increases iron utilization, and redistributes body iron in transfusion-dependent thalassemia. Am J Hematol 2024; 99:182-192. [PMID: 37782758 DOI: 10.1002/ajh.27102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 09/06/2023] [Accepted: 09/09/2023] [Indexed: 10/04/2023]
Abstract
Luspatercept, a ligand-trapping fusion protein, binds select TGF-β superfamily ligands implicated in thalassemic erythropoiesis, promoting late-stage erythroid maturation. Luspatercept reduced transfusion burden in the BELIEVE trial (NCT02604433) of 336 adults with transfusion-dependent thalassemia (TDT). Analysis of biomarkers in BELIEVE offers novel physiological and clinical insights into benefits offered by luspatercept. Transfusion iron loading rates decreased 20% by 1.4 g (~7 blood units; median iron loading rate difference: -0.05 ± 0.07 mg Fe/kg/day, p< .0001) and serum ferritin (s-ferritin) decreased 19.2% by 269.3 ± 963.7 μg/L (p < .0001), indicating reduced macrophage iron. However, liver iron content (LIC) did not decrease but showed statistically nonsignificant increases from 5.3 to 6.7 mg/g dw. Erythropoietin, growth differentiation factor 15, soluble transferrin receptor 1 (sTfR1), and reticulocytes rose by 93%, 59%, 66%, and 112%, respectively; accordingly, erythroferrone increased by 51% and hepcidin decreased by 53% (all p < .0001). Decreased transfusion with luspatercept in patients with TDT was associated with increased erythropoietic markers and decreasing hepcidin. Furthermore, s-ferritin reduction associated with increased erythroid iron incorporation (marked by sTfR1) allowed increased erythrocyte marrow output, consequently reducing transfusion needs and enhancing rerouting of hemolysis (heme) iron and non-transferrin-bound iron to the liver. LIC increased in patients with intact spleens, consistent with iron redistribution given the hepcidin reduction. Thus, erythropoietic and hepcidin changes with luspatercept in TDT lower transfusion dependency and may redistribute iron from macrophages to hepatocytes, necessitating the use of concomitant chelator cover for effective iron management.
Collapse
Affiliation(s)
- Maciej W Garbowski
- UCL Cancer Institute Hematology Department, University College London, University College London Hospitals, London, UK
| | - Manuel Ugidos
- BMS Center for Innovation and Translational Research Europe (CITRE), Bristol Myers Squibb, Seville, Spain
| | - Alberto Risueño
- BMS Center for Innovation and Translational Research Europe (CITRE), Bristol Myers Squibb, Seville, Spain
| | - Jeevan K Shetty
- Celgene International Sàrl a Bristol-Myers Squibb Company, Boudry, Switzerland
| | | | - Olivier Hermine
- Necker Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Imagine Institute, INSERM Unité 1163, University of Paris, Paris, France
| | - John B Porter
- UCL Cancer Institute Hematology Department, University College London, University College London Hospitals, London, UK
| | | | | |
Collapse
|
44
|
Galy B, Conrad M, Muckenthaler M. Mechanisms controlling cellular and systemic iron homeostasis. Nat Rev Mol Cell Biol 2024; 25:133-155. [PMID: 37783783 DOI: 10.1038/s41580-023-00648-1] [Citation(s) in RCA: 123] [Impact Index Per Article: 123.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 10/04/2023]
Abstract
In mammals, hundreds of proteins use iron in a multitude of cellular functions, including vital processes such as mitochondrial respiration, gene regulation and DNA synthesis or repair. Highly orchestrated regulatory systems control cellular and systemic iron fluxes ensuring sufficient iron delivery to target proteins is maintained, while limiting its potentially deleterious effects in iron-mediated oxidative cell damage and ferroptosis. In this Review, we discuss how cells acquire, traffick and export iron and how stored iron is mobilized for iron-sulfur cluster and haem biogenesis. Furthermore, we describe how these cellular processes are fine-tuned by the combination of various sensory and regulatory systems, such as the iron-regulatory protein (IRP)-iron-responsive element (IRE) network, the nuclear receptor co-activator 4 (NCOA4)-mediated ferritinophagy pathway, the prolyl hydroxylase domain (PHD)-hypoxia-inducible factor (HIF) axis or the nuclear factor erythroid 2-related factor 2 (NRF2) regulatory hub. We further describe how these pathways interact with systemic iron homeostasis control through the hepcidin-ferroportin axis to ensure appropriate iron fluxes. This knowledge is key for the identification of novel therapeutic opportunities to prevent diseases of cellular and/or systemic iron mismanagement.
Collapse
Affiliation(s)
- Bruno Galy
- German Cancer Research Center (DKFZ), Division of Virus-associated Carcinogenesis (F170), Heidelberg, Germany
| | - Marcus Conrad
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Neuherberg, Germany
| | - Martina Muckenthaler
- Department of Paediatric Hematology, Oncology and Immunology, University of Heidelberg, Heidelberg, Germany.
- Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner site Heidelberg/Mannheim, Heidelberg, Germany.
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
45
|
Tseng YJ, Kageyama Y, Murdaugh RL, Kitano A, Kim JH, Hoegenauer KA, Tiessen J, Smith MH, Uryu H, Takahashi K, Martin JF, Samee MAH, Nakada D. Increased iron uptake by splenic hematopoietic stem cells promotes TET2-dependent erythroid regeneration. Nat Commun 2024; 15:538. [PMID: 38225226 PMCID: PMC10789814 DOI: 10.1038/s41467-024-44718-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 01/02/2024] [Indexed: 01/17/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are capable of regenerating the blood system, but the instructive cues that direct HSCs to regenerate particular lineages lost to the injury remain elusive. Here, we show that iron is increasingly taken up by HSCs during anemia and induces erythroid gene expression and regeneration in a Tet2-dependent manner. Lineage tracing of HSCs reveals that HSCs respond to hemolytic anemia by increasing erythroid output. The number of HSCs in the spleen, but not bone marrow, increases upon anemia and these HSCs exhibit enhanced proliferation, erythroid differentiation, iron uptake, and TET2 protein expression. Increased iron in HSCs promotes DNA demethylation and expression of erythroid genes. Suppressing iron uptake or TET2 expression impairs erythroid genes expression and erythroid differentiation of HSCs; iron supplementation, however, augments these processes. These results establish that the physiological level of iron taken up by HSCs has an instructive role in promoting erythroid-biased differentiation of HSCs.
Collapse
Affiliation(s)
- Yu-Jung Tseng
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yuki Kageyama
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Rebecca L Murdaugh
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ayumi Kitano
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jong Hwan Kim
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kevin A Hoegenauer
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jonathan Tiessen
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Mackenzie H Smith
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Hidetaka Uryu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - James F Martin
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, 77030, USA
| | - Md Abul Hassan Samee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Daisuke Nakada
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
46
|
Appleby S, Frampton C, Holdaway M, Chew-Harris J, Liew OW, Chong JPC, Lewis L, Troughton R, Ooi SBS, Kuan WS, Ibrahim I, Chan SP, Richards AM, Pemberton CJ. Circulating erythroferrone has diagnostic utility for acute decompensated heart failure in patients presenting with acute or worsening dyspnea. Front Cardiovasc Med 2024; 10:1195082. [PMID: 38259307 PMCID: PMC10800458 DOI: 10.3389/fcvm.2023.1195082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Objectives In dyspneic patients with atrial fibrillation (AF) or obesity, the diagnostic performance of NT-proBNP for acute heart failure is reduced. We evaluated the erythroblast derived protein erythroferrone (ERFE) as an ancillary biomarker for the diagnosis of acute decompensated heart failure (ADHF) in these comorbid subgroups in both Western and Asian populations. Methods The diagnostic performance of ERFE (Intrinsic Lifesciences) and NT-proBNP (Roche Cobas e411) for ADHF was assessed in 479 New Zealand (NZ) and 475 Singapore (SG) patients presenting with breathlessness. Results Plasma ERFE was higher in ADHF, compared with breathlessness from other causes, in both countries (NZ; 4.9 vs. 1.4 ng/ml, p < 0.001) and (SG; 4.2 vs. 0.4 ng/ml, p = 0.021). The receiver operating characteristic (ROC) areas under the curve (AUCs) for discrimination of ADHF were reduced in the NZ cohort compared to SG for ERFE (0.75 and 0.84, p = 0.007) and NT-proBNP (0.86 and 0.92, p = 0.004). Optimal cut-off points for ERFE yielded comparable sensitivity and positive predictive values in both cohorts, but slightly better specificity, negative predictive values and accuracy in SG compared with NZ. In patients with AF, the AUC decreased for ERFE in each cohort (NZ: 0.71, n = 105, SG: 0.61, n = 44) but increased in patients with obesity (NZ: 0.79, n = 150, SG: 0.87, n = 164). Conclusions Circulating ERFE is higher in patients with ADHF than in other causes of new onset breathlessness with fair diagnostic utility, performing better in Asian than in Western patients. The diagnostic performance of ERFE is impaired in patients with AF but not patients with obesity.
Collapse
Affiliation(s)
- Sarah Appleby
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Chris Frampton
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Mark Holdaway
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Janice Chew-Harris
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Oi Wah Liew
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| | - Jenny Pek Ching Chong
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| | - Lynley Lewis
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| | - Richard Troughton
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
- Department of Cardiology, Te Whatu Ora, Christchurch, New Zealand
| | | | - Win Sen Kuan
- Emergency Department, National University Hospital, Singapore, Singapore
| | - Irwani Ibrahim
- Emergency Department, National University Hospital, Singapore, Singapore
| | - Siew Pang Chan
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
| | - A. Mark Richards
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
- Cardiovascular Research Institute, National University of Singapore, Singapore, Singapore
- Department of Cardiology, Te Whatu Ora, Christchurch, New Zealand
| | - Christopher J. Pemberton
- Department of Medicine, Christchurch Heart Institute, University of Otago Christchurch, Christchurch, New Zealand
| |
Collapse
|
47
|
McKay AKA, Anderson B, Peeling P, Whitfield J, Tee N, Zeder C, Zimmermann MB, Burke LM, Moretti D. Iron Absorption in Highly Trained Male Runners: Does it Matter When and Where You Eat Your Iron? Med Sci Sports Exerc 2024; 56:118-127. [PMID: 38098150 DOI: 10.1249/mss.0000000000003272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
PURPOSE We examined iron absorption and its regulation during two common scenarios experienced by endurance athletes. Our aims were to: (i) compare the effects of preexercise versus postexercise iron intake on iron absorption; and (ii) compare the impact of training at altitude (1800 m) on iron absorption preexercise. METHODS Male runners (n = 18) completed three exercise trials over a 5-wk period, each preceded by 24 h of standardized low-iron diets. First, athletes completed two 60-min treadmill running trials at 65% V̇O2max at near sea-level (580 m). In a randomized order, preexercise and postexercise test meals labeled with 4 mg of 57Fe or 58Fe were consumed 30 min before or 30 min after exercise. Then, the same exercise trial was performed after living and training at altitude (~1800 m) for 7 d, with the labeled test meal consumed 30 min preexercise. We collected venous blood samples preexercise and postexercise for markers of iron status and regulation, and 14 d later to measure erythrocyte isotope incorporation. RESULTS No differences in fractional iron absorption were evident when test meals were consumed preexercise (7.3% [4.4, 12.1]) or postexercise (6.2% [3.1, 12.5]) (n = 18; P = 0.058). Iron absorption preexercise was greater at altitude (18.4% [10.6, 32.0]) than at near sea-level (n = 17; P < 0.001) and hepcidin concentrations at altitude were lower at rest and 3 h postexercise compared with near sea level (P < 0.001). CONCLUSIONS In an acute setting, preexercise and postexercise iron absorption is comparable if consumed within 30 min of exercise. Preexercise iron absorption increases 2.6-fold at altitude compared with near sea-level, likely due to the homeostatic response to provide iron for enhanced erythropoiesis and maintain iron stores.
Collapse
Affiliation(s)
- Alannah K A McKay
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Vic, AUSTRALIA
| | | | | | - Jamie Whitfield
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Vic, AUSTRALIA
| | - Nicolin Tee
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Vic, AUSTRALIA
| | - Christophe Zeder
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, ETH Zürich, Zurich, SWITZERLAND
| | - Michael B Zimmermann
- Medical Research Council Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UNITED KINGDOM
| | - Louise M Burke
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Vic, AUSTRALIA
| | - Diego Moretti
- Nutrition Group, Health Department, Swiss Distance University of Applied Sciences (FFHS), Zürich, SWITZERLAND
| |
Collapse
|
48
|
Mast JF, Leach EAE, Thompson TB. Characterization of erythroferrone oligomerization and its impact on BMP antagonism. J Biol Chem 2024; 300:105452. [PMID: 37949218 PMCID: PMC10772735 DOI: 10.1016/j.jbc.2023.105452] [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: 07/30/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/12/2023] Open
Abstract
Hepcidin, a peptide hormone that negatively regulates iron metabolism, is expressed by bone morphogenetic protein (BMP) signaling. Erythroferrone (ERFE) is an extracellular protein that binds and inhibits BMP ligands, thus positively regulating iron import by indirectly suppressing hepcidin. This allows for rapid erythrocyte regeneration after blood loss. ERFE belongs to the C1Q/TNF-related protein family and is suggested to adopt multiple oligomeric forms: a trimer, a hexamer, and a high molecular weight species. The molecular basis for how ERFE binds BMP ligands and how the different oligomeric states impact BMP inhibition are poorly understood. In this study, we demonstrated that ERFE activity is dependent on the presence of stable dimeric or trimeric ERFE and that larger species are dispensable for BMP inhibition. Additionally, we used an in silico approach to identify a helix, termed the ligand-binding domain, that was predicted to bind BMPs and occlude the type I receptor pocket. We provide evidence that the ligand-binding domain is crucial for activity through luciferase assays and surface plasmon resonance analysis. Our findings provide new insight into how ERFE oligomerization impacts BMP inhibition, while identifying critical molecular features of ERFE essential for binding BMP ligands.
Collapse
Affiliation(s)
- Jacob F Mast
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, Ohio, USA
| | - Edmund A E Leach
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, Ohio, USA
| | - Thomas B Thompson
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, Ohio, USA.
| |
Collapse
|
49
|
Rostoker G, Dekeyser M, Francisco S, Loridon C, Griuncelli M, Languille-Llitjos E, Boulahia G, Cohen Y. Relationship between bone marrow iron load and liver iron concentration in dialysis-associated haemosiderosis. EBioMedicine 2024; 99:104929. [PMID: 38128412 PMCID: PMC10776950 DOI: 10.1016/j.ebiom.2023.104929] [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: 06/14/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Iron overload due to the excessive use of parenteral iron in haemodialysis is now an increasingly recognised clinical issue. Before erythropoiesis-stimulating agents (ESA) were introduced, a specific feature of patients treated by dialysis and having iron overload was that iron levels in the bone marrow were paradoxically low in most of them, despite severe hepatosplenic siderosis. Whether or not this paradox persists in the actual ESA era was unknown until recently, when an autopsy study in 21 patients treated by haemodialysis revealed similarities between liver and bone marrow iron content. The aim of this study was to further explore these recent findings in a cohort of alive patients on dialysis and to analyse the determinants of iron bone marrow. METHODS Liver iron concentration (LIC) and vertebral T2∗ (a surrogate marker of bone marrow iron) were analysed retrospectively in 152 alive patients on dialysis (38.8% female) of whom 47.4% had iron overload by quantitative magnetic resonance imaging (MRI). FINDINGS Vertebral T2∗ differed significantly between patients classified according to liver iron content at MRI: those with mild or moderate and severe liver iron overload had increased vertebral iron content at R2∗ relaxometry MRI (mild: vertebral T2∗ = 9.9 ms (4-24.8); moderate and severe: vertebral T2∗ = 8.5 ms (4.9-22.8)) when compared to patients with normal LIC (vertebral T2∗ = 13.2 ms (6.6-30.5) (p < 0.0001 Kruskal-Wallis test)). INTERPRETATION The paradoxical discrepancy between bone marrow and liver iron-storage compartments observed in the pre-ESA era has disappeared today, as shown by a recent autopsy study and the present study in a cohort of alive patients treated by dialysis. FUNDING None.
Collapse
Affiliation(s)
- Guy Rostoker
- Division of Nephrology and Dialysis, Ramsay Santé, Hôpital Privé Claude Galien, Quincy-sous-Sénart 91480, France; Collège de Médecine des Hôpitaux de Paris, 10 Rue des Fossés Saint-Marcel, Paris 75005, France.
| | - Manon Dekeyser
- Department of Nephrology, Regional University Centre, Orléans and INSERM 1186, Gustave Roussy Institute, Paris-Saclay University, Villejuif, Paris, France
| | - Sergio Francisco
- Division of Radiology, Ramsay Santé, Hôpital Privé Claude Galien, Quincy-sous-Sénart 91480, France
| | - Christelle Loridon
- Division of Nephrology and Dialysis, Ramsay Santé, Hôpital Privé Claude Galien, Quincy-sous-Sénart 91480, France
| | - Mireille Griuncelli
- Division of Nephrology and Dialysis, Ramsay Santé, Hôpital Privé Claude Galien, Quincy-sous-Sénart 91480, France
| | - Eva Languille-Llitjos
- Division of Nephrology and Dialysis, Ramsay Santé, Hôpital Privé Claude Galien, Quincy-sous-Sénart 91480, France
| | - Ghada Boulahia
- Division of Nephrology and Dialysis, Ramsay Santé, Hôpital Privé Claude Galien, Quincy-sous-Sénart 91480, France
| | - Yves Cohen
- Division of Radiology, Ramsay Santé, Hôpital Privé Claude Galien, Quincy-sous-Sénart 91480, France
| |
Collapse
|
50
|
Krumm B, Saugy JJ, Botrè F, Donati F, Faiss R. Indirect biomarkers of blood doping: A systematic review. Drug Test Anal 2024; 16:49-64. [PMID: 37160638 DOI: 10.1002/dta.3514] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/13/2023] [Accepted: 05/02/2023] [Indexed: 05/11/2023]
Abstract
The detection of blood doping represents a current major issue in sports and an ongoing challenge for antidoping research. Initially focusing on direct detection methods to identify a banned substance or its metabolites, the antidoping effort has been progressively complemented by indirect approaches. The longitudinal and individual monitoring of specific biomarkers aims to identify nonphysiological variations that may be related to doping practices. From this perspective, the identification of markers sensitive to erythropoiesis alteration is key in the screening of blood doping. The current Athlete Biological Passport implemented since 2009 is composed of 14 variables (including two primary markers, i.e., hemoglobin concentration and OFF score) for the hematological module to be used for indirect detection of blood doping. Nevertheless, research has continually proposed and investigated new markers sensitive to an alteration of the erythropoietic cascade and specific to blood doping. If multiple early markers have been identified (at the transcriptomic level) or developed directly in a diagnostics' kit (at a proteomic level), other target variables at the end of the erythropoietic process (linked with the red blood cell functions) may strengthen the hematological module in the future. Therefore, this review aims to provide a global systematic overview of the biomarkers considered to date in the indirect investigation of blood doping.
Collapse
Affiliation(s)
- Bastien Krumm
- REDs, Research & Expertise in AntiDoping Sciences, Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Jonas J Saugy
- REDs, Research & Expertise in AntiDoping Sciences, Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Francesco Botrè
- REDs, Research & Expertise in AntiDoping Sciences, Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
- Laboratorio Antidoping, Federazione Medico Sportiva Italiana, Rome, Italy
| | - Francesco Donati
- Laboratorio Antidoping, Federazione Medico Sportiva Italiana, Rome, Italy
| | - Raphael Faiss
- REDs, Research & Expertise in AntiDoping Sciences, Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|