51
|
Nai A, Cordero-Sanchez C, Tanzi E, Pagani A, Silvestri L, Di Modica SM. Cellular and animal models for the investigation of β-thalassemia. Blood Cells Mol Dis 2024; 104:102761. [PMID: 37271682 DOI: 10.1016/j.bcmd.2023.102761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/06/2023]
Abstract
β-Thalassemia is a genetic form of anemia due to mutations in the β-globin gene, that leads to ineffective and extramedullary erythropoiesis, abnormal red blood cells and secondary iron-overload. The severity of the disease ranges from mild to lethal anemia based on the residual levels of globins production. Despite being a monogenic disorder, the pathophysiology of β-thalassemia is multifactorial, with different players contributing to the severity of anemia and secondary complications. As a result, the identification of effective therapeutic strategies is complex, and the treatment of patients is still suboptimal. For these reasons, several models have been developed in the last decades to provide experimental tools for the study of the disease, including erythroid cell lines, cultures of primary erythroid cells and transgenic animals. Years of research enabled the optimization of these models and led to decipher the mechanisms responsible for globins deregulation and ineffective erythropoiesis in thalassemia, to unravel the role of iron homeostasis in the disease and to identify and validate novel therapeutic targets and agents. Examples of successful outcomes of these analyses include iron restricting agents, currently tested in the clinics, several gene therapy vectors, one of which was recently approved for the treatment of most severe patients, and a promising gene editing strategy, that has been shown to be effective in a clinical trial. This review provides an overview of the available models, discusses pros and cons, and the key findings obtained from their study.
Collapse
Affiliation(s)
- Antonella Nai
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, via Olgettina 60, Milan, Italy; Vita-Salute San Raffaele University, via Olgettina 58, Milan, Italy.
| | - Celia Cordero-Sanchez
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, via Olgettina 60, Milan, Italy
| | - Emanuele Tanzi
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, via Olgettina 60, Milan, Italy
| | - Alessia Pagani
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, via Olgettina 60, Milan, Italy
| | - Laura Silvestri
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, via Olgettina 60, Milan, Italy; Vita-Salute San Raffaele University, via Olgettina 58, Milan, Italy
| | - Simona Maria Di Modica
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, via Olgettina 60, Milan, Italy
| |
Collapse
|
52
|
An W, Feola M, Levy M, Aluri S, Ruiz-Martinez M, Sridharan A, Fibach E, Zhu X, Verma A, Ginzburg Y. Iron chelation improves ineffective erythropoiesis and iron overload in myelodysplastic syndrome mice. eLife 2023; 12:e83103. [PMID: 38153418 PMCID: PMC10754500 DOI: 10.7554/elife.83103] [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: 08/31/2022] [Accepted: 11/27/2023] [Indexed: 12/29/2023] Open
Abstract
Myelodysplastic syndrome (MDS) is a heterogeneous group of bone marrow stem cell disorders characterized by ineffective hematopoiesis and cytopenias, most commonly anemia. Red cell transfusion therapy for anemia in MDS results in iron overload, correlating with reduced overall survival. Whether the treatment of iron overload benefits MDS patients remains controversial. We evaluate underlying iron-related pathophysiology and the effect of iron chelation using deferiprone on erythropoiesis in NUP98-HOXD13 transgenic mice, a highly penetrant well-established MDS mouse model. Our results characterize an iron overload phenotype with aberrant erythropoiesis in these mice which was reversed by deferiprone-treatment. Serum erythropoietin levels decreased while erythroblast erythropoietin receptor expression increased in deferiprone-treated MDS mice. We demonstrate, for the first time, normalized expression of the iron chaperones Pcbp1 and Ncoa4 and increased ferritin stores in late-stage erythroblasts from deferiprone-treated MDS mice, evidence of aberrant iron trafficking in MDS erythroblasts. Importantly, erythroblast ferritin is increased in response to deferiprone, correlating with decreased erythroblast ROS. Finally, we confirmed increased expression of genes involved in iron uptake, sensing, and trafficking in stem and progenitor cells from MDS patients. Taken together, our findings provide evidence that erythroblast-specific iron metabolism is a novel potential therapeutic target to reverse ineffective erythropoiesis in MDS.
Collapse
Affiliation(s)
- Wenbin An
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Division of Pediatric Blood Diseases Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Maria Feola
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Maayan Levy
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Srinivas Aluri
- Division of Hematology and Medical Oncology, Albert Einstein College of MedicineBronxUnited States
| | - Marc Ruiz-Martinez
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Ashwin Sridharan
- Division of Hematology and Medical Oncology, Albert Einstein College of MedicineBronxUnited States
| | - Eitan Fibach
- Department of Hematology, Hadassah Medical Center, Hebrew UniversityJerusalemIsrael
| | - Xiaofan Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Division of Pediatric Blood Diseases Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
| | - Amit Verma
- Division of Hematology and Medical Oncology, Albert Einstein College of MedicineBronxUnited States
| | - Yelena Ginzburg
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| |
Collapse
|
53
|
Mina E, Wyart E, Sartori R, Angelino E, Zaggia I, Rausch V, Maldotti M, Pagani A, Hsu MY, Friziero A, Sperti C, Menga A, Graziani A, Hirsch E, Oliviero S, Sandri M, Conti L, Kautz L, Silvestri L, Porporato PE. FK506 bypasses the effect of erythroferrone in cancer cachexia skeletal muscle atrophy. Cell Rep Med 2023; 4:101306. [PMID: 38052214 PMCID: PMC10772350 DOI: 10.1016/j.xcrm.2023.101306] [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: 02/22/2023] [Revised: 09/29/2023] [Accepted: 11/07/2023] [Indexed: 12/07/2023]
Abstract
Skeletal muscle atrophy is a hallmark of cachexia, a wasting condition typical of chronic pathologies, that still represents an unmet medical need. Bone morphogenetic protein (BMP)-Smad1/5/8 signaling alterations are emerging drivers of muscle catabolism, hence, characterizing these perturbations is pivotal to develop therapeutic approaches. We identified two promoters of "BMP resistance" in cancer cachexia, specifically the BMP scavenger erythroferrone (ERFE) and the intracellular inhibitor FKBP12. ERFE is upregulated in cachectic cancer patients' muscle biopsies and in murine cachexia models, where its expression is driven by STAT3. Moreover, the knock down of Erfe or Fkbp12 reduces muscle wasting in cachectic mice. To bypass the BMP resistance mediated by ERFE and release the brake on the signaling, we targeted FKBP12 with low-dose FK506. FK506 restores BMP-Smad1/5/8 signaling, rescuing myotube atrophy by inducing protein synthesis. In cachectic tumor-bearing mice, FK506 prevents muscle and body weight loss and protects from neuromuscular junction alteration, suggesting therapeutic potential for targeting the ERFE-FKBP12 axis.
Collapse
Affiliation(s)
- Erica Mina
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy
| | - Elisabeth Wyart
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy
| | - Roberta Sartori
- Department of Biomedical Sciences, University of Padova, Padova, Italy; VIMM: Veneto Institute of Molecular Medicine, Padova, Italy
| | - Elia Angelino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy; Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Ivan Zaggia
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy
| | - Valentina Rausch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy
| | - Mara Maldotti
- Department of Life Sciences and Systems Biology, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy; Italian Institute for Genomic Medicine (IIGM), Sp142 Km 3.95, 10060 Candiolo, Torino, Italy
| | - Alessia Pagani
- Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Myriam Y Hsu
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy; Division of Cell Fate Dynamics and Therapeutics, Department of Biosystems Science, Institute for Life and Medical Sciences (LiMe), Kyoto University, Kyoto, Japan
| | - Alberto Friziero
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; General Surgery 1, Padova University Hospital, Padova, Italy
| | - Cosimo Sperti
- General Surgery 2, Hepato-Pancreato-Biliary Surgery and Liver Transplantation Unit, Padova University Hospital, Padova, Italy
| | - Alessio Menga
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy
| | - Andrea Graziani
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy; Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy
| | - Salvatore Oliviero
- Department of Life Sciences and Systems Biology, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy; Italian Institute for Genomic Medicine (IIGM), Sp142 Km 3.95, 10060 Candiolo, Torino, Italy
| | - Marco Sandri
- Department of Biomedical Sciences, University of Padova, Padova, Italy; VIMM: Veneto Institute of Molecular Medicine, Padova, Italy
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy
| | - Léon Kautz
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, University Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Laura Silvestri
- Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy; Vita Salute San Raffaele University, Milan, Italy
| | - Paolo E Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center "Guido Tarone", University of Torino, 10126 Torino, Italy.
| |
Collapse
|
54
|
Yoo JJ, Cohen HJ, Artz AS, Price E, Fill JA, Prchal J, Sapp S, Barnhart H. Biomarkers of erythropoiesis response to intravenous iron in a crossover pilot study in unexplained anemia of the elderly. Hematology 2023; 28:1-8. [PMID: 37114660 DOI: 10.1080/16078454.2023.2204613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023] Open
Abstract
Anemia is common in older adults, but often unexplained. Previously, we conducted a randomized, controlled trial of intravenous (IV) iron sucrose to study its impact on the 6-minute walk test and hemoglobin in older adults with unexplained anemia and ferritin levels of 20-200 ng/mL. In this report, we present for the first time the response of hemoglobin, as well as the dynamic response of biomarkers of erythropoiesis and iron indices, in a pooled analysis of the initially IV iron-treated group of 9 subjects and the subsequently IV iron treated 10 subjects from the delayed treatment group. We hypothesized that there would be a reproducible hemoglobin response from IV iron, and that iron indices and erythropoietic markers would reflect appropriate iron loading and reduced erythropoietic stress. To investigate the biochemical response of anemia to IV iron, we studied the dynamics of soluble transferrin receptor (STfR), hepcidin, erythropoietin (EPO), and iron indices over 12 weeks after treatment. In total, all 19 treated subjects were evaluable: 9 from initial treatment and 10 after cross-over. Hemoglobin rose from 11.0 to 11.7 g/dL, 12 weeks after initiating IV iron treatment of 1000 mg divided weekly over 5 weeks. We found early changes of iron loading after 1-2 IV iron dose: serum iron increased by 184 mcg/dL from a baseline of 66 mcg/dL, ferritin by 184 ng/mL from 68 ng/mL, and hepcidin by 7.49 ng/mL from 19.2 ng/mL, while STfR and serum EPO declined by 0.55 mg/L and 3.5 mU/mL from 19.2 ng/mL and 14 mU/mL, respectively. The erythroid response and evidence of enhanced iron trafficking are consistent with the hypothesis that IV iron overcomes iron deficient or iron-restricted erythropoiesis. These data provide new insight that iron-restricted erythropoiesis is a potential and targetable mechanism for patients diagnosed with unexplained anemia of the elderly and offers support for larger prospective trials of IV iron among anemic older adults of low to normal ferritin.
Collapse
Affiliation(s)
- Justin J Yoo
- Duke University School of Medicine, Durham, NC, USA
| | - Harvey J Cohen
- Center for the Study of Aging, Duke University School of Medicine, Durham, NC, USA
| | - Andrew S Artz
- Department of Hematology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Elizabeth Price
- Division of Hematology, Stanford University Medical Center, Palo Alto, NC, USA
| | | | - Josef Prchal
- University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Shelly Sapp
- Duke University School of Medicine, Clinical Research Institute, Durham, NC, USA
| | - Huiman Barnhart
- Duke University School of Medicine, Clinical Research Institute, Durham, NC, USA
| |
Collapse
|
55
|
Casimir M, Colard M, Dussiot M, Roussel C, Martinez A, Peyssonnaux C, Mayeux P, Benghiat S, Manceau S, Francois A, Marin N, Pène F, Buffet PA, Hermine O, Amireault P. Erythropoietin downregulates red blood cell clearance, increasing transfusion efficacy in severely anemic recipients. Am J Hematol 2023; 98:1923-1933. [PMID: 37792521 DOI: 10.1002/ajh.27117] [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: 08/09/2023] [Revised: 09/12/2023] [Accepted: 09/19/2023] [Indexed: 10/06/2023]
Abstract
Red blood cells (RBC) transfusion is used to alleviate symptoms and prevent complications in anemic patients by restoring oxygen delivery to tissues. RBC transfusion efficacy, that can be measured by a rise in hemoglobin (Hb) concentration, is influenced by donor-, product-, and recipient-related characteristics. In some studies, severe pre-transfusion anemia is associated with a greater than expected Hb increment following transfusion but the biological mechanism underpinning this relationship remains poorly understood. We conducted a prospective study in critically ill patients and quantified Hb increment following one RBC transfusion. In a murine model, we investigated the possibility that, in conjunction with the host erythropoietic response, the persistence of transfused donor RBC is improved to maintain a highest RBC biomass. We confirmed a correlation between a greater Hb increment and a deeper pre-transfusion anemia in a cohort of 17 patients. In the mouse model, Hb increment and post-transfusion recovery were increased in anemic recipients. Post-transfusion RBC recovery was improved in hypoxic mice or those receiving an erythropoiesis-stimulating agent and decreased in those treated with erythropoietin (EPO)-neutralizing antibodies, suggesting that EPO signaling is necessary to observe this effect. Irradiated recipients also showed decreased post-transfusion RBC recovery. The EPO-induced post-transfusion RBC recovery improvement was abrogated in irradiated or in macrophage-depleted recipients, but maintained in splenectomized recipients, suggesting a mechanism requiring erythroid progenitors and macrophages, but which is not spleen-specific. Our study highlights a physiological role of EPO in downregulating post-transfusion RBC clearance, contributing to maintain a vital RBC biomass to rapidly cope with hypoxemia.
Collapse
Affiliation(s)
- Madeleine Casimir
- Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Institut Imagine, Université Paris Cité, Paris, France
- Département d'Hématologie, Hôpital Erasme, Université Libre de Bruxelles, Bruxelles, Belgium
- Laboratory of Excellence GR-Ex, Paris, France
| | - Martin Colard
- Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Institut Imagine, Université Paris Cité, Paris, France
- Département d'Hématologie, Hôpital Erasme, Université Libre de Bruxelles, Bruxelles, Belgium
- Laboratory of Excellence GR-Ex, Paris, France
| | - Michael Dussiot
- Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Institut Imagine, Université Paris Cité, Paris, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Camille Roussel
- Laboratory of Excellence GR-Ex, Paris, France
- Université Paris Cité et Université des Antilles, INSERM, BIGR, Paris, France
- Laboratoire d'Hématologie Générale, Hôpital Universitaire Necker Enfants Malades, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Anaïs Martinez
- Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Institut Imagine, Université Paris Cité, Paris, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Carole Peyssonnaux
- Laboratory of Excellence GR-Ex, Paris, France
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | - Patrick Mayeux
- Laboratory of Excellence GR-Ex, Paris, France
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | - Samantha Benghiat
- Département d'Hématologie, Hôpital Erasme, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Sandra Manceau
- Laboratory of Excellence GR-Ex, Paris, France
- Biotherapy Department, French National Sickle Cell Disease Referral Center, Clinical Investigation Center, Hôpital Necker, Assistance-Publique Hôpitaux de Paris, Paris, France
| | - Anne Francois
- Établissement Français du Sang d'Ile de France, Site Hôpital Européen Georges Pompidou, Paris, France
| | - Nathalie Marin
- Service de Médecine Intensive-Réanimation, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Centre-Université Paris Cité, Paris, France
| | - Frédéric Pène
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
- Service de Médecine Intensive-Réanimation, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Centre-Université Paris Cité, Paris, France
| | - Pierre A Buffet
- Laboratory of Excellence GR-Ex, Paris, France
- Université Paris Cité et Université des Antilles, INSERM, BIGR, Paris, France
- Service Des Maladies Infectieuses et Tropicales, Hôpital Universitaire Necker Enfants Malades, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Olivier Hermine
- Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Institut Imagine, Université Paris Cité, Paris, France
- Laboratory of Excellence GR-Ex, Paris, France
- Département d'Hématologie, Hôpital Universitaire Necker Enfants Malades, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Pascal Amireault
- Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Institut Imagine, Université Paris Cité, Paris, France
- Laboratory of Excellence GR-Ex, Paris, France
- Université Paris Cité et Université des Antilles, INSERM, BIGR, Paris, France
| |
Collapse
|
56
|
Srole DN, Jung G, Waring AJ, Nemeth E, Ganz T. Characterization of erythroferrone structural domains relevant to its iron-regulatory function. J Biol Chem 2023; 299:105374. [PMID: 37866631 PMCID: PMC10692919 DOI: 10.1016/j.jbc.2023.105374] [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: 07/28/2023] [Revised: 09/29/2023] [Accepted: 10/12/2023] [Indexed: 10/24/2023] Open
Abstract
Iron delivery to the plasma is closely coupled to erythropoiesis, the production of red blood cells, as this process consumes most of the circulating plasma iron. In response to hemorrhage and other erythropoietic stresses, increased erythropoietin stimulates the production of the hormone erythroferrone (ERFE) by erythrocyte precursors (erythroblasts) developing in erythropoietic tissues. ERFE acts on the liver to inhibit bone morphogenetic protein (BMP) signaling and thereby decrease hepcidin production. Decreased circulating hepcidin concentrations then allow the release of iron from stores and increase iron absorption from the diet. Guided by evolutionary analysis and Alphafold2 protein complex modeling, we used targeted ERFE mutations, deletions, and synthetic ERFE segments together with cell-based bioassays and surface plasmon resonance to probe the structural features required for bioactivity and BMP binding. We define the ERFE active domain and multiple structural features that act together to entrap BMP ligands. In particular, the hydrophobic helical segment 81 to 86 and specifically the highly conserved tryptophan W82 in the N-terminal region are essential for ERFE bioactivity and Alphafold2 modeling places W82 between two tryptophans in its ligands BMP2, BMP6, and the BMP2/6 heterodimer, an interaction similar to those that bind BMPs to their cognate receptors. Finally, we identify the cationic region 96-107 and the globular TNFα-like domain 186-354 as structural determinants of ERFE multimerization that increase the avidity of ERFE for BMP ligands. Collectively, our results provide further insight into the ERFE-mediated inhibition of BMP signaling in response to erythropoietic stress.
Collapse
Affiliation(s)
- Daniel N Srole
- Department of Medicine, Center for Iron Disorders, David Geffen School of Medicine at UCLA, Los Angeles, California, USA; Molecular and Medical Pharmacology Graduate Program, Graduate Programs in Bioscience, Los Angeles, California, USA
| | - Grace Jung
- Department of Medicine, Center for Iron Disorders, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Alan J Waring
- Department of Medicine, Harbor-UCLA Medical Center, Lundquist Institute, Los Angeles, California, USA
| | - Elizabeta Nemeth
- Department of Medicine, Center for Iron Disorders, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Tomas Ganz
- Department of Medicine, Center for Iron Disorders, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.
| |
Collapse
|
57
|
Zhao M, Wang Y, Yang J, Wang Y, Feng Y, Chen L, Shao Z, Wang H, Xing L. Iron metabolism abnormalities in autoimmune hemolytic anemia and Jianpishengxue keli can ameliorate hemolysis and improve iron metabolism in AIHA mouse models. Ann Med 2023; 55:231-240. [PMID: 36576329 PMCID: PMC9809345 DOI: 10.1080/07853890.2022.2157475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Autoimmune hemolytic anemia (AIHA) is rare heterogeneous disorder characterized by red blood cell (RBC) destruction via auto-antibodies, and after RBC is destroyed, proinflammatory danger-associated molecular patterns including extracellular hemoglobin, heme, and iron which causing cell injury. And oxidative stress represents one of the most significant effects of chronic hemolysis. Jianpishengxue keli can improve the symptoms of anemia patients with kidney disease and tumors and are beneficial in promoting recovery from chronic inflammation. Therefore, it is presumed that Jianpishengxue keli can improve the symptoms of AIHA. We aimed to investigate iron metabolism in AIHA and effects of Jianpishengxue keli on AIHA murine model. METHODS Nineteen hemolytic episode AIHA patients, 10 remission patients and 10 healthy controls (HCs) were enrolled in this study. Serum hepcidin, ferritin and other related indicators of iron metabolism were measured. Mouse models of AIHA were established and received high, medium, or low doses of Jianpishengxue keli by gavage daily for 14 and 28 days respectively. The level of RBCs, Hb, bilirubin, LDH, hepcidin, and the expression level of hepcidin mRNA, and hepatic ferroportin 1(FPN1) protein were evaluated. RESULTS Serum hepcidin in hemolytic episode AIHA patients and remission patients were significantly higher than that in HCs (p = 0.0083 and p = 0.0473, respectively). Serum ferritin in hemolytic AIHA patients was significantly higher than that in HCs (p = 0.008). Serum transferrin saturation levels are increased in patients with AIHA[ (57.21 ± 8.96) %]. EPO in hemolytic group was higher than that in healthy control (p<0.05). In AIHA mouse models, IBIL decreased after 14 days of high dose drug intervention. After 28 days, TBIL and IBIL both significantly decreased in all dose groups and LDH significantly decreased in the medium-and high-dose groups. Body weight improved, and the level of RBCs, Hb and hepcidin in the high-dose group returned to normal. After 14 and 28 days of intervention, hepatic hepcidin mRNA in all dose group significantly decreased. Hepatic FPN1 protein which were significantly lower in the AIHA mouse models, increased in all dose groups after drug intervention for 28 days. CONCLUSION Iron metabolism abnormalities exists in AIHA patients and Jianpishengxue keli can ameliorate hemolysis and improve iron metabolism in AIHA mouse models.KEY MESSAGESIron metabolism abnormalities exists in hemolytic episode AIHA patients. Hepcidin and ferritin levels significantly elevated and also correlated with the severity of AIHA patients. Jianpishengxue keli can ameliorate hemolysis and prompt the recovery of AIHA.
Collapse
Affiliation(s)
- Manjun Zhao
- Department of Hematology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Yan Wang
- Department of Hematology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Jin Yang
- Department of Hematology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Yi Wang
- Third Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yingying Feng
- Department of Hematology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Lei Chen
- Department of Hematology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Zonghong Shao
- Department of Hematology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Huaquan Wang
- Department of Hematology, General Hospital of Tianjin Medical University, Tianjin, China
| | - Limin Xing
- Department of Hematology, General Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
58
|
Ramasamy J, Jagadish C, Sukumaran A, Varghese J, Mani T, Joseph AJ, Simon EG, Jacob M. Low Serum Hepcidin Levels in Patients with Ulcerative Colitis - Implications for Treatment of Co-existent Iron-Deficiency Anemia. Inflammation 2023; 46:2209-2222. [PMID: 37486527 DOI: 10.1007/s10753-023-01872-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 07/02/2023] [Accepted: 07/04/2023] [Indexed: 07/25/2023]
Abstract
Ulcerative colitis (UC) is often associated with anemia. Hepcidin, the central regulator of iron homeostasis, is known to be induced by inflammation and suppressed by anemia. It is not clear how hepcidin is affected in those with UC, when both inflammation and anemia may co-exist.Such knowledge may hold implications for treatment. Hematological and iron-related parameters, C-reactive protein (CRP), growth differentiation factor 15 (GDF-15) and erythroferrone (ERFE) (erythroid regulators of hepcidin) levels were estimated in blood from those with UC and in control subjects. Values for hematological and iron-related parameters showed evidence of iron-deficiency and resultant anemia, in patients with UC. The presence of UC was significantly associated with inflammation. Serum levels of ERFE, but not of GDF-15, were significantly higher in patients with UC than in control patients, while hepcidin levels were significantly lower. Serum hepcidin concentrations in patients with UC correlated positively with serum iron, ferritin and GDF-15, and negatively with serum ERFE. The iron status and serum hepcidin levels in UC patients with co-existent anemia were significantly lower and serum ERFE values significantly higher than in those with UC without anemia. The effect of anemia on hepcidin predominated over that of inflammation in patients with UC, resulting in suppressed hepcidin levels. This effect is possibly mediated through erythroferrone. We suggest that a serum hepcidin-guided approach may be useful to guide use of oral iron supplements to treat co-existent iron-deficiency anemia in patients with UC and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Jagadish Ramasamy
- Department of Biochemistry, Christian Medical College, Vellore, Tamil Nadu, 632002, India
- Department of Biochemistry, Velammal Medical College Hospital and Research Institute, Madurai, Tamil Nadu, 625009, India
| | - Chinmai Jagadish
- Department of Biochemistry, Christian Medical College, Vellore, Tamil Nadu, 632002, India
- Department of Biochemistry, Dr. Chandramma Dayanada Sagar Institute of Medical Education and Research, Deverakaggalahalli, Karnataka, 562112, India
| | - Abitha Sukumaran
- Department of Biochemistry, Christian Medical College, Vellore, Tamil Nadu, 632002, India
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Joe Varghese
- Department of Biochemistry, Christian Medical College, Vellore, Tamil Nadu, 632002, India
| | - Thenmozhi Mani
- Department of Biostatistics, Christian Medical College, Vellore, Tamil Nadu, 632002, India
| | - A J Joseph
- Department of Gastroenterology, Christian Medical College and Hospital, Vellore, Tamil Nadu, 632002, India
| | - Ebby George Simon
- Department of Gastroenterology, Christian Medical College and Hospital, Vellore, Tamil Nadu, 632002, India
| | - Molly Jacob
- Department of Biochemistry, Christian Medical College, Vellore, Tamil Nadu, 632002, India.
| |
Collapse
|
59
|
Ruivard M, Lobbes H. [Diagnosis and treatment of iron overload]. Rev Med Interne 2023; 44:656-661. [PMID: 37507250 DOI: 10.1016/j.revmed.2023.07.002] [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: 06/07/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023]
Abstract
Etiological investigation of hyperferritinemia includes a full clinical examination, with the measurement of waist circumference, and simple biological tests including transferrin saturation. The classification between hyperferritinemia without iron overload (inflammation, excessive alcohol intake, cytolysis, L-ferritin mutation) or with iron overload is then relatively easy. Dysmetabolic iron overload syndrome is the most common iron overload disease and is defined by an unexplained serum ferritin level elevation associated with various metabolic syndrome criteria and mild hepatic iron content increase assessed by magnetic resonance imaging. Bloodlettings are often poorly tolerated without clear benefit. Type 1 genetic hemochromatosis (homozygous C282Y mutation on the HFE gene) leads to iron accumulation through an increase of dietary iron absorption due to hypohepcidinemia. More than 95% of hemochromatosis are type 1 hemochromatosis but the phenotypic expression is highly variable. Elastography is recommended to identify advanced hepatic fibrosis when serum ferritin exceeds 1000μg/L. Life expectancy is normal when bloodlettings are started early. Ferroportin gene mutation is an autosomal dominant disease with generally moderate iron overload. Chelators are used in iron overload associated with anaemia (myelodysplastic syndromes or transfusion-dependent thalassemia). Chelation is initiated when hepatic iron content exceeds 120μmol/g. Deferasirox is often used as first-line therapy, but deferiprone may be of interest despite haematological toxicity (neutropenia). Deferoxamine (parenteral route) is the treatment of choice for severe iron overload or emergency conditions.
Collapse
Affiliation(s)
- M Ruivard
- Service médecine Interne, CHU de Clermont-Ferrand, CHU d'Estaing, 63003 Clermont-Ferrand, France; UMR 6602 UCA/CNRS/SIGMA, thérapies guidées par l'image (TGI), université Clermont Auvergne, 63000 Clermont-Ferrand, France.
| | - H Lobbes
- Service médecine Interne, CHU de Clermont-Ferrand, CHU d'Estaing, 63003 Clermont-Ferrand, France; UMR 6602 UCA/CNRS/SIGMA, thérapies guidées par l'image (TGI), université Clermont Auvergne, 63000 Clermont-Ferrand, France.
| |
Collapse
|
60
|
Wang X, Wei C, Zhao D, Sun X, Zhu F, Mei Y, Ma Q, Cai G, Chen X, Li P. Iron Supplements Concomitant within Hypoxia-Inducible Factor Prolyl Hydroxylase Domain Inhibitors in the Treatment of Chronic Kidney Disease Anemia. KIDNEY DISEASES (BASEL, SWITZERLAND) 2023; 9:485-497. [PMID: 38098876 PMCID: PMC10719729 DOI: 10.1159/000533304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 07/20/2023] [Indexed: 12/17/2023]
Abstract
Background Anemia is a common and important complication in patients with chronic kidney disease (CKD). Accordingly, the current treatment is based on erythropoiesis-stimulating agents (ESAs) and iron. Hypoxia-inducible factor (HIF) prolyl hydroxylase domain inhibitors (HIF-PHIs) have been developed to treat renal anemia through a novel mechanism. HIF-PHIs increase erythropoietin at physiologic blood concentrations and also improve the supply of hematopoietic iron. Iron is the main component of hemoglobin, and ensuring efficient iron metabolism is essential in the treatment of anemia. Summary HIF-PHIs may have advantages in improving iron utilization and mobilization compared to ESAs. Most HIF-PHI trials revealed a significant decline of hepcidin, increase in transferrin level and total iron binding capacity in patients. From a clinical point of view, improvements in iron metabolism should translate into reductions in iron supplementation. There are differences in the iron treatment regimentation currently used, so it is important to evaluate and timely iron supplementation across studies. Key Messages This review summarizes the mechanism of HIF-PHIs on improved iron metabolism and the route of iron usage in the trials for dialysis-dependent CKD and non-dialysis CKD. And this review also makes an interpretation of the clinical practice guidelines in China and recommendation by Asia Pacific Society of Nephrology.
Collapse
Affiliation(s)
- Xue Wang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Medical School, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Cuiting Wei
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Medical School, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Delong Zhao
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Medical School, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Xuefeng Sun
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Medical School, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Fengge Zhu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Medical School, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Yan Mei
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Medical School, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Qian Ma
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Medical School, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Guangyan Cai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Medical School, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Medical School, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Ping Li
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Medical School, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| |
Collapse
|
61
|
Zhao Y, Xiong W, Li C, Zhao R, Lu H, Song S, Zhou Y, Hu Y, Shi B, Ge J. Hypoxia-induced signaling in the cardiovascular system: pathogenesis and therapeutic targets. Signal Transduct Target Ther 2023; 8:431. [PMID: 37981648 PMCID: PMC10658171 DOI: 10.1038/s41392-023-01652-9] [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: 07/10/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 11/21/2023] Open
Abstract
Hypoxia, characterized by reduced oxygen concentration, is a significant stressor that affects the survival of aerobic species and plays a prominent role in cardiovascular diseases. From the research history and milestone events related to hypoxia in cardiovascular development and diseases, The "hypoxia-inducible factors (HIFs) switch" can be observed from both temporal and spatial perspectives, encompassing the occurrence and progression of hypoxia (gradual decline in oxygen concentration), the acute and chronic manifestations of hypoxia, and the geographical characteristics of hypoxia (natural selection at high altitudes). Furthermore, hypoxia signaling pathways are associated with natural rhythms, such as diurnal and hibernation processes. In addition to innate factors and natural selection, it has been found that epigenetics, as a postnatal factor, profoundly influences the hypoxic response and progression within the cardiovascular system. Within this intricate process, interactions between different tissues and organs within the cardiovascular system and other systems in the context of hypoxia signaling pathways have been established. Thus, it is the time to summarize and to construct a multi-level regulatory framework of hypoxia signaling and mechanisms in cardiovascular diseases for developing more therapeutic targets and make reasonable advancements in clinical research, including FDA-approved drugs and ongoing clinical trials, to guide future clinical practice in the field of hypoxia signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Yongchao Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Weidong Xiong
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China
| | - Chaofu Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Shuai Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - You Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Yiqing Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| | - Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| | - Junbo Ge
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
62
|
Nai A. BMP5: a novel tile of the hepcidin regulatory pathway. Blood 2023; 142:1260-1261. [PMID: 37824161 DOI: 10.1182/blood.2023021643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023] Open
Affiliation(s)
- Antonella Nai
- IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University
| |
Collapse
|
63
|
Xiao X, Xu Y, Moschetta GA, Yu Y, Fisher AL, Alfaro-Magallanes VM, McMillen S, Phillips S, Wang CY, Christian J, Babitt JL. BMP5 contributes to hepcidin regulation and systemic iron homeostasis in mice. Blood 2023; 142:1312-1322. [PMID: 37478395 PMCID: PMC10613724 DOI: 10.1182/blood.2022019195] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 06/14/2023] [Accepted: 06/22/2023] [Indexed: 07/23/2023] Open
Abstract
Hepcidin is the master regulator of systemic iron homeostasis. The bone morphogenetic protein (BMP) signaling pathway is a critical regulator of hepcidin expression in response to iron and erythropoietic drive. Although endothelial-derived BMP6 and BMP2 ligands have key functional roles as endogenous hepcidin regulators, both iron and erythropoietic drives still regulate hepcidin in mice lacking either or both ligands. Here, we used mice with an inactivating Bmp5 mutation (Bmp5se), either alone or together with a global or endothelial Bmp6 knockout, to investigate the functional role of BMP5 in hepcidin and systemic iron homeostasis regulation. We showed that Bmp5se-mutant mice exhibit hepcidin deficiency at age 10 days, blunted hepcidin induction in response to oral iron gavage, and mild liver iron loading when fed on a low- or high-iron diet. Loss of 1 or 2 functional Bmp5 alleles also leads to increased iron loading in Bmp6-heterozygous mice and more profound hemochromatosis in global or endothelial Bmp6-knockout mice. Moreover, double Bmp5- and Bmp6-mutant mice fail to induce hepcidin in response to long-term dietary iron loading. Finally, erythroferrone binds directly to BMP5 and inhibits BMP5 induction of hepcidin in vitro. Although erythropoietin suppresses hepcidin in Bmp5se-mutant mice, it fails to suppress hepcidin in double Bmp5- and Bmp6-mutant males. Together, these data demonstrate that BMP5 plays a functional role in hepcidin and iron homeostasis regulation, particularly under conditions in which BMP6 is limited.
Collapse
Affiliation(s)
- Xia Xiao
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Yang Xu
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Gillian A. Moschetta
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Yang Yu
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Allison L. Fisher
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Víctor M. Alfaro-Magallanes
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- LFE Research Group, Department of Health and Human Performance, Faculty of Physical Activity and Sport Sciences, Universidad Politécnica de Madrid, Madrid, Spain
| | - Shasta McMillen
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Sydney Phillips
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Chia-Yu Wang
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Jan Christian
- Division of Hematology and Hematologic Malignancies, Department of Neurobiology and Internal Medicine, University of Utah, Salt Lake City, UT
| | - Jodie L. Babitt
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
64
|
Wang XM, Wang QY, Huang J. Anemia status of infants and young children aged six to thirty-six months in Ma'anshan City: A retrospective study. World J Clin Cases 2023; 11:6744-6753. [PMID: 37901006 PMCID: PMC10600847 DOI: 10.12998/wjcc.v11.i28.6744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 09/25/2023] Open
Abstract
BACKGROUND Anemia in infants and young children can have long-term effects on cognitive and physical development. In Ma'anshan City, China, there has been growing concern about the prevalence of anemia among children aged 6 to 36 mo. Understanding the factors influencing this condition is crucial for targeted interventions and improving overall child health in the region. AIM To analyze the anemia status and influencing factors of infants and young children aged 6 to 36 mo in Ma'anshan City, China. Providing scientific evidence for reducing the incidence of anemia and improving the health level of children in this age group. METHODS The study encompassed 37698 infants and young children, aged from 6 to 36 mo, who underwent health examinations at the Ma'anshan Maternal and Child Health Hospital from January 2018 to October 2022 were included in the study. Basic information, physical examination, and hemoglobin detection data were collected. Descriptive analysis was used to analyze the prevalence of anemia in children in the region, and univariate analysis was used to analyze the influencing factors of anemia. RESULTS The mean hemoglobin level of infants and young children aged 9 to 36 mo increased with age, and the anemia detection rate decreased with age. The anemia detection rate in rural infants aged 6, 9, and 12 mo was higher than that in urban infants. Although the anemia detection rate was higher in 6-mo-old boys than girls, it was higher in 24-mo-old girls than boys. There were statistically significant differences in the anemia detection rates among 9-mo-old and 12-mo-old infants with different nutritional statuses (emaciation, overweight, obese, and normal). Moreover, there were no statistically significant differences in anemia detection rates among infants and young children with different nutritional statuses at other ages. Besides, the anemia detection rates in obese infants aged 9 and 12 mo were higher than those in normal and overweight infants, with statistically significant differences. Finally, there were no statistically significant differences in the anemia detection rates between emaciation infants and those with other nutritional statuses. CONCLUSION The anemia situation among infants and young children aged 6 to 36 mo in Ma'anshan City, China, is relatively prominent and influenced by various factors. Our result shown that attention should be paid to the anemic infant and young child population, with strengthened education and targeted prevention and dietary guidance to help them establish good living habits, improve nutritional status, and reduce the occurrence of anemia to improve children's health levels.
Collapse
Affiliation(s)
- Xue-Mei Wang
- Department of Pediatric, Ma’anshan Maternal and Child Health Center, Ma’anshan 243011, Anhui Province, China
| | - Qiong-Yao Wang
- Department of Pediatric, Ma’anshan Maternal and Child Health Center, Ma’anshan 243011, Anhui Province, China
| | - Jie Huang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu Province, China
| |
Collapse
|
65
|
Zhou M, Tamburini IJ, Van C, Molendijk J, Nguyen CM, Chang IYY, Johnson C, Velez LM, Cheon Y, Yeo RX, Bae H, Le J, Larson N, Pulido R, Filho C, Jang C, Marazzi I, Justice JN, Pannunzio N, Hevener A, Sparks LM, Kershaw EE, Nicholas D, Parker B, Masri S, Seldin M. Leveraging inter-individual transcriptional correlation structure to infer discrete signaling mechanisms across metabolic tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.10.540142. [PMID: 37214953 PMCID: PMC10197628 DOI: 10.1101/2023.05.10.540142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Abstract/IntroductionInter-organ communication is a vital process to maintain physiologic homeostasis, and its dysregulation contributes to many human diseases. Beginning with the discovery of insulin over a century ago, characterization of molecules responsible for signal between tissues has required careful and elegant experimentation where these observations have been integral to deciphering physiology and disease. Given that circulating bioactive factors are stable in serum, occur naturally, and are easily assayed from blood, they present obvious focal molecules for therapeutic intervention and biomarker development. For example, physiologic dissection of the actions of soluble proteins such as proprotein convertase subtilisin/kexin type 9 (PCSK9) and glucagon-like peptide 1 (GLP1) have yielded among the most promising therapeutics to treat cardiovascular disease and obesity, respectively1–4. A major obstacle in the characterization of such soluble factors is that defining their tissues and pathways of action requires extensive experimental testing in cells and animal models. Recently, studies have shown that secreted proteins mediating inter-tissue signaling could be identified by “brute-force” surveys of all genes within RNA-sequencing measures across tissues within a population5–9. Expanding on this intuition, we reasoned that parallel strategies could be used to understand how individual genes mediate signaling across metabolic tissues through correlative analyses of gene variation between individuals. Thus, comparison of quantitative levels of gene expression relationships between organs in a population could aid in understanding cross-organ signaling. Here, we surveyed gene-gene correlation structure across 18 metabolic tissues in 310 human individuals and 7 tissues in 103 diverse strains of mice fed a normal chow or HFHS diet. Variation of genes such asFGF21, ADIPOQ, GCGandIL6showed enrichments which recapitulate experimental observations. Further, similar analyses were applied to explore both within-tissue signaling mechanisms (liverPCSK9) as well as genes encoding enzymes producing metabolites (adiposePNPLA2), where inter-individual correlation structure aligned with known roles for these critical metabolic pathways. Examination of sex hormone receptor correlations in mice highlighted the difference of tissue-specific variation in relationships with metabolic traits. We refer to this resource asGene-DerivedCorrelationsAcrossTissues (GD-CAT) where all tools and data are built into a web portal enabling users to perform these analyses without a single line of code (gdcat.org). This resource enables querying of any gene in any tissue to find correlated patterns of genes, cell types, pathways and network architectures across metabolic organs.
Collapse
Affiliation(s)
- Mingqi Zhou
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Ian J. Tamburini
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Cassandra Van
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Jeffrey Molendijk
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Christy M Nguyen
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | | | - Casey Johnson
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Leandro M. Velez
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Youngseo Cheon
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Reichelle X. Yeo
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Hosung Bae
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Johnny Le
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Natalie Larson
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Ron Pulido
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Carlos Filho
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Ivan Marazzi
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Jamie N. Justice
- Veterans Administration Greater Los Angeles Healthcare System, Geriatric Research Education and Clinical Center (GRECC), Los Angeles, CA, USA
| | - Nicholas Pannunzio
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Andrea Hevener
- Department of Medicine, Division of Endocrinology, Diabetes, and Hypertension, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Iris Cantor-UCLA Women’s Health Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Lauren M. Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Erin E. Kershaw
- Department of Internal Medicine, Section On Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Dequina Nicholas
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Benjamin Parker
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Selma Masri
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
- Center for Epigenetics and Metabolism, UC Irvine. Irvine, CA, USA
| | - Marcus Seldin
- Department of Biological Chemistry, UC Irvine. Irvine, CA, USA
| |
Collapse
|
66
|
Guerra A, Parhiz H, Rivella S. Novel potential therapeutics to modify iron metabolism and red cell synthesis in diseases associated with defective erythropoiesis. Haematologica 2023; 108:2582-2593. [PMID: 37345473 PMCID: PMC10542825 DOI: 10.3324/haematol.2023.283057] [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: 04/01/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023] Open
Abstract
Under normal conditions, iron metabolism is carefully regulated to sustain normal cellular functions and the production of hemoglobin in erythroid cells. Perturbation to the erythropoiesis-iron metabolism axis can result in iron imbalances and cause anemia or organ toxicity. Various congenital and acquired diseases associated with abnormal red cell production are characterized by aberrant iron absorption. Several recent studies have shown that improvements in red blood cell production also ameliorate iron metabolism and vice versa. Many therapeutics are now under development with the potential to improve a variety of hematologic diseases, from β-thalassemia and iron-refractory iron deficiency anemia to anemia of inflammation and polycythemia vera. This review summarizes selected mechanisms related to red cell production and iron metabolism and describes potential therapeutics and their current uses. We also consider the potential application of the discussed therapeutics on various diseases, alone or in combination. The vast repertoire of drugs under development offers new opportunities to improve the clinical care of patients suffering from congenital or acquired red blood cell disorders with limited or no treatment options.
Collapse
Affiliation(s)
- Amaliris Guerra
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA
| | - Hamideh Parhiz
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA; RNA Institute, University of Pennsylvania, Philadelphia, PA
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA; Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA; RNA Institute, University of Pennsylvania, 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.
| |
Collapse
|
67
|
Schmitz L, Pirotte M, Lebeau A, Ernst M, Fillet M, Devey A, Schmitt J, Cobraiville G, Binsfeld M, Gofflot S, Beguin Y, Vertenoeil G. Alterations of erythropoiesis in Covid-19 patients: prevalence of positive Coombs tests and iron metabolism. Ther Adv Hematol 2023; 14:20406207231199837. [PMID: 37780945 PMCID: PMC10540584 DOI: 10.1177/20406207231199837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 08/18/2023] [Indexed: 10/03/2023] Open
Abstract
Background For more than 2 years medical practice has been dealing with the Covid-19 pandemic. Atypical symptoms, such as frostbites and acrosyndromes, have appeared, and autoimmune anemias (some of which with cold agglutinins) have been described. Objectives We planned to study the prevalence of positive direct Coombs tests (DCTs) and hemolytic autoimmune anemia in patients infected with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and its correlation with complications, and then investigate the impact of the infection on iron metabolism. Design This is an observational, cross-sectional, single-center, exploratory study. Methods We obtained Coombs tests in a population of 179 infected patients at the CHU of Liège. We then studied iron metabolism in some of these patients, by measuring serum ferritin, erythropoietin (EPO), erythroferrone and hepcidin. Results We did not identify any case of autoimmune hemolysis. However, there was a 20.3% prevalence of positive DCT, mainly with IgG (91.7%). These patients, compared to DCT-negative patients, were not only more anemic and transfused, but also required more transfers to intensive care units and had longer hospital stays and mechanical ventilation. The pattern of anemia was consistent with the anemia of inflammation, showing elevated hepcidin and ferritin levels, while EPO and erythroferrone values were lower than expected at this degree of anemia. Erythroferrone was higher and Hb was lower in DCT-positive patients. Finally, we identified a correlation between iron parameters and complicated forms of infection. Conclusion Covid-19 patients suffered from inflammatory anemia with more severe forms of infection correlated to positive DCT status. This could potentially be of interest for future clinical practice.
Collapse
Affiliation(s)
- Léa Schmitz
- Department of Hematology and GIGA Laboratory of Hematology, University Hospital of Liège and ULiège, Liège, Belgium
| | - Michelle Pirotte
- Department of Hematology and GIGA Laboratory of Hematology, University Hospital of Liège and ULiège, Liège, Belgium
| | - Alizée Lebeau
- Laboratory of Experimental Pathology, GIGA Cancer, University Hospital of Liège and ULiège, Liège, Belgium
| | - Marie Ernst
- Department of Biostatistics and Medico-Economics, University Hospital of Liège and ULiège, Liège, Belgium
| | - Marianne Fillet
- Laboratory for the Analysis of Medicines, CIRM, ULiège, Liège, Belgium
| | - Anais Devey
- Department of Clinical Biology, University Hospital of Liège and ULiège, Liège, Belgium
| | - Justine Schmitt
- Department of Clinical Biology, University Hospital of Liège and ULiège, Liège, Belgium
| | - Gaël Cobraiville
- Department of Rhumatology and GIGA Laboratory of Rhumatology, University Hospital of Liège and ULiège, Liège, Belgium
| | - Marilène Binsfeld
- Hematology Research Unit, GIGA-I3, University of Liège, Liège, Belgium
| | - Stéphanie Gofflot
- Biobank of the CHU of Liège, CHU de Liège – Hôpital du Sart Tilman, Liège, Belgium
| | - Yves Beguin
- Department of Hematology and GIGA Laboratory of Hematology, University Hospital of Liège and ULiège, Liège, Belgium
| | - Gaëlle Vertenoeil
- Department of Hematology, CHU of Liège, 1 avenue de l’Hôpital, Liège 4000, Belgium
| |
Collapse
|
68
|
Helman SL, Wilkins SJ, Chan JCJ, Hartel G, Wallace DF, Anderson GJ, Frazer DM. A Decrease in Maternal Iron Levels Is the Predominant Factor Suppressing Hepcidin during Pregnancy in Mice. Int J Mol Sci 2023; 24:14379. [PMID: 37762679 PMCID: PMC10532249 DOI: 10.3390/ijms241814379] [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: 08/16/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
In order to supply adequate iron during pregnancy, the levels of the iron regulatory hormone hepcidin in the maternal circulation are suppressed, thereby increasing dietary iron absorption and storage iron release. Whether this decrease in maternal hepcidin is caused by changes in factors known to regulate hepcidin expression, or by other unidentified pregnancy factors, is not known. To investigate this, we examined iron parameters during pregnancy in mice. We observed that hepatic iron stores and transferrin saturation, both established regulators of hepcidin production, were decreased in mid and late pregnancy in normal and iron loaded dams, indicating an increase in iron utilization. This can be explained by a significant increase in maternal erythropoiesis, a known suppressor of hepcidin production, by mid-pregnancy, as indicated by an elevation in circulating erythropoietin and an increase in spleen size and splenic iron uptake. Iron utilization increased further in late pregnancy due to elevated fetal iron demand. By increasing maternal iron levels in late gestation, we were able to stimulate the expression of the gene encoding hepcidin, suggesting that the iron status of the mother is the predominant factor influencing hepcidin levels during pregnancy. Our data indicate that pregnancy-induced hepcidin suppression likely occurs because of reductions in maternal iron reserves due to increased iron requirements, which predominantly reflect stimulated erythropoiesis in mid-gestation and increased fetal iron requirements in late gestation, and that there is no need to invoke other factors, including novel pregnancy factor(s), to explain these changes.
Collapse
Affiliation(s)
- Sheridan L. Helman
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (S.L.H.); (J.C.J.C.)
| | - Sarah J. Wilkins
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia (G.J.A.)
| | - Jennifer C. J. Chan
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (S.L.H.); (J.C.J.C.)
| | - Gunter Hartel
- Statistics Unit, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia;
- School of Public Health, The University of Queensland, Herston, QLD 4006, Australia
- School of Nursing, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| | - Daniel F. Wallace
- School of Biomedical Sciences and Centre for Genomics and Personalised Health, Queensland University of Technology, Kelvin Grove, Brisbane, QLD 4059, Australia;
| | - Gregory J. Anderson
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia (G.J.A.)
| | - David M. Frazer
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (S.L.H.); (J.C.J.C.)
- School of Biomedical Sciences, Queensland University of Technology, Gardens Point, QLD 4000, Australia
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4067, Australia
| |
Collapse
|
69
|
Saliba AN, Musallam KM, Taher AT. How I treat non-transfusion-dependent β-thalassemia. Blood 2023; 142:949-960. [PMID: 37478396 PMCID: PMC10644094 DOI: 10.1182/blood.2023020683] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/23/2023] Open
Abstract
The intricate interplay of anemia and iron overload under the pathophysiological umbrella of ineffective erythropoiesis in non-transfusion-dependent β-thalassemia (NTDT) results in a complex variety of clinical phenotypes that are challenging to diagnose and manage. In this article, we use a clinical framework rooted in pathophysiology to present 4 common scenarios of patients with NTDT. Starting from practical considerations in the diagnosis of NTDT, we delineate our strategy for the longitudinal care of patients who exhibit different constellations of symptoms and complications. We highlight the use of transfusion therapy and novel agents, such as luspatercept, in the patient with anemia-related complications. We also describe our approach to chelation therapy in the patient with iron overload. Although tackling every specific complication of NTDT is beyond the scope of this article, we touch on the management of the various morbidities and multisystem manifestations of the disease.
Collapse
Affiliation(s)
| | - Khaled M. Musallam
- Thalassemia Center, Burjeel Medical City, Abu Dhabi, United Arab Emirates
| | - Ali T. Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
70
|
Woźniak M, Borkowska A, Jastrzębska M, Sochal M, Małecka-Wojciesko E, Talar-Wojnarowska R. Clinical Significance of Erythroferrone and Bone Morphogenetic Protein-6 in Patients with Anemia in the Course of Inflammatory Bowel Disease. Metabolites 2023; 13:1006. [PMID: 37755286 PMCID: PMC10537870 DOI: 10.3390/metabo13091006] [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: 06/26/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/28/2023] Open
Abstract
In recent years, a steady increase in the incidence of inflammatory bowel diseases (IBD) has been observed with anemia as their most common extraintestinal symptom. Erythroferrone and Bone Morphogenetic Protein 6 (BMP-6) are recently identified cytokines involved in the process of increased erythropoiesis in anemia of various pathomechanisms. The aim of this study was to analyze the concentration of erythroferrone and BMP-6 in IBD patients in relation to clinical and laboratory data. The study comprised 148 patients: 118 with IBD, including 73 (61.85%) diagnosed with anemia (42 with Crohn's disease (CD) (66.7%) and 31 (56.4%) with ulcerative colitis (UC)) and 30 as a control group. The erythroferrone concentration was significantly higher in IBD patients with anemia (p = 0.009) and higher in UC patients both with and without anemia (p = 0.018), compared to the control group. In CD, no similar difference was observed between patients with and without anemia. Regarding BMP-6, higher levels were found in CD patients with anemia compared to the control group (p = 0.021). The positive correlation between BMP-6 and iron concentration in UC was also noticed. In conclusion, we confirm an increase in erythroferrone concentration in the entire group of IBD patients with anemia, while BMP-6 levels were higher only in anemic CD patients. Due to the clinical importance of anemia in IBD, this problem is worth further analysis and research projects.
Collapse
Affiliation(s)
- Małgorzata Woźniak
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-419 Lodz, Poland; (A.B.); (E.M.-W.); (R.T.-W.)
| | - Anna Borkowska
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-419 Lodz, Poland; (A.B.); (E.M.-W.); (R.T.-W.)
| | - Marta Jastrzębska
- Department of Gastroenterology, Health Care Center, 26-200 Konskie, Poland;
| | - Marcin Sochal
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 90-419 Lodz, Poland;
| | - Ewa Małecka-Wojciesko
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-419 Lodz, Poland; (A.B.); (E.M.-W.); (R.T.-W.)
| | - Renata Talar-Wojnarowska
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-419 Lodz, Poland; (A.B.); (E.M.-W.); (R.T.-W.)
| |
Collapse
|
71
|
Li X, Lozovatsky L, Tommasini SM, Fretz J, Finberg KE. Bone marrow sinusoidal endothelial cells are a site of Fgf23 upregulation in a mouse model of iron deficiency anemia. Blood Adv 2023; 7:5156-5171. [PMID: 37417950 PMCID: PMC10480544 DOI: 10.1182/bloodadvances.2022009524] [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: 12/12/2022] [Revised: 06/16/2023] [Accepted: 07/05/2023] [Indexed: 07/08/2023] Open
Abstract
Iron deficiency is a potent stimulator of fibroblast growth factor 23 (FGF23), a hormonal regulator of phosphate and vitamin D metabolism, that is classically thought to be produced by bone-embedded osteocytes. Here, we show that iron-deficient transmembrane serine protease 6 knockout (Tmprss6-/-) mice exhibit elevated circulating FGF23 and Fgf23 messenger RNA (mRNA) upregulation in the bone marrow (BM) but not the cortical bone. To clarify sites of Fgf23 promoter activity in Tmprss6-/- mice, we introduced a heterozygous enhanced green fluorescent protein (eGFP) reporter allele at the endogenous Fgf23 locus. Heterozygous Fgf23 disruption did not alter the severity of systemic iron deficiency or anemia in the Tmprss6-/- mice. Tmprss6-/-Fgf23+/eGFP mice showed green fluorescence in the vascular regions of BM sections and showed a subset of BM endothelial cells that were GFPbright by flow cytometry. Mining of transcriptomic data sets from mice with normal iron balance revealed higher Fgf23 mRNA in BM sinusoidal endothelial cells (BM-SECs) than that in other BM endothelial cell populations. Anti-GFP immunohistochemistry of fixed BM sections from Tmprss6-/-Fgf23+/eGFP mice revealed GFP expression in BM-SECs, which was more intense than in nonanemic controls. In addition, in mice with intact Tmprss6 alleles, Fgf23-eGFP reporter expression increased in BM-SECs following large-volume phlebotomy and also following erythropoietin treatment both ex vivo and in vivo. Collectively, our results identified BM-SECs as a novel site for Fgf23 upregulation in both acute and chronic anemia. Given the elevated serum erythropoietin in both anemic models, our findings raise the possibility that erythropoietin may act directly on BM-SECs to promote FGF23 production during anemia.
Collapse
Affiliation(s)
- Xiuqi Li
- Department of Pathology, Yale School of Medicine, New Haven, CT
| | | | - Steven M. Tommasini
- Department of Orthopaedics & Rehabilitation, Yale School of Medicine, New Haven, CT
| | - Jackie Fretz
- Department of Orthopaedics & Rehabilitation, Yale School of Medicine, New Haven, CT
| | | |
Collapse
|
72
|
Tomosugi N, Koshino Y, Ogawa C, Maeda K, Shimada N, Tomita K, Daimon S, Shikano T, Ryu K, Takatani T, Sakamoto K, Ueyama S, Nagasaku D, Nakamura M, Ra S, Nishimura M, Takagi C, Ishii Y, Kudo N, Takechi S, Ishizu T, Yanagawa T, Fukuda M, Nitta Y, Yamaoka T, Saito T, Imayoshi S, Omata M, Oshima J, Onozaki A, Ichihashi H, Matsushima Y, Takae H, Nakazawa R, Ikeda K, Tsuboi M, Konishi K, Kato S, Ooura M, Koyama M, Naganuma T, Ogi M, Katayama S, Okumura T, Kameda S, Shirai S. Oral Iron Absorption of Ferric Citrate Hydrate and Hepcidin-25 in Hemodialysis Patients: A Prospective, Multicenter, Observational Riona-Oral Iron Absorption Trial. Int J Mol Sci 2023; 24:13779. [PMID: 37762085 PMCID: PMC10531220 DOI: 10.3390/ijms241813779] [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: 08/15/2023] [Revised: 08/27/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Oral ferric citrate hydrate (FCH) is effective for iron deficiencies in hemodialysis patients; however, how iron balance in the body affects iron absorption in the intestinal tract remains unclear. This prospective observational study (Riona-Oral Iron Absorption Trial, R-OIAT, UMIN 000031406) was conducted at 42 hemodialysis centers in Japan, wherein 268 hemodialysis patients without inflammation were enrolled and treated with a fixed amount of FCH for 6 months. We assessed the predictive value of hepcidin-25 for iron absorption and iron shift between ferritin (FTN) and red blood cells (RBCs) following FCH therapy. Serum iron changes at 2 h (ΔFe2h) after FCH ingestion were evaluated as iron absorption. The primary outcome was the quantitative delineation of iron variables with respect to ΔFe2h, and the secondary outcome was the description of the predictors of the body's iron balance. Generalized estimating equations (GEEs) were used to identify the determinants of iron absorption during each phase of FCH treatment. ΔFe2h increased when hepcidin-25 and TSAT decreased (-0.459, -0.643 to -0.276, p = 0.000; -0.648, -1.099 to -0.197, p = 0.005, respectively) in GEEs. FTN increased when RBCs decreased (-1.392, -1.749 to -1.035, p = 0.000) and hepcidin-25 increased (0.297, 0.239 to 0.355, p = 0.000). Limiting erythropoiesis to maintain hemoglobin levels induces RBC reduction in hemodialysis patients, resulting in increased hepcidin-25 and FTN levels. Hepcidin-25 production may prompt an iron shift from RBC iron to FTN iron, inhibiting iron absorption even with continued FCH intake.
Collapse
Affiliation(s)
- Naohisa Tomosugi
- Division of Systems Bioscience for Drug Discovery, Project Research Center, Medical Research Institute, Kanazawa Medical University, Kahoku 920-0293, Ishikawa, Japan
| | | | - Chie Ogawa
- Maeda Institute of Renal Research Musashikosugi, Kawasaki 211-0063, Kanagawa, Japan;
| | - Kunimi Maeda
- Maeda Institute of Renal Research Shakujii, Nerima 177-0041, Tokyo, Japan;
| | | | - Kimio Tomita
- The Chronic Kidney Disease Research Center, Tomei Atsugi General Hospital, Atsugi 243-8571, Kanagawa, Japan;
| | - Shoichiro Daimon
- Department of Nephrology, Daimon Clinic for Internal Medicine, Nonoichi 921-8802, Ishikawa, Japan;
| | - Tsutomu Shikano
- Kyoto Okamoto Memorial Hospital, Kuze 613-0034, Kyoto, Japan; (T.S.); (K.R.)
| | - Kazuyuki Ryu
- Kyoto Okamoto Memorial Hospital, Kuze 613-0034, Kyoto, Japan; (T.S.); (K.R.)
| | - Toru Takatani
- Nephrology Division, Tojinkai Hospital, Fushimi 612-8026, Kyoto, Japan;
| | - Kazuya Sakamoto
- Department of Urology, Tomakomai Nisshou Hospital, Tomakomai 053-0803, Hokkaido, Japan;
| | - Satonori Ueyama
- Jinaikai Ueyama Hospital, Kagoshima 890-0073, Kagoshima, Japan;
| | | | | | - Shibun Ra
- Noheji Clinic, Noheji 039-3152, Aomori, Japan;
| | | | | | - Yoji Ishii
- Nozatomon Clinic, Himeji 670-0011, Hyogo, Japan;
| | | | | | - Takashi Ishizu
- Department of Nephrology, Tsukuba Central Hospital, Ushiku 300-1211, Ibaraki, Japan; (T.I.); (T.Y.)
| | - Takamoto Yanagawa
- Department of Nephrology, Tsukuba Central Hospital, Ushiku 300-1211, Ibaraki, Japan; (T.I.); (T.Y.)
| | | | - Yutaka Nitta
- The Department of Nephrology, Saiseikai Shimonoseki General Hospital, Shimonoseki 759-6603, Yamaguchi, Japan; (Y.N.); (T.Y.)
| | - Takayuki Yamaoka
- The Department of Nephrology, Saiseikai Shimonoseki General Hospital, Shimonoseki 759-6603, Yamaguchi, Japan; (Y.N.); (T.Y.)
| | - Taku Saito
- Saito Memorial Hospital, Kawaguchi 332-0034, Saitama, Japan; (T.S.); (S.I.)
| | - Suzuko Imayoshi
- Saito Memorial Hospital, Kawaguchi 332-0034, Saitama, Japan; (T.S.); (S.I.)
| | - Momoyo Omata
- Department of Internal Medicine, Hachioji Azumacho Clinic, Hachioji-shi 192-0082, Tokyo, Japan;
| | - Joji Oshima
- Kubojima Clinic, Kumagaya 360-0831, Saitama, Japan;
| | - Akira Onozaki
- Tokatsu-Clinic Hospital, Matsudo 271-0067, Chiba, Japan;
| | | | | | | | | | - Koichi Ikeda
- Tokatsu Clinic Koiwa, Edogawa 133-0056, Tokyo, Japan;
| | - Masato Tsuboi
- Kaikoukai Anjo Kyoritsu Clinic, Anjo 446-0065, Aichi, Japan;
| | | | - Shouzaburo Kato
- Nishi Interchange Clinic for Internal Medicine and Dialysis, Kanazawa 921-8001, Ishikawa, Japan;
| | - Maki Ooura
- Maro Clinic, Tanabe 646-0004, Wakayama, Japan;
| | | | - Tsukasa Naganuma
- Department of Nephrology, Yamanashi Prefectural Central Hospital, Kofu 400-0027, Yamanashi, Japan;
| | - Makoto Ogi
- Department of Internal Medicine, Yuurinkouseikai Fuji Hospital, Gotemba 412-0043, Shizuoka, Japan;
| | | | | | - Shigemi Kameda
- Joetsu General Hospital, Joetsu 943-8507, Niigata, Japan;
| | - Sayuri Shirai
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University Yokohama Seibu Hospital, Yokohama 241-0811, Kanagawa, Japan;
| |
Collapse
|
73
|
Liu W, Zhang S, Li Q, Wu Y, Jia X, Feng W, Li Z, Shi Y, Hou Q, Ma J, Liu Y, Gao P, Ganz T, Liu S. Lactate modulates iron metabolism by binding soluble adenylyl cyclase. Cell Metab 2023; 35:1597-1612.e6. [PMID: 37480842 DOI: 10.1016/j.cmet.2023.06.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/18/2023] [Accepted: 06/27/2023] [Indexed: 07/24/2023]
Abstract
Overproduction of lactate (LA) can occur during exercise and in many diseases such as cancers. Individuals with hyperlactatemia often display anemia, decreased serum iron, and elevated hepcidin, a key regulator of iron metabolism. However, it is unknown whether and how LA regulates hepcidin expression. Here, we show LA binds to soluble adenylyl cyclase (sAC) in normal hepatocytes and affects systemic iron homeostasis in mice by increasing hepcidin expression. Comprehensive in vitro, in vivo, and in silico experiments show that the LA-sAC interaction raises cyclic adenosine monophosphate (cAMP) levels, which activates the PKA-Smad1/5/8 signaling pathway to increase hepcidin transcription. We verified this regulatory axis in wild-type mice and in mice with disordered iron homeostasis. LA also regulates hepcidin in humans at rest and subjected to extensive exercise that produce elevated LA. Our study links hyperlactatemia to iron deficiency, offering a mechanistic explanation for anemias seen in athletes and patients with lactic acidosis.
Collapse
Affiliation(s)
- Wei Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Shuping Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Quanjin Li
- University of Chinese Academy of Sciences, Beijing 100049, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yue Wu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuan Jia
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenya Feng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhaolong Li
- University of Chinese Academy of Sciences, Beijing 100049, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yali Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingzhi Hou
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Juan Ma
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yajun Liu
- National Center for Orthopaedics, Beijing Jishuitan Hospital, Beijing 100035, China; Beijing Research Institute of Traumatology and Orthopaedics, Beijing 100035, China
| | - Pu Gao
- University of Chinese Academy of Sciences, Beijing 100049, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sijin Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
74
|
Mast JF, Leach EAE, Thompson TB. Characterization of erythroferrone oligomerization and its impact on BMP antagonism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.01.555965. [PMID: 37693455 PMCID: PMC10491252 DOI: 10.1101/2023.09.01.555965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
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 (CTRP) 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 (LBD), that was predicted to bind BMPs and occlude the type I receptor pocket. We provide evidence that the LBD is crucial for activity through luciferase assays and surface plasmon resonance (SPR) 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
| | - Edmund A E Leach
- Department of Molecular and Cellular Biosciences, University of Cincinnati
| | - Thomas B Thompson
- Department of Molecular and Cellular Biosciences, University of Cincinnati
| |
Collapse
|
75
|
Bäckström F, Chmielewska A, Domellöf M, Berglund SK. Normal range and predictors of serum erythroferrone in infants. Pediatr Res 2023; 94:965-970. [PMID: 37069224 PMCID: PMC10444606 DOI: 10.1038/s41390-023-02594-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/09/2023] [Accepted: 03/20/2023] [Indexed: 04/19/2023]
Abstract
BACKGROUND Erythroferrone (ERFE) has been identified as a hepcidin-regulating hormone synthetized by erythroblasts correlating to the erythropoietic activity and the needs for iron substrate in bone marrow of adults. The present study aimed to assess the ERFE serum concentrations and its predictors in infants. METHODS ERFE was explored at 4 time points during the first year of life in 45 healthy, breastfed, normal birth weight (NBW) infants, and 136 marginally low birth weight infants (LBW, 2000-2500 g) receiving iron (N = 58) or placebo (N = 78) between 6 weeks and 6 months of age. RESULTS ERFE concentrations were low at birth, increasing gradually during the first year of life. In NBW infants, reference ranges (5th to 95th percentile) were at 6 weeks <0.005-0.99 ng/mL and at 12 months <0.005-33.7 ng/mL. ERFE was higher in LBW infants at 6 weeks but lower at 12 months compared to NBW and minimally affected by iron supplementation among LBW infants. Correlations of ERFE with erythropoietic and iron status markers were weak and inconsistent. CONCLUSIONS The role of ERFE in the crosstalk of erythropoiesis and iron homeostasis remains unclear in infants and further studies on ERFE in infants and older children are warranted within the framework of the erythropoietin-ERFE-hepcidin axis. IMPACT Normal range of erythroferrone in healthy infants is described for the first time. Erythroferrone in infants lacks correlation to iron status and markers of erythropoiesis. The findings indicate differences in infant regulation of iron homeostasis as compared to adults. The findings point to a need to study infant erythropoiesis separately from its adult counterpart. The findings may have clinical impact on management strategies of iron-loading anemia in infancy.
Collapse
Affiliation(s)
- Fredrik Bäckström
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Anna Chmielewska
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Magnus Domellöf
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Staffan K Berglund
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden.
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden.
| |
Collapse
|
76
|
Hoving V, Nijssen LE, Donker AE, Roelofs R, Schols SEM, Swinkels DW. Erythropoiesis-hepcidin-iron axis in patients with X-linked sideroblastic anaemia: An explorative biomarker study. Br J Haematol 2023; 202:1216-1219. [PMID: 37469032 DOI: 10.1111/bjh.18986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/28/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023]
Affiliation(s)
- Vera Hoving
- Department of Haematology, Radboud university medical center, Nijmegen, The Netherlands
| | - Lieke E Nijssen
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Department of Paediatrics, Máxima Medical Center, Veldhoven, The Netherlands
| | - Albertine E Donker
- Department of Paediatrics, Máxima Medical Center, Veldhoven, The Netherlands
| | - Rian Roelofs
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Saskia E M Schols
- Department of Haematology, Radboud university medical center, Nijmegen, The Netherlands
| | - Dorine W Swinkels
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Sanquin Blood Bank, Sanquin Diagnostics BV, Amsterdam, The Netherlands
| |
Collapse
|
77
|
Carson JL, Brittenham GM. How I treat anemia with red blood cell transfusion and iron. Blood 2023; 142:777-785. [PMID: 36315909 PMCID: PMC10485845 DOI: 10.1182/blood.2022018521] [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: 09/26/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2022] Open
Abstract
Severe anemia is commonly treated with red blood cell transfusion. Clinical trials have demonstrated that a restrictive transfusion strategy of 7 to 8 g/dL is as safe as a liberal transfusion strategy of 9 to 10 g/dL in many clinical settings. Evidence is lacking for subgroups of patients, including those with preexisting coronary artery disease, acute myocardial infarction, congestive heart failure, and myelodysplastic neoplasms. We present 3 clinical vignettes that highlight the clinical challenges in caring for patients with coronary artery disease with gastrointestinal bleeding, congestive heart failure, or myelodysplastic neoplasms. We emphasize that transfusion practice should be guided by patient symptoms and preferences in conjunction with the patient's hemoglobin concentration. Along with the transfusion decision, evaluation and management of the etiology of the anemia is essential. Iron-restricted erythropoiesis is a common cause of anemia severe enough to be considered for red blood cell transfusion but diagnosis and management of absolute iron deficiency anemia, the anemia of inflammation with functional iron deficiency, or their combination may be problematic. Intravenous iron therapy is generally the treatment of choice for absolute iron deficiency in patients with complex medical disorders, with or without coexisting functional iron deficiency.
Collapse
Affiliation(s)
- Jeffrey L. Carson
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Gary M. Brittenham
- Departments of Pediatrics and Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| |
Collapse
|
78
|
Farag NM, Mousa M, Elsayed E, Ismeil A. GDF-15 and hepcidin as a therapeutic target for anemia in chronic kidney disease. Ital J Pediatr 2023; 49:106. [PMID: 37649102 PMCID: PMC10469522 DOI: 10.1186/s13052-023-01505-9] [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: 01/04/2023] [Accepted: 07/31/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Anaemia is a common presenting feature among patients with chronic kidney disease (CKD) and associated with poor clinical outcomes. We evaluated the diagnostic validity of growth differentiation factor-15 (GDF-15) and hepcidin as it is not clear if they are useful as a biomarkers of anaemia among non-dialysis CKD egyptian patients. METHOD An analytical cross-sectional study was conducted among non-dialysis CKD patients (n = 60) and apparently healthy controls (n = 28) at Minia University maternity & children Hospital. Serum levels of GDF-15 and hepcidin were determined. Predictive logistic regression models were built and post estimation receiver operator characteristics were determined to evaluate diagnostic validity of hepcidin and GDF-15 for iron deficiency anaemia. RESULTS Hepcidin and GDF-15 are significantly higher in cases than control p value (0.047 < 0.0001) respectively. The predictive value of diagnosing anaemia among CKD patients using hepcidin and GDF-15 was 72.0%, 70.0%. There was a weak negative correlation between hepcidin levels and glomerular filtration rate GFR (r = -.175, p = 0.105) in CKD patients, and significant correlation between serum GDF-15 and haemoglobin (r = -0.897, p < 0.0001), ferritin (r = 0.489, P < 0.000), Iron (r = -0.314, P = 0.002), CRP (r = 0.409, P < 0.0001). CONCLUSION Hepcidin and GDF-15 is a potential biomarker for predicting anaemia connected with inflammation among CKD Egyptian patients.
Collapse
Affiliation(s)
| | | | - Eman Elsayed
- Clinical Pathology, Minia University, Minia, Egypt
| | | |
Collapse
|
79
|
Sangkhae V, Fisher AL, Ganz T, Nemeth E. Iron Homeostasis During Pregnancy: Maternal, Placental, and Fetal Regulatory Mechanisms. Annu Rev Nutr 2023; 43:279-300. [PMID: 37253681 PMCID: PMC10723031 DOI: 10.1146/annurev-nutr-061021-030404] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Pregnancy entails a large negative balance of iron, an essential micronutrient. During pregnancy, iron requirements increase substantially to support both maternal red blood cell expansion and the development of the placenta and fetus. As insufficient iron has long been linked to adverse pregnancy outcomes, universal iron supplementation is common practice before and during pregnancy. However, in high-resource countries with iron fortification of staple foods and increased red meat consumption, the effects of too much iron supplementation during pregnancy have become a concern because iron excess has also been linked to adverse pregnancy outcomes. In this review, we address physiologic iron homeostasis of the mother, placenta, and fetus and discuss perturbations in iron homeostasis that result in pathological pregnancy. As many mechanistic regulatory systems have been deduced from animal models, we also discuss the principles learned from these models and how these may apply to human pregnancy.
Collapse
Affiliation(s)
- Veena Sangkhae
- Center for Iron Disorders, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA;
| | - Allison L Fisher
- Endocrine Unit and Nephrology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tomas Ganz
- Center for Iron Disorders, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA;
| | - Elizabeta Nemeth
- Center for Iron Disorders, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA;
| |
Collapse
|
80
|
Ginzburg Y, An X, Rivella S, Goldfarb A. Normal and dysregulated crosstalk between iron metabolism and erythropoiesis. eLife 2023; 12:e90189. [PMID: 37578340 PMCID: PMC10425177 DOI: 10.7554/elife.90189] [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: 06/16/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023] Open
Abstract
Erythroblasts possess unique characteristics as they undergo differentiation from hematopoietic stem cells. During terminal erythropoiesis, these cells incorporate large amounts of iron in order to generate hemoglobin and ultimately undergo enucleation to become mature red blood cells, ultimately delivering oxygen in the circulation. Thus, erythropoiesis is a finely tuned, multifaceted process requiring numerous properly timed physiological events to maintain efficient production of 2 million red blood cells per second in steady state. Iron is required for normal functioning in all human cells, the erythropoietic compartment consuming the majority in light of the high iron requirements for hemoglobin synthesis. Recent evidence regarding the crosstalk between erythropoiesis and iron metabolism sheds light on the regulation of iron availability by erythroblasts and the consequences of insufficient as well as excess iron on erythroid lineage proliferation and differentiation. In addition, significant progress has been made in our understanding of dysregulated iron metabolism in various congenital and acquired malignant and non-malignant diseases. Finally, we report several actual as well as theoretical opportunities for translating the recently acquired robust mechanistic understanding of iron metabolism regulation to improve management of patients with disordered erythropoiesis, such as anemia of chronic inflammation, β-thalassemia, polycythemia vera, and myelodysplastic syndromes.
Collapse
Affiliation(s)
- Yelena Ginzburg
- Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Xiuli An
- LFKRI, New York Blood CenterNew YorkUnited States
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Cell and Molecular Biology affinity group (CAMB), University of PennsylvaniaPhiladelphiaUnited States
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics at the Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Penn Center for Musculoskeletal Disorders at the Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Institute for Regenerative Medicine at University of PennsylvaniaPhiladelphiaUnited States
- RNA Institute at University of PennsylvaniaPhiladelphiaUnited States
| | - Adam Goldfarb
- Department of Pathology, University of VirginiaCharlottesvilleUnited States
| |
Collapse
|
81
|
Ozaki Y, Ohashi K, Otaka N, Kawanishi H, Takikawa T, Fang L, Takahara K, Tatsumi M, Ishihama S, Takefuji M, Kato K, Shimizu Y, Bando YK, Inoue A, Kuzuya M, Miura S, Murohara T, Ouchi N. Myonectin protects against skeletal muscle dysfunction in male mice through activation of AMPK/PGC1α pathway. Nat Commun 2023; 14:4675. [PMID: 37542026 PMCID: PMC10403505 DOI: 10.1038/s41467-023-40435-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 07/28/2023] [Indexed: 08/06/2023] Open
Abstract
To maintain and restore skeletal muscle mass and function is essential for healthy aging. We have found that myonectin acts as a cardioprotective myokine. Here, we investigate the effect of myonectin on skeletal muscle atrophy in various male mouse models of muscle dysfunction. Disruption of myonectin exacerbates skeletal muscle atrophy in age-associated, sciatic denervation-induced or dexamethasone (DEX)-induced muscle atrophy models. Myonectin deficiency also contributes to exacerbated mitochondrial dysfunction and reduces expression of mitochondrial biogenesis-associated genes including PGC1α in denervated muscle. Myonectin supplementation attenuates denervation-induced muscle atrophy via activation of AMPK. Myonectin also reverses DEX-induced atrophy of cultured myotubes through the AMPK/PGC1α signaling. Furthermore, myonectin treatment suppresses muscle atrophy in senescence-accelerated mouse prone (SAMP) 8 mouse model of accelerated aging or mdx mouse model of Duchenne muscular dystrophy. These data indicate that myonectin can ameliorate skeletal muscle dysfunction through AMPK/PGC1α-dependent mechanisms, suggesting that myonectin could represent a therapeutic target of muscle atrophy.
Collapse
Affiliation(s)
- Yuta Ozaki
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Koji Ohashi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Naoya Otaka
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Kawanishi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomonobu Takikawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Lixin Fang
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kunihiko Takahara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Minako Tatsumi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sohta Ishihama
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mikito Takefuji
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Katsuhiro Kato
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuuki Shimizu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuko K Bando
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Aiko Inoue
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masafumi Kuzuya
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinji Miura
- Laboratory of Nutritional Biochemistry, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Noriyuki Ouchi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
82
|
Olivera J, Zhang V, Nemeth E, Ganz T. Erythroferrone exacerbates iron overload and ineffective extramedullary erythropoiesis in a mouse model of β-thalassemia. Blood Adv 2023; 7:3339-3349. [PMID: 36995275 PMCID: PMC10345853 DOI: 10.1182/bloodadvances.2022009307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/08/2023] [Accepted: 03/29/2023] [Indexed: 03/31/2023] Open
Abstract
β-thalassemia is characterized by chronic hepcidin suppression and iron overload, even in patients who have not undergone transfusion. The HbbTh3/+ (Th3/+) mouse model of nontransfusion-dependent β-thalassemia (NTDBT) partially recapitulates the human phenotype but lacks chronic hepcidin suppression, progressive iron accumulation into adulthood, or the interindividual variation of the rate of iron loading observed in patients. Erythroferrone (ERFE) is an erythroid regulator that suppresses hepcidin during increased erythropoiesis. ERFE concentrations in the sera of patients with NTDBT correlate negatively with hepcidin levels but vary over a broad range, possibly explaining the variability of iron overload in patients. To analyze the effect of high ERFE concentrations on hepcidin and iron overload in NTDBT, we crossed Th3/+ mice with erythroid ERFE-overexpressing transgenic mice. Th3/ERFE-transgenic mice suffered high perinatal mortality, but embryos at E18.5 showed similar viability, appearance, and anemia effects as Th3/+ mice. Compared with Th3/+ littermates, adult Th3/ERFE mice had similarly severe anemia but manifested greater suppression of serum hepcidin and increased iron accumulation in the liver, kidney, and spleen. The Th3/ERFE mice had much higher concentrations of serum ERFE than either parental strain, a finding attributable to both a higher number of erythroblasts and higher production of ERFE by each erythroblast.Th3/+ and Th3/ERFE mice had similar red blood cell count and shortened erythrocyte lifespan, but Th3/ERFE mice had an increased number of erythroid precursors in their larger spleens, indicative of aggravated ineffective extramedullary erythropoiesis. Thus, high ERFE concentrations increase the severity of nontransfusional iron overload and ineffective erythropoiesis in thalassemic mice but do not substantially affect anemia or hemolysis.
Collapse
Affiliation(s)
- Joseph Olivera
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Vida Zhang
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Elizabeta Nemeth
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA
- Department of Pathology, David Geffen School of Medicine, UCLA, Los Angeles, CA
| |
Collapse
|
83
|
Yu Y, Woloshun RR, Lee JK, Ebea PO, Zhu S, Nemeth E, Garrick LM, Garrick MD, Collins JF. Fetal factors disrupt placental and maternal iron homeostasis in murine β-thalassemia. Blood 2023; 142:185-196. [PMID: 37146247 PMCID: PMC10352602 DOI: 10.1182/blood.2022018839] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/28/2023] [Accepted: 04/17/2023] [Indexed: 05/07/2023] Open
Abstract
Pregnancy rates in β-thalassemia are increasing but the risk of complications is higher; thus, better understanding of maternal and fetal iron homeostasis in this disorder is needed. HbbTh3/+ (Th3/+) mice model human β-thalassemia. Both the murine and human diseases are characterized by low hepcidin, high iron absorption, and tissue iron overload, with concurrent anemia. We hypothesized that disordered iron metabolism in pregnant Th3/+ mice would negatively affect their unborn offspring. The experimental design included these groups: wild-type (WT) dams carrying WT fetuses (WT1); WT dams carrying WT and Th3/+ fetuses (WT2); Th3/+ dams carrying WT and Th3/+ fetuses (Th3/+); and age-matched, nonpregnant adult females. Serum hepcidin was low, and mobilization of splenic and hepatic storage iron was enhanced in all 3 groups of experimental dams. Intestinal 59Fe absorption was lower in Th3/+ dams (as compared with WT1/2 dams) but splenic 59Fe uptake was higher. Th3/+ dams had hyperferremia, which led to fetal and placenta iron loading, fetal growth restriction, and placentomegaly. Notably, Th3/+ dams loaded Th3/+ and WT fetuses, with the latter situation more closely mirroring human circumstances when mothers with thalassemia have relatively unaffected (thalassemia trait) offspring. Iron-related oxidative stress likely contributed to fetal growth impairment; enhanced placental erythropoiesis is a probable cause of placental enlargement. Moreover, high fetal liver iron transactivated Hamp; fetal hepcidin downregulated placental ferroportin expression, limiting placental iron flux and thus mitigating fetal iron loading. Whether gestational iron loading occurs in human thalassemic pregnancy, when blood transfusion can further elevate serum iron, is worth consideration.
Collapse
Affiliation(s)
- Yang Yu
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL
| | - Regina R. Woloshun
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL
| | - Jennifer K. Lee
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL
| | - Pearl Onuwa Ebea
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL
| | - Sean Zhu
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL
| | - Elizabeta Nemeth
- Department of Medicine, University of California, Los Angeles, CA
| | | | | | - James F. Collins
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL
| |
Collapse
|
84
|
Huang Q, You M, Huang W, Chen J, Zeng Q, Jiang L, Du X, Liu X, Hong M, Wang J. Comparative effectiveness and acceptability of HIF prolyl-hydroxylase inhibitors versus for anemia patients with chronic kidney disease undergoing dialysis: a systematic review and network meta-analysis. Front Pharmacol 2023; 14:1050412. [PMID: 37521459 PMCID: PMC10374033 DOI: 10.3389/fphar.2023.1050412] [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: 09/21/2022] [Accepted: 06/19/2023] [Indexed: 08/01/2023] Open
Abstract
Background: The comparative benefits and acceptability of HIF-PHIs for treating anemia have not been well researched to date. We sought to compare the effectiveness of 6 HIF-PHIs and 3 ESAs for the treatment of renal anemia patients undergoing dialysis. Data sources: Cochrane Central Register of Controlled Trials, PubMed, Embase, Cochrane Library, MEDLINE, Web of Science, and clinicaltrials.gov databases. Results: Twenty-five RCTs (involving 17,204 participants) were included, all of which were designed to achieve target Hb levels by adjusting thee dose of HIF-PHIs. Regarding the efficacy in achieving target Hb levels, no significant differences were found between HIF-PHIs and ESAs in Hb response at the dose-adjusted designed RCTs selected for comparison. Intervention with roxadustat showed a significantly lower risk of RBC transfusion than rhEPO, with an OR and 95% CI of 0.76 (0.56-0.93). Roxadustat and vadadustat had higher risks of increasing the discontinuation rate than ESAs; the former had ORs and 95% CIs of 1.58 (95% CI: 1.21-2.06) for rhEPO, 1.66 (1.16-2.38) for DPO (darbepoetin alfa), and 1.76 (1.70-4.49) for MPG-EPO, and the latter had ORs and 95% CIs of 1.71 (1.09-2.67) for rhEPO, 1.79 (1.29-2.49) for DPO, and 2.97 (1.62-5.46) for MPG-EPO. No differences were observed in the AEs and SAEs among patients who received the studied drugs. Results of a meta-analysis of gastrointestinal disorders among AEs revealed that vadadustat was less effect on causing diarrea than DPO, with an OR of 0.97 (95% CI, 0.9-0.99). Included HIF-PHIs, were proven to be more effective than ESAs in reducing hepcidin levels and increasing TIBC and serum iron level with OR of -0.17 (95% CI, -0.21 to -0.12), OR of 0.79 (95% CI, 0.63-0.95), and OR of 0.39 (95% CI, 0.33-0.45), respectively. Conclusion: HIF-PHIs and ESAs have their characteristics and advantages in treating anemia undergoing dialysis. With the selected dose-adjusted mode, some HIF-PHIs appeared to be a potential treatment for DD-CKD patients when ompared with rhEPO, due to its effectiveness in decreasing the risk of RBC transfusion rate or regulating iron or lipid metabolism while achieving target Hb levels. Systematic Review Registration: https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=306511; Identifier: CRD42022306511.
Collapse
Affiliation(s)
- Qiong Huang
- Department of Nephropathy, Luohu District Traditional Chinese Medicine Hospital, Shenzhen, China
- Guangzhou University of Chinese Traditional Medicine, Guangzhou, China
| | - Minling You
- Department of Nephropathy, Luohu District Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Weijuan Huang
- Department of Nephropathy, Luohu District Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Jian Chen
- Department of Nephropathy, Luohu District Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Qinming Zeng
- Department of Nephropathy, Luohu District Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Longfeng Jiang
- Department of Nephropathy, Luohu District Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Xiuben Du
- LuoHu Center for Chronic Disease Control, Shenzhen, China
| | - Xusheng Liu
- Guangzhou University of Chinese Traditional Medicine, Guangzhou, China
| | - Ming Hong
- Institute of Advanced Diagnostic and Clinical Medicine, Zhongshan City People’s Hospital, Affiliated Zhongshan Hospital of Sun Yat-sen University, Zhongshan, China
| | - Jing Wang
- Department of Nephropathy, Luohu District Traditional Chinese Medicine Hospital, Shenzhen, China
| |
Collapse
|
85
|
Courbon G, Thomas JJ, Martinez-Calle M, Wang X, Spindler J, Von Drasek J, Hunt-Tobey B, Mehta R, Isakova T, Chang W, Creemers JWM, Ji P, Martin A, David V. Bone-derived C-terminal FGF23 cleaved peptides increase iron availability in acute inflammation. Blood 2023; 142:106-118. [PMID: 37053547 PMCID: PMC10356820 DOI: 10.1182/blood.2022018475] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/24/2023] [Accepted: 04/06/2023] [Indexed: 04/15/2023] Open
Abstract
Inflammation leads to functional iron deficiency by increasing the expression of the hepatic iron regulatory peptide hepcidin. Inflammation also stimulates fibroblast growth factor 23 (FGF23) production by increasing both Fgf23 transcription and FGF23 cleavage, which paradoxically leads to excess in C-terminal FGF23 peptides (Cter-FGF23), rather than intact FGF23 (iFGF23) hormone. We determined that the major source of Cter-FGF23 is osteocytes and investigated whether Cter-FGF23 peptides play a direct role in the regulation of hepcidin and iron metabolism in response to acute inflammation. Mice harboring an osteocyte-specific deletion of Fgf23 showed a ∼90% reduction in Cter-FGF23 levels during acute inflammation. Reduction in Cter-FGF23 led to a further decrease in circulating iron in inflamed mice owing to excessive hepcidin production. We observed similar results in mice showing impaired FGF23 cleavage owing to osteocyte-specific deletion of Furin. We next showed that Cter-FGF23 peptides bind members of the bone morphogenetic protein (BMP) family, BMP2 and BMP9, which are established inducers of hepcidin. Coadministration of Cter-FGF23 and BMP2 or BMP9 prevented the increase in Hamp messenger RNA and circulating hepcidin levels induced by BMP2/9, resulting in normal serum iron levels. Finally, injection of Cter-FGF23 in inflamed Fgf23KO mice and genetic overexpression of Cter-Fgf23 in wild type mice also resulted in lower hepcidin and higher circulating iron levels. In conclusion, during inflammation, bone is the major source of Cter-FGF23 secretion, and independently of iFGF23, Cter-FGF23 reduces BMP-induced hepcidin secretion in the liver.
Collapse
Affiliation(s)
- Guillaume Courbon
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Jane Joy Thomas
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Marta Martinez-Calle
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Xueyan Wang
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Jadeah Spindler
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - John Von Drasek
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Bridget Hunt-Tobey
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Rupal Mehta
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Tamara Isakova
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Wenhan Chang
- Endocrine Research Unit, San Francisco Veterans Affairs Medical Center, University of California San Francisco, San Francisco, CA
| | | | - Peng Ji
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Aline Martin
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Valentin David
- Division of Nephrology and Hypertension, Department of Medicine, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
86
|
Hilton C, Sabaratnam R, Drakesmith H, Karpe F. Iron, glucose and fat metabolism and obesity: an intertwined relationship. Int J Obes (Lond) 2023; 47:554-563. [PMID: 37029208 PMCID: PMC10299911 DOI: 10.1038/s41366-023-01299-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 03/08/2023] [Accepted: 03/16/2023] [Indexed: 04/09/2023]
Abstract
A bidirectional relationship exists between adipose tissue metabolism and iron regulation. Total body fat, fat distribution and exercise influence iron status and components of the iron-regulatory pathway, including hepcidin and erythroferrone. Conversely, whole body and tissue iron stores associate with fat mass and distribution and glucose and lipid metabolism in adipose tissue, liver, and muscle. Manipulation of the iron-regulatory proteins erythroferrone and erythropoietin affects glucose and lipid metabolism. Several lines of evidence suggest that iron accumulation and metabolism may play a role in the development of metabolic diseases including obesity, type 2 diabetes, hyperlipidaemia and non-alcoholic fatty liver disease. In this review we summarise the current understanding of the relationship between iron homoeostasis and metabolic disease.
Collapse
Affiliation(s)
- Catriona Hilton
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK.
| | - Rugivan Sabaratnam
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| |
Collapse
|
87
|
Olivari V, Di Modica SM, Lidonnici MR, Aghajan M, Cordero-Sanchez C, Tanzi E, Pettinato M, Pagani A, Tiboni F, Silvestri L, Guo S, Ferrari G, Nai A. A single approach to targeting transferrin receptor 2 corrects iron and erythropoietic defects in murine models of anemia of inflammation and chronic kidney disease. Kidney Int 2023; 104:61-73. [PMID: 36990212 DOI: 10.1016/j.kint.2023.03.012] [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: 07/27/2022] [Revised: 02/24/2023] [Accepted: 03/08/2023] [Indexed: 03/29/2023]
Abstract
Anemia is a common complication of systemic inflammation. Proinflammatory cytokines both decrease erythroblast sensitivity to erythropoietin (EPO) and increase the levels of the hepatic hormone hepcidin, sequestering iron in stores and causing functional iron deficiency. Anemia of chronic kidney disease (CKD) is a peculiar form of anemia of inflammation, characterized by impaired EPO production paralleling progressive kidney damage. Traditional therapy based on increased EPO (often in combination with iron) may have off-target effects due to EPO interaction with its non-erythroid receptors. Transferrin Receptor 2 (Tfr2) is a mediator of the iron-erythropoiesis crosstalk. Its deletion in the liver hampers hepcidin production, increasing iron absorption, whereas its deletion in the hematopoietic compartment increases erythroid EPO sensitivity and red blood cell production. Here, we show that selective hematopoietic Tfr2 deletion ameliorates anemia in mice with sterile inflammation in the presence of normal kidney function, promoting EPO responsiveness and erythropoiesis without increasing serum EPO levels. In mice with CKD, characterized by absolute rather than functional iron deficiency, Tfr2 hematopoietic deletion had a similar effect on erythropoiesis but anemia improvement was transient because of limited iron availability. Also, increasing iron levels by downregulating only hepatic Tfr2 had a minor effect on anemia. However, simultaneous deletion of hematopoietic and hepatic Tfr2, stimulating erythropoiesis and increased iron supply, was sufficient to ameliorate anemia for the entire protocol. Thus, our results suggest that combined targeting of hematopoietic and hepatic Tfr2 may be a therapeutic option to balance erythropoiesis stimulation and iron increase, without affecting EPO levels.
Collapse
Affiliation(s)
- Violante Olivari
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (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, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Maria Rosa Lidonnici
- Gene Transfer into Stem Cell Unit, SR-Tiget, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | | | - Celia Cordero-Sanchez
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Emanuele Tanzi
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Mariateresa Pettinato
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Alessia Pagani
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Francesca Tiboni
- Gene Transfer into Stem Cell Unit, SR-Tiget, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Laura Silvestri
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Shuling Guo
- Ionis Pharmaceuticals Inc., Carlsbad, California, USA
| | - Giuliana Ferrari
- Vita-Salute San Raffaele University, Milan, Italy; Gene Transfer into Stem Cell Unit, SR-Tiget, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Antonella Nai
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
88
|
Delaney KM, Barad A, Castillo LF, Hasund CM, Guillet R, Pressman EK, Katzman PJ, Ganz T, Nemeth E, O'Brien KO. Placental Erythroferrone and Erythropoietin mRNA Expression is not Associated with Maternal or Neonatal Iron Status in Adolescents Carrying Singletons and Adult Women Carrying Multiples. J Nutr 2023; 153:1950-1958. [PMID: 37253412 PMCID: PMC10375499 DOI: 10.1016/j.tjnut.2023.05.023] [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: 02/25/2023] [Revised: 04/27/2023] [Accepted: 05/26/2023] [Indexed: 06/01/2023] Open
Abstract
BACKGROUND The iron regulatory hormones erythroferrone (ERFE), erythropoietin (EPO), and hepcidin, and the cargo receptor nuclear receptor coactivator 4 (NCOA4) are expressed in the placenta. However, determinants of placental expression of these proteins and their associations with maternal or neonatal iron status are unknown. OBJECTIVES To characterize expression of placental ERFE, EPO, and NCOA4 mRNA in placentae from newborns at increased risk of iron deficiency and to evaluate these in relation to maternal and neonatal iron status and regulatory hormones. METHODS Placentae were collected from 114 neonates born to adolescents carrying singletons (14-18 y) and 110 neonates born to 54 adults (20-46 y) carrying multiples. Placental EPO, ERFE, and NCOA4 mRNA expression were measured by RT-qPCR and compared with maternal and neonatal iron status indicators (SF, sTfR, total body iron, serum iron) and hormones. RESULTS Placental ERFE, EPO, and NCOA4 mRNA were detected in all placentae delivered between 25 and 42 wk of gestation. Relationships between placental ERFE and EPO differed by cohort. In the multiples cohort, placental EPO and ERFE were positively correlated (P = 0.004), but only a positive trend (P = 0.08) was evident in the adolescents. Placental EPO and ERFE were not associated with maternal or neonatal iron status markers or hormones in either cohort. Placental NCOA4 was not associated with placental EPO or ERFE in either cohort but was negatively associated with maternal SF (P = 0.03) in the multiples cohort and positively associated with neonatal sTfR (P = 0.009) in the adolescents. CONCLUSIONS The human placenta expresses ERFE, EPO, and NCOA4 mRNA as early as 25 wk of gestation. Placental expression of ERFE and EPO transcripts was not associated with maternal or neonatal iron status. Greater placental NCOA4 transcript expression was evident in women and newborns with poor iron status (lower SF and higher sTfR, respectively). Further research is needed to characterize the roles of these proteins in the human placenta. TRIAL REGISTRATION NUMBER These clinical trials were registered at clinicaltrials.gov as NCT01019902 (https://clinicaltrials.gov/ct2/show/NCT01019902) and NCT01582802 (https://clinicaltrials.gov/ct2/show/NCT01582802).
Collapse
Affiliation(s)
- Katherine M Delaney
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Alexa Barad
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Luisa F Castillo
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Chloe M Hasund
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Ronnie Guillet
- Department of Pediatrics Division of Neonatology, University of Rochester School of Medicine, Rochester, NY, United States
| | - Eva K Pressman
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine, Rochester, NY, United States
| | - Philip J Katzman
- Department of Pathology and Clinical Laboratory Medicine, University of Rochester School of Medicine, Rochester, NY, United States
| | - Tomas Ganz
- David Geffen School of Medicine, Center for Iron Disorders, University of California Los Angeles, Los Angeles, CA, United States
| | - Elizabeta Nemeth
- David Geffen School of Medicine, Center for Iron Disorders, University of California Los Angeles, Los Angeles, CA, United States
| | - Kimberly O O'Brien
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States.
| |
Collapse
|
89
|
Cui F, Sun J, Mi H, Li B, Tang L, Wang R, Du Y, Guo B, Li Y, Shi M. Chronic intermittent hypobaric hypoxia improves iron metabolism disorders via the IL-6/JAK2/STAT3 and Epo/STAT5/ERFE signaling pathways in metabolic syndrome rats. J Trace Elem Med Biol 2023; 79:127259. [PMID: 37413927 DOI: 10.1016/j.jtemb.2023.127259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/09/2023] [Accepted: 06/28/2023] [Indexed: 07/08/2023]
Abstract
AIM Our previous study demonstrated that chronic intermittent hypobaric hypoxia (CIHH) improved iron metabolism disorder in obese rats through the downregulation of hepcidin. This study aimed to observe the molecular mechanism of CIHH in improving iron metabolism disorders, especially by Janus kinase/signal transducer and activation of the transcription (JAK/STAT) signaling pathway in metabolic syndrome (MS) rats. METHODS Six-week-old male Sprague-Dawley rats were randomly divided into four groups: CON, CIHH (subjected to hypobaric hypoxia simulating 5000-m altitude for 28 days, 6 h daily), MS (induced by high fat diet and fructose water), and MS+CIHH. The serum levels of glucose, lipid metabolism, iron metabolism, interleukin-6 (IL-6), erythropoietin (Epo) and hepcidin were measured. The protein expressions of JAK2, STAT3, STAT5, bone morphogenetic protein 6 (BMP6), small mothers against decapentaplegic 1 (SMAD1) and hepcidin were examined. The mRNA expressions of erythroferrone (ERFE) and hepcidin were analyzed. RESULTS The MS rats displayed obesity, hyperglycemia, hyperlipidemia, iron metabolism disorder, increased IL-6 and hepcidin serum levels, upregulation of JAK2/STAT3 signaling pathway, decreased Epo serum levels, downregulation of STAT5/ERFE signaling pathway in spleen, upregulation of BMP/SMAD signaling pathway in liver, and increased hepcidin mRNA and protein expression compared to CON rats. All the aforementioned abnormalities in MS rats were ameliorated in MS + CIHH rats. CONCLUSIONS CIHH improved iron metabolism disorders, possibly by inhibiting IL-6/JAK2/STAT3 and activating Epo/STAT5/ERFE signaling pathway, thus downregulating hepcidin in MS rats.
Collapse
Affiliation(s)
- Fang Cui
- Department of Clinical Laboratory, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, PR China; Department of Electron Microscope Laboratory Centre, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Jie Sun
- Department of Clinical Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, PR China
| | - Haichao Mi
- Department of Laboratory Medicine, Linyi Peoples' Hospital, Linyi 276000, PR China
| | - Bo Li
- Department of Emergency, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang, 050001, Hebei, PR China
| | - Longmei Tang
- School of Public Health, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Province Key Laboratory of Environment and Human Health, Shijiazhuang 050017, PR China
| | - Ruotong Wang
- Department of Clinical Laboratory, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, PR China
| | - Yutao Du
- Department of Clinical Laboratory, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, PR China
| | - Bingyan Guo
- Department of Cardiovascular Medicine, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, PR China
| | - Yongjun Li
- Department of Clinical Laboratory, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, PR China
| | - Min Shi
- Department of Clinical Laboratory, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, PR China.
| |
Collapse
|
90
|
Bennett C, Jackson VE, Pettikiriarachchi A, Hayman T, Schaeper U, Moir-Meyer G, Fielding K, Ataide R, Clucas D, Baldi A, Garnham AL, Li-Wai-Suen CSN, Loughran SJ, Baxter EJ, Green AR, Alexander WS, Bahlo M, Burbury K, Ng AP, Pasricha SR. Iron homeostasis governs erythroid phenotype in polycythemia vera. Blood 2023; 141:3199-3214. [PMID: 36928379 PMCID: PMC10646816 DOI: 10.1182/blood.2022016779] [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: 04/22/2022] [Revised: 03/08/2023] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
Polycythemia vera (PV) is a myeloproliferative neoplasm driven by activating mutations in JAK2 that result in unrestrained erythrocyte production, increasing patients' hematocrit and hemoglobin concentrations, placing them at risk of life-threatening thrombotic events. Our genome-wide association study of 440 PV cases and 403 351 controls using UK Biobank data showed that single nucleotide polymorphisms in HFE known to cause hemochromatosis are highly associated with PV diagnosis, linking iron regulation to PV. Analysis of the FinnGen dataset independently confirmed overrepresentation of homozygous HFE variants in patients with PV. HFE influences the expression of hepcidin, the master regulator of systemic iron homeostasis. Through genetic dissection of mouse models of PV, we show that the PV erythroid phenotype is directly linked to hepcidin expression: endogenous hepcidin upregulation alleviates erythroid disease whereas hepcidin ablation worsens it. Furthermore, we demonstrate that in PV, hepcidin is not regulated by expanded erythropoiesis but is likely governed by inflammatory cytokines signaling via GP130-coupled receptors. These findings have important implications for understanding the pathophysiology of PV and offer new therapeutic strategies for this disease.
Collapse
Affiliation(s)
- Cavan Bennett
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Victoria E. Jackson
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Anne Pettikiriarachchi
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Thomas Hayman
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | | | - Gemma Moir-Meyer
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Katherine Fielding
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Diagnostic Haematology, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Ricardo Ataide
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Infectious Diseases, Peter Doherty Institute, University of Melbourne, Parkville, VIC, Australia
| | - Danielle Clucas
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Diagnostic Haematology, The Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Andrew Baldi
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Alexandra L. Garnham
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Connie S. N. Li-Wai-Suen
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Stephen J. Loughran
- Wellcome–MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - E. Joanna Baxter
- Wellcome–MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Anthony R. Green
- Wellcome–MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Warren S. Alexander
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Melanie Bahlo
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Kate Burbury
- Clinical Haematology at the Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Ashley P. Ng
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Blood Cells and Blood Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Clinical Haematology at the Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Sant-Rayn Pasricha
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Diagnostic Haematology, The Royal Melbourne Hospital, Parkville, VIC, Australia
- Clinical Haematology at the Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, VIC, Australia
| |
Collapse
|
91
|
Gao G, You L, Zhang J, Chang YZ, Yu P. Brain Iron Metabolism, Redox Balance and Neurological Diseases. Antioxidants (Basel) 2023; 12:1289. [PMID: 37372019 DOI: 10.3390/antiox12061289] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The incidence of neurological diseases, such as Parkinson's disease, Alzheimer's disease and stroke, is increasing. An increasing number of studies have correlated these diseases with brain iron overload and the resulting oxidative damage. Brain iron deficiency has also been closely linked to neurodevelopment. These neurological disorders seriously affect the physical and mental health of patients and bring heavy economic burdens to families and society. Therefore, it is important to maintain brain iron homeostasis and to understand the mechanism of brain iron disorders affecting reactive oxygen species (ROS) balance, resulting in neural damage, cell death and, ultimately, leading to the development of disease. Evidence has shown that many therapies targeting brain iron and ROS imbalances have good preventive and therapeutic effects on neurological diseases. This review highlights the molecular mechanisms, pathogenesis and treatment strategies of brain iron metabolism disorders in neurological diseases.
Collapse
Affiliation(s)
- Guofen Gao
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| | - Linhao You
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| | - Jianhua Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| | - Yan-Zhong Chang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| | - Peng Yu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, No. 20 Nan'erhuan Eastern Road, Shijiazhuang 050024, China
| |
Collapse
|
92
|
Pandur E, Pap R, Jánosa G, Horváth A, Sipos K. The Role of Fractalkine in the Regulation of Endometrial Iron Metabolism in Iron Deficiency. Int J Mol Sci 2023; 24:9917. [PMID: 37373063 PMCID: PMC10298019 DOI: 10.3390/ijms24129917] [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: 05/15/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Iron is a crucial element in the human body. Endometrial iron metabolism is implicated in endometrium receptivity and embryo implantation. Disturbances of the maternal as well as the endometrial iron homeostasis, such as iron deficiency, can contribute to the reduced development of the fetus and could cause an increased risk of adverse pregnancy outcomes. Fractalkine is a unique chemokine that plays a role in the communication between the mother and the fetus. It has been demonstrated that FKN is involved in the development of endometrial receptivity and embryo implantation, and it functions as a regulator of iron metabolism. In the present study, we examined the effect of FKN on the iron metabolism of HEC-1A endometrial cells in a state of iron deficiency mediated by desferrioxamine treatment. Based on the findings, FKN enhances the expression of iron metabolism-related genes in iron deficiency and modifies the iron uptake via transferrin receptor 1 and divalent metal transporter-1, and iron release via ferroportin. FKN can activate the release of iron from heme-containing proteins by elevating the level of heme oxygenase-1, contributing to the redistribution of intracellular iron content. It was revealed that the endometrium cells express both mitoferrin-1 and 2 and that their levels are not dependent on the iron availability of the cells. FKN may also contribute to maintaining mitochondrial iron homeostasis. FKN can improve the deteriorating effect of iron deficiency in HEC-1A endometrium cells, which may contribute to the development of receptivity and/or provide iron delivery towards the embryo.
Collapse
Affiliation(s)
- Edina Pandur
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, H-7624 Pécs, Hungary; (R.P.); (G.J.); (A.H.); (K.S.)
- National Laboratory on Human Reproduction, University of Pécs, H-7624 Pécs, Hungary
| | - Ramóna Pap
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, H-7624 Pécs, Hungary; (R.P.); (G.J.); (A.H.); (K.S.)
- National Laboratory on Human Reproduction, University of Pécs, H-7624 Pécs, Hungary
| | - Gergely Jánosa
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, H-7624 Pécs, Hungary; (R.P.); (G.J.); (A.H.); (K.S.)
| | - Adrienn Horváth
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, H-7624 Pécs, Hungary; (R.P.); (G.J.); (A.H.); (K.S.)
| | - Katalin Sipos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, H-7624 Pécs, Hungary; (R.P.); (G.J.); (A.H.); (K.S.)
- National Laboratory on Human Reproduction, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
93
|
Bayır H, Dixon SJ, Tyurina YY, Kellum JA, Kagan VE. Ferroptotic mechanisms and therapeutic targeting of iron metabolism and lipid peroxidation in the kidney. Nat Rev Nephrol 2023; 19:315-336. [PMID: 36922653 DOI: 10.1038/s41581-023-00689-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 03/17/2023]
Abstract
Ferroptosis is a mechanism of regulated necrotic cell death characterized by iron-dependent, lipid peroxidation-driven membrane destruction that can be inhibited by glutathione peroxidase 4. Morphologically, it is characterized by cellular, organelle and cytoplasmic swelling and the loss of plasma membrane integrity, with the release of intracellular components. Ferroptosis is triggered in cells with dysregulated iron and thiol redox metabolism, whereby the initial robust but selective accumulation of hydroperoxy polyunsaturated fatty acid-containing phospholipids is further propagated through enzymatic and non-enzymatic secondary mechanisms, leading to formation of oxidatively truncated electrophilic species and their adducts with proteins. Thus, ferroptosis is dependent on the convergence of iron, thiol and lipid metabolic pathways. The kidney is particularly susceptible to redox imbalance. A growing body of evidence has linked ferroptosis to acute kidney injury in the context of diverse stimuli, such as ischaemia-reperfusion, sepsis or toxins, and to chronic kidney disease, suggesting that ferroptosis may represent a novel therapeutic target for kidney disease. However, further work is needed to address gaps in our understanding of the triggers, execution and spreading mechanisms of ferroptosis.
Collapse
Affiliation(s)
- Hülya Bayır
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Free Radical and Antioxidant Health, Departments of Environmental Health, Pharmacology and Chemical Biology, Chemistry, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Pediatrics, Division of Critical Care and Hospital Medicine, Redox Health Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Yulia Y Tyurina
- Center for Free Radical and Antioxidant Health, Departments of Environmental Health, Pharmacology and Chemical Biology, Chemistry, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - John A Kellum
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, Departments of Environmental Health, Pharmacology and Chemical Biology, Chemistry, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
94
|
Rodrigues F, Coman T, Fouquet G, Côté F, Courtois G, Trovati Maciel T, Hermine O. A deep dive into future therapies for microcytic anemias and clinical considerations. Expert Rev Hematol 2023; 16:349-364. [PMID: 37092971 DOI: 10.1080/17474086.2023.2206556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
INTRODUCTION Microcytic anemias (MA) have frequent or rare etiologies. New discoveries in understanding and treatment of microcytic anemias need to be reviewed. AREAS COVERED Microcytic anemias with a focus on most frequent causes and on monogenic diseases that are relevant for understanding biocellular mechanisms of MA. All treatments excepting gene therapy, with a focus on recent advances. Pubmed search with references selected by expert opinion. EXPERT OPINION As the genetic and cellular background of dyserythropoiesis will continue to be clarified, collaboration with bioengineering of treatments acting specifically at the protein domain level will continue to provide new therapies in haematology as well as oncology and neurology.
Collapse
Affiliation(s)
- François Rodrigues
- Université de Paris, service d'hématologie adultes, Hôpital Necker - Enfants Malades, Asrsistance Publique- Hôpitaux de Paris, France
- Inserm U1163, CNRS ERL8254 Imagine Institute, Paris, France
| | - Tereza Coman
- Inserm U1163, CNRS ERL8254 Imagine Institute, Paris, France
- Département d'hématologie, Institut Gustave Roussy, Villejuif, France
| | - Guillemette Fouquet
- Université de Paris, service d'hématologie adultes, Hôpital Necker - Enfants Malades, Asrsistance Publique- Hôpitaux de Paris, France
- Hématologie clinique, Centre Hospitalier Sud Francilien, Corbeil Essonnes, France
| | - Francine Côté
- Inserm U1163, CNRS ERL8254 Imagine Institute, Paris, France
| | | | | | - Olivier Hermine
- Université de Paris, service d'hématologie adultes, Hôpital Necker - Enfants Malades, Asrsistance Publique- Hôpitaux de Paris, France
- Inserm U1163, CNRS ERL8254 Imagine Institute, Paris, France
| |
Collapse
|
95
|
Yang J, Li Q, Feng Y, Zeng Y. Iron Deficiency and Iron Deficiency Anemia: Potential Risk Factors in Bone Loss. Int J Mol Sci 2023; 24:ijms24086891. [PMID: 37108056 PMCID: PMC10138976 DOI: 10.3390/ijms24086891] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/01/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Iron is one of the essential mineral elements for the human body and this nutrient deficiency is a worldwide public health problem. Iron is essential in oxygen transport, participates in many enzyme systems in the body, and is an important trace element in maintaining basic cellular life activities. Iron also plays an important role in collagen synthesis and vitamin D metabolism. Therefore, decrease in intracellular iron can lead to disturbance in the activity and function of osteoblasts and osteoclasts, resulting in imbalance in bone homeostasis and ultimately bone loss. Indeed, iron deficiency, with or without anemia, leads to osteopenia or osteoporosis, which has been revealed by numerous clinical observations and animal studies. This review presents current knowledge on iron metabolism under iron deficiency states and the diagnosis and prevention of iron deficiency and iron deficiency anemia (IDA). With emphasis, studies related to iron deficiency and bone loss are discussed, and the potential mechanisms of iron deficiency leading to bone loss are analyzed. Finally, several measures to promote complete recovery and prevention of iron deficiency are listed to improve quality of life, including bone health.
Collapse
Affiliation(s)
- Jiancheng Yang
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Qingmei Li
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Yan Feng
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| | - Yuhong Zeng
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, China
| |
Collapse
|
96
|
Petro JL, Fragozo-Ramos MC, Milán AF, Aristizabal JC, Gallo-Villegas JA, Calderón JC. Serum Levels of Myonectin Are Lower in Adults with Metabolic Syndrome and Are Negatively Correlated with Android Fat Mass. Int J Mol Sci 2023; 24:ijms24086874. [PMID: 37108038 PMCID: PMC10138930 DOI: 10.3390/ijms24086874] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 04/29/2023] Open
Abstract
Myonectin has shown beneficial effects on lipid regulation in murine models; therefore, it may have implications in the pathophysiology of metabolic syndrome (MS). We evaluated the relationship between serum myonectin and serum lipids, global and regional fat mass, intramuscular lipid content, and insulin resistance (IR) in adults with metabolic risk factors. This was a cross-sectional study in sedentary adults who were diagnosed with MS or without MS (NMS). Serum myonectin was quantified by enzyme-linked immunosorbent assay, lipid profile by conventional techniques, and free fatty acids (FFA) by gas chromatography. Body composition was assessed by dual-energy X-ray absorptiometry and intramuscular lipid content through proton nuclear magnetic resonance spectroscopy in the right vastus lateralis muscle. IR was estimated with the homeostatic model assessment (HOMA-IR). The MS (n = 61) and NMS (n = 29) groups were comparable in age (median (interquartile range): 51.0 (46.0-56.0) vs. 53.0 (45.5-57.5) years, p > 0.05) and sex (70.5% men vs. 72.4% women). MS subjects had lower serum levels of myonectin than NMS subjects (1.08 (0.87-1.35) vs. 1.09 (0.93-4.05) ng·mL-1, p < 0.05). Multiple linear regression models adjusted for age, sex, fat mass index and lean mass index showed that serum myonectin was negatively correlated with the android/gynoid fat mass ratio (R2 = 0.48, p < 0.01), but not with the lipid profile, FFA, intramuscular lipid content or HOMA-IR. In conclusion, serum myonectin is lower in subjects with MS. Myonectin negatively correlates with a component relevant to the pathophysiology of MS, such as the android/gynoid fat mass ratio, but not with other components such as FFA, intramuscular fat or IR.
Collapse
Affiliation(s)
- Jorge L Petro
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellín 050010, Colombia
- Research Group in Physical Activity, Sports and Health Sciences-GICAFS, Universidad de Córdoba, Montería 230002, Colombia
| | - María Carolina Fragozo-Ramos
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellín 050010, Colombia
| | - Andrés F Milán
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellín 050010, Colombia
| | - Juan C Aristizabal
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellín 050010, Colombia
| | - Jaime A Gallo-Villegas
- Sports Medicine Postgraduate Program, and GRINMADE Research Group, SICOR Center, Faculty of Medicine, University of Antioquia, Medellín 050010, Colombia
| | - Juan C Calderón
- Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellín 050010, Colombia
| |
Collapse
|
97
|
Sardo U, Perrier P, Cormier K, Sotin M, 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 the recovery from hemorrhage-induced anemia in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.06.535920. [PMID: 37066218 PMCID: PMC10104156 DOI: 10.1101/2023.04.06.535920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
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 adequate supply of iron to the bone marrow for red blood cells production. However, mounting evidence suggested that another factor may exert a similar function. We identified the hepatokine 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 a blunted repression of hepcidin after bleeding. FGL1 exerts its activity by direct binding to BMP6, thereby inhibiting the canonical BMP-SMAD signaling cascade that controls hepcidin transcription. Key points 1/ FGL1 regulates iron metabolism during the recovery from anemia. 2/ FGL1 is an antagonist of the BMP/SMAD signaling pathway.
Collapse
|
98
|
Ganz T, Nemeth E, Rivella S, Goldberg P, Dibble AR, McCaleb ML, Guo S, Monia BP, Barrett TD. TMPRSS6 as a Therapeutic Target for Disorders of Erythropoiesis and Iron Homeostasis. Adv Ther 2023; 40:1317-1333. [PMID: 36690839 PMCID: PMC10070284 DOI: 10.1007/s12325-022-02421-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 12/23/2022] [Indexed: 01/25/2023]
Abstract
TMPRSS6 is a serine protease highly expressed in the liver. Its role in iron regulation was first reported in 2008 when mutations in TMPRSS6 were shown to be the cause of iron-refractory iron deficiency anemia (IRIDA) in humans and in mouse models. TMPRSS6 functions as a negative regulator of the expression of the systemic iron-regulatory hormone hepcidin. Over the last decade and a half, growing understanding of TMPRSS6 biology and mechanism of action has enabled development of new therapeutic approaches for patients with diseases of erythropoiesis and iron homeostasis.ClinicalTrials.gov identifier NCT03165864.
Collapse
Affiliation(s)
- Tomas Ganz
- Department of Medicine and Pathology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
| | - Elizabeta Nemeth
- Department of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology, Children's Hospital of Philadelphia (CHOP), 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Cell and Molecular Biology Graduate Group (CAMB), University of Pennsylvania, Abramson Research Center, 3615 Civic Center Boulevard, Room 316B, Philadelphia, PA, 19104, USA
| | - Paul Goldberg
- Prilenia Therapeutics, Herzliya, Israel
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | | | | | - Shuling Guo
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | | | | |
Collapse
|
99
|
Abstract
Iron overload remains a lethal complication of β-thalassemia and other anemias caused by ineffective erythropoiesis. This review discusses the pathogenetic mechanisms of iron overload in thalassemia, at organismal, cellular, and molecular levels.
Collapse
Affiliation(s)
- Tomas Ganz
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA.
| | - Elizabeta Nemeth
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA
| |
Collapse
|
100
|
Ginzburg YZ. Hepcidin and its multiple partners: Complex regulation of iron metabolism in health and disease. VITAMINS AND HORMONES 2023; 123:249-284. [PMID: 37717987 DOI: 10.1016/bs.vh.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
The peptide hormone hepcidin is central to the regulation of iron metabolism, influencing the movement of iron into the circulation and determining total body iron stores. Its effect on a cellular level involves binding ferroportin, the main iron export protein, preventing iron egress and leading to iron sequestration within ferroportin-expressing cells. Hepcidin expression is enhanced by iron loading and inflammation and suppressed by erythropoietic stimulation. Aberrantly increased hepcidin leads to systemic iron deficiency and/or iron restricted erythropoiesis as occurs in anemia of chronic inflammation. Furthermore, insufficiently elevated hepcidin occurs in multiple diseases associated with iron overload such as hereditary hemochromatosis and iron loading anemias. Abnormal iron metabolism as a consequence of hepcidin dysregulation is an underlying factor resulting in pathophysiology of multiple diseases and several agents aimed at manipulating this pathway have been designed, with some already in clinical trials. In this chapter, we assess the complex regulation of hepcidin, delineate the many binding partners involved in its regulation, and present an update on the development of hepcidin agonists and antagonists in various clinical scenarios.
Collapse
Affiliation(s)
- Yelena Z Ginzburg
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, United Sates.
| |
Collapse
|