1
|
De la Cruz-Garcinuño M, Juárez-Vela R, Lasa-Berasain P, Ruiz de Viñaspre-Hernández R, Czapla M, García-Muñoz L, Polo-Andrade E, Sarmiento C, Rodero-Martínez J, Alonso-Arias M, López-Tornero S, Quintana-Díaz M. Utility of reticulocyte hemoglobin as a new predictor of anemia in intensive care unit patients. Front Med (Lausanne) 2025; 12:1577047. [PMID: 40270491 PMCID: PMC12014566 DOI: 10.3389/fmed.2025.1577047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 03/25/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction Iron deficiency and anemia are common complications in critically ill patients, particularly in the Intensive Care Unit setting (ICU), where inflammation and infection are prevalent. Traditional markers like ferritin are unreliable in these contexts due to their behavior as acute-phase reactants. New hematimetric indices, such as Reticulocyte Hemoglobin Equivalent (RET-He) and Delta Hemoglobin Equivalent (Delta-He), may offer better predictive value for anemia in ICU patients. Objectives This study aimed to evaluate the predictive utility of RET-He and Delta-He for anemia in critically ill patients and compare their performance with serum ferritin levels. Methods A pilot, observational, prospective study was conducted on 40 ICU patients admitted for burns or polytrauma. Hematological and hematimetric parameters were analyzed at admission, 48 h, 4 days, and 7 days post-admission. Mixed-effects regression models were used to assess the predictive value of RET-He, Delta-He, and ferritin for hemoglobin levels and anemia. Results Significant reductions in hemoglobin and hematocrit were observed within the first 48 h of ICU admission, while RET-He and Delta-He remained stable. Over 4 and 7 days, decreases in RET-He and Delta-He were strongly associated with lower hemoglobin levels and increased risk of anemia (p < 0.01). Ferritin levels did not predict anemia in either period. Conclusion RET-He and Delta-He are valuable predictors of anemia in critically ill ICU patients, outperforming ferritin in this context. Their routine use could improve the early detection and management of iron deficiency and anemia in ICU settings.
Collapse
Affiliation(s)
| | - Raúl Juárez-Vela
- Faculty of Health Sciences, GRUPAC Research Group, University of La Rioja, Logroño, Spain
- Research Group on Bleeding Patient Management, IDIPAZ Research Institute, Madrid, Spain
| | - Pablo Lasa-Berasain
- Intensive Care Medicine Service, University Hospital of Navarra, Pamplona, Spain
| | | | - Michał Czapla
- Faculty of Health Sciences, GRUPAC Research Group, University of La Rioja, Logroño, Spain
- Department of Emergency Medical Service, Faculty of Nursing and Midwifery, Wroclaw Medical University, Wroclaw, Poland
| | | | | | | | | | | | | | - Manuel Quintana-Díaz
- Research Group on Bleeding Patient Management, IDIPAZ Research Institute, Madrid, Spain
- Intensive Care Medicine Service, La Paz Hospital, Madrid, Spain
| |
Collapse
|
2
|
Gembillo G, Soraci L, Peritore L, Siligato R, Labbozzetta V, Giuffrida AE, Cuzzola F, Spinella C, Romeo A, Calabrese V, Montesanto A, Corsonello A, Santoro D. Impact of SARS-CoV-2 Infection on Erythropoietin Resistance Index in Hemodialysis Patients. Geriatrics (Basel) 2025; 10:33. [PMID: 40126283 PMCID: PMC11932237 DOI: 10.3390/geriatrics10020033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/06/2025] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Background/Objectives: Hemodialysis (HD) patients with advanced chronic kidney disease (CKD) are highly vulnerable to complications from SARS-CoV-2 infection. Anemia management in this population is complex, particularly due to erythropoietin resistance, which may be exacerbated by COVID-19-related inflammation. To this aim, in this small-scale retrospective study, we investigated trends in the erythropoietin resistance index (ERI) over time in patients with and without SARS-CoV-2 infection. Methods: This single-center retrospective study included 25 HD patients, divided into two groups: 15 with a history of SARS-CoV-2 infection (CoV2 group) and 10 without (nonCoV2 group). The ERI was assessed over four visits, with 70-100-day intervals between them. Linear mixed models were used to evaluate factors associated with ERI changes. Results: Patients in the CoV2 group exhibited significantly higher ERI increases between T1 (baseline) and T2 (post-infection) compared to the nonCoV2 group (median ΔERI: +4.65 vs. -0.27, p < 0.001). During the T2-T4 recovery period, CoV2 patients demonstrated a delayed but substantial decline in the ERI, converging to baseline levels by T4. Male sex and hemoglobin levels were negatively associated with the ERI. Conclusions: SARS-CoV-2 infection induces transient but significant erythropoietin resistance in HD patients, likely due to inflammation and disrupted erythropoiesis. Tailored anemia management strategies, including the potential use of hypoxia-inducible factor stabilizers, are warranted. Larger, multicenter studies are needed to validate these findings and improve treatment protocols.
Collapse
Affiliation(s)
- Guido Gembillo
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (L.P.); (V.L.); (A.E.G.); (F.C.); (C.S.); (A.R.); (V.C.)
| | - Luca Soraci
- Unit of Geriatric Medicine, Italian National Research Center on Aging (IRCCS INRCA), 87100 Cosenza, Italy; (L.S.); (A.C.)
| | - Luigi Peritore
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (L.P.); (V.L.); (A.E.G.); (F.C.); (C.S.); (A.R.); (V.C.)
| | - Rossella Siligato
- Nephrology Unit, University Hospital of Ferrara, 44121 Ferrara, Italy;
| | - Vincenzo Labbozzetta
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (L.P.); (V.L.); (A.E.G.); (F.C.); (C.S.); (A.R.); (V.C.)
| | - Alfio Edoardo Giuffrida
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (L.P.); (V.L.); (A.E.G.); (F.C.); (C.S.); (A.R.); (V.C.)
| | - Felicia Cuzzola
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (L.P.); (V.L.); (A.E.G.); (F.C.); (C.S.); (A.R.); (V.C.)
| | - Claudia Spinella
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (L.P.); (V.L.); (A.E.G.); (F.C.); (C.S.); (A.R.); (V.C.)
| | - Adolfo Romeo
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (L.P.); (V.L.); (A.E.G.); (F.C.); (C.S.); (A.R.); (V.C.)
| | - Vincenzo Calabrese
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (L.P.); (V.L.); (A.E.G.); (F.C.); (C.S.); (A.R.); (V.C.)
| | - Alberto Montesanto
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy;
| | - Andrea Corsonello
- Unit of Geriatric Medicine, Italian National Research Center on Aging (IRCCS INRCA), 87100 Cosenza, Italy; (L.S.); (A.C.)
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Domenico Santoro
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (L.P.); (V.L.); (A.E.G.); (F.C.); (C.S.); (A.R.); (V.C.)
| |
Collapse
|
3
|
Li S, Huang S, Feng Y, Mao Y. White blood cell-to-haemoglobin ratio as a predictor of 30-day mortality in ICU patients with pulmonary hypertension: a MIMIC-IV database study. BMC Cardiovasc Disord 2024; 24:729. [PMID: 39707222 PMCID: PMC11660688 DOI: 10.1186/s12872-024-04396-8] [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: 10/26/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND AND OBJECTIVE The white blood cell-to-haemoglobin ratio (WHR) is a comprehensive indicator of inflammation and anaemia status. However, the relationship between the WHR and the risk of 30-day mortality among intensive care unit (ICU) patients with pulmonary hypertension (PH) remains unclear. The purpose of this study was to investigate the association between the WHR and 30-day mortality in critically ill patients with PH. METHODS Clinical data of patients with PH were extracted from the MIMIC-IV (2.2) database. Restricted cubic splines and logistic regression analysis were used to investigate the relationship between the WHR and 30-day mortality. Subgroup analysis was used to assess the robustness of the results. RESULTS A total of 451 patients with PH were enrolled, with 78 (17.3%) dying within 30 days. Restricted cubic spline analysis revealed a linear relationship between the WHR and 30-day mortality. Logistic regression analysis revealed the WHR was an independent predictor for 30-day mortality in critically ill patients with PH (OR, 1.58; 95% CI, 1.05-2.37; P = 0.028). The AUC of the WHR was 0.646 (95% CI: 0.60-0.69). CONCLUSIONS A higher WHR was an independent predictor for 30-day mortality in critically ill patients with PH. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Shuangping Li
- College of Clinical Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China
| | - Shenshen Huang
- College of Clinical Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yuxuan Feng
- College of Clinical Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yimin Mao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, China.
| |
Collapse
|
4
|
Conde Díez S, de Las Cuevas Allende R, Conde García E. Anemia of inflammation and iron metabolism in chronic diseases. Rev Clin Esp 2024; 224:598-608. [PMID: 39236980 DOI: 10.1016/j.rceng.2024.09.002] [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: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 09/07/2024]
Abstract
Anemia of Inflammation begins with the activation of the immune system and the subsequent release of cytokines that lead to an elevation of hepcidin, responsible for hypoferremia, and a suppression of erythropoiesis due to lack of iron. The anemia is usually mild/moderate, normocytic/normochromic and is the most prevalent, after iron deficiency anemia, and is the most common in patients with chronic diseases, in the elderly and in hospitalized patients. Anemia can influence the patient's quality of life and have a negative impact on survival. Treatment should be aimed at improving the underlying disease and correcting the anemia. Intravenous iron, erythropoietin and prolyl hydroxylase inhibitors are the current basis of treatment, but future therapy is directed against hepcidin, which is ultimately responsible for anemia.
Collapse
Affiliation(s)
- S Conde Díez
- Medicina de Familia, Servicio Cántabro de Salud, Centro de Salud Camargo Costa, Maliaño, Cantabria, Spain
| | - R de Las Cuevas Allende
- Medicina de Familia, Servicio Cántabro de Salud, Centro de Salud Altamira, Puente de San Miguel, Cantabria, Spain
| | - E Conde García
- Hematólogo jubilado [jefe del Servicio de Hematología del Hospital Marqués de Valdecilla, Santander. Catedrático de Medicina de la Universidad de Cantabria], Santander, Spain.
| |
Collapse
|
5
|
Wacka E, Nicikowski J, Jarmuzek P, Zembron-Lacny A. Anemia and Its Connections to Inflammation in Older Adults: A Review. J Clin Med 2024; 13:2049. [PMID: 38610814 PMCID: PMC11012269 DOI: 10.3390/jcm13072049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 03/29/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
Anemia is a common hematological disorder that affects 12% of the community-dwelling population, 40% of hospitalized patients, and 47% of nursing home residents. Our understanding of the impact of inflammation on iron metabolism and erythropoiesis is still lacking. In older adults, anemia can be divided into nutritional deficiency anemia, bleeding anemia, and unexplained anemia. The last type of anemia might be caused by reduced erythropoietin (EPO) activity, progressive EPO resistance of bone marrow erythroid progenitors, and the chronic subclinical pro-inflammatory state. Overall, one-third of older patients with anemia demonstrate a nutritional deficiency, one-third have a chronic subclinical pro-inflammatory state and chronic kidney disease, and one-third suffer from anemia of unknown etiology. Understanding anemia's pathophysiology in people aged 65 and over is crucial because it contributes to frailty, falls, cognitive decline, decreased functional ability, and higher mortality risk. Inflammation produces adverse effects on the cells of the hematological system. These effects include iron deficiency (hypoferremia), reduced EPO production, and the elevated phagocytosis of erythrocytes by hepatic and splenic macrophages. Additionally, inflammation causes enhanced eryptosis due to oxidative stress in the circulation. Identifying mechanisms behind age-related inflammation is essential for a better understanding and preventing anemia in older adults.
Collapse
Affiliation(s)
- Eryk Wacka
- Department of Applied and Clinical Physiology, Collegium Medicum University of Zielona Gora, 65-417 Zielona Gora, Poland; (J.N.); (A.Z.-L.)
| | - Jan Nicikowski
- Department of Applied and Clinical Physiology, Collegium Medicum University of Zielona Gora, 65-417 Zielona Gora, Poland; (J.N.); (A.Z.-L.)
| | - Pawel Jarmuzek
- Department of Neurosurgery and Neurology, Collegium Medicum University of Zielona Gora, 65-417 Zielona Gora, Poland;
| | - Agnieszka Zembron-Lacny
- Department of Applied and Clinical Physiology, Collegium Medicum University of Zielona Gora, 65-417 Zielona Gora, Poland; (J.N.); (A.Z.-L.)
| |
Collapse
|
6
|
Hanson AL, Mulè MP, Ruffieux H, Mescia F, Bergamaschi L, Pelly VS, Turner L, Kotagiri P, Göttgens B, Hess C, Gleadall N, Bradley JR, Nathan JA, Lyons PA, Drakesmith H, Smith KGC. Iron dysregulation and inflammatory stress erythropoiesis associates with long-term outcome of COVID-19. Nat Immunol 2024; 25:471-482. [PMID: 38429458 PMCID: PMC10907301 DOI: 10.1038/s41590-024-01754-8] [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: 03/03/2023] [Accepted: 01/12/2024] [Indexed: 03/03/2024]
Abstract
Persistent symptoms following SARS-CoV-2 infection are increasingly reported, although the drivers of post-acute sequelae (PASC) of COVID-19 are unclear. Here we assessed 214 individuals infected with SARS-CoV-2, with varying disease severity, for one year from COVID-19 symptom onset to determine the early correlates of PASC. A multivariate signature detected beyond two weeks of disease, encompassing unresolving inflammation, anemia, low serum iron, altered iron-homeostasis gene expression and emerging stress erythropoiesis; differentiated those who reported PASC months later, irrespective of COVID-19 severity. A whole-blood heme-metabolism signature, enriched in hospitalized patients at month 1-3 post onset, coincided with pronounced iron-deficient reticulocytosis. Lymphopenia and low numbers of dendritic cells persisted in those with PASC, and single-cell analysis reported iron maldistribution, suggesting monocyte iron loading and increased iron demand in proliferating lymphocytes. Thus, defects in iron homeostasis, dysregulated erythropoiesis and immune dysfunction due to COVID-19 possibly contribute to inefficient oxygen transport, inflammatory disequilibrium and persisting symptomatology, and may be therapeutically tractable.
Collapse
Affiliation(s)
- Aimee L Hanson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Matthew P Mulè
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- NIH-Oxford-Cambridge Scholars Program, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Hélène Ruffieux
- MRC Biostatistics Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Federica Mescia
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Laura Bergamaschi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Victoria S Pelly
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Lorinda Turner
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Prasanti Kotagiri
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Berthold Göttgens
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
| | - Christoph Hess
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Nicholas Gleadall
- Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Botnar Research Centre for Child Health (BRCCH), University of Basel and ETH Zurich, Basel, Switzerland
| | - John R Bradley
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - James A Nathan
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Hal Drakesmith
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
7
|
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
|
8
|
Randrianarisoa RMF, Ramanandafy H, Mania A, Monjanel H, Trouillier S. Prevalence and diagnostic performance of iron deficiency in polycythemia. Hematology 2023; 28:2204621. [PMID: 37115586 DOI: 10.1080/16078454.2023.2204621] [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
INTRODUCTION Several observations have shown that patients with polycythemia have iron deficiency. Our objectives were to report the prevalence of iron deficiency and to evaluate the diagnostic performance of serum ferritin in polycythemia vera. PATIENTS AND METHOD This is a retrospective descriptive and analytical study carried out in the internal medicine department of the Henri Mondor Hospital, Aurillac, France. The study involved 114 patients with polycythemia, followed in the department from January 1, 2010 to December 31, 2021. To evaluate the diagnostic performance, the JAK2 mutation was considered as the gold standard of diagnosis. RESULTS Thirty-three patients had polycythemia vera and 76 patients had secondary polycythemia. The mean age of the patients was 61.79 years (±15.44) with a sex ratio of 4.43. The overall prevalence of iron deficiency was 21.05%. The prevalence was 53% in polycythemia vera group and 1.32% in secondary polycythemia group. The risk of iron deficiency was high in polycythemia vera (OR = 115; 95% CI [14.4-918.2], p < 0.0001) and the sensitivity and specificity of serum ferritin were 52.63% and 100% respectively. CONCLUSION Assessment of iron deficiency should be part of the initial evaluation of polycythemia. Iron deficiency had a high specificity during polycythemia vera.
Collapse
Affiliation(s)
| | - Herveat Ramanandafy
- Department of Internal Medicine, Joseph Raseta Befelatanana Hospital, Antananarivo, Madagascar
| | - Alexandre Mania
- Department of Internal Medicine, Henri Mondor Hospital, Aurillac, France
| | - Hélène Monjanel
- Department of Internal Medicine, Henri Mondor Hospital, Aurillac, France
| | | |
Collapse
|
9
|
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
|
10
|
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: 7] [Impact Index Per Article: 3.5] [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
|
11
|
Ganz T, Nemeth E, Rivella S, Goldberg P, Dibble AR, McCaleb ML, Guo S, Monia BP, Barrett TD. TMPRSS6 as a Therapeutic Target for Disorders of Erythropoiesis and Iron Homeostasis. Adv Ther 2023; 40:1317-1333. [PMID: 36690839 PMCID: PMC10070284 DOI: 10.1007/s12325-022-02421-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 12/23/2022] [Indexed: 01/25/2023]
Abstract
TMPRSS6 is a serine protease highly expressed in the liver. Its role in iron regulation was first reported in 2008 when mutations in TMPRSS6 were shown to be the cause of iron-refractory iron deficiency anemia (IRIDA) in humans and in mouse models. TMPRSS6 functions as a negative regulator of the expression of the systemic iron-regulatory hormone hepcidin. Over the last decade and a half, growing understanding of TMPRSS6 biology and mechanism of action has enabled development of new therapeutic approaches for patients with diseases of erythropoiesis and iron homeostasis.ClinicalTrials.gov identifier NCT03165864.
Collapse
Affiliation(s)
- Tomas Ganz
- Department of Medicine and Pathology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
| | - Elizabeta Nemeth
- Department of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology, Children's Hospital of Philadelphia (CHOP), 3401 Civic Center Boulevard, Philadelphia, PA, 19104, USA
- Cell and Molecular Biology Graduate Group (CAMB), University of Pennsylvania, Abramson Research Center, 3615 Civic Center Boulevard, Room 316B, Philadelphia, PA, 19104, USA
| | - Paul Goldberg
- Prilenia Therapeutics, Herzliya, Israel
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | | | | | - Shuling Guo
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | | | | |
Collapse
|
12
|
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
|
13
|
Handa S, Ginzburg Y, Hoffman R, Kremyanskaya M. Hepcidin mimetics in polycythemia vera: resolving the irony of iron deficiency and erythrocytosis. Curr Opin Hematol 2023; 30:45-52. [PMID: 36728649 PMCID: PMC9908837 DOI: 10.1097/moh.0000000000000747] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE OF REVIEW Development of hepcidin therapeutics has been a ground-breaking discovery in restoring iron homeostasis in several haematological disorders. The hepcidin mimetic, rusfertide, is in late-stage clinical development for treating polycythemia vera patients with a global phase 3 trial [NCT05210790] currently underway. Rusfertide serves as the first possible noncytoreductive therapeutic option to maintain haematocrit control and avoid phlebotomy in polycythemia vera patients. In this comprehensive review, we discuss the pathobiology of dysregulated iron metabolism in polycythemia vera, provide the rationale for targeting the hepcidin-ferroportin axis and elaborate on the preclinical and clinical trial evidence supporting the role of hepcidin mimetics in polycythemia vera. RECENT FINDINGS Recently, updated results from two phase 2 clinical trials [NCT04057040 & NCT04767802] of rusfertide (PTG300) demonstrate that the drug is highly effective in eliminating the need for therapeutic phlebotomies, normalizing haematological parameters, repleting iron stores and relieving constitutional symptoms in patients with polycythemia vera. In light of these findings, additional hepcidin mimetic agents are also being evaluated in polycythemia vera patients. SUMMARY Hepcidin agonists essentially serve as a 'chemical phlebotomy' and are poised to vastly improve the quality of life for phlebotomy requiring polycythemia vera patients.
Collapse
Affiliation(s)
- Shivani Handa
- Tisch Cancer Institute, Division of Hematology-Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yelena Ginzburg
- Tisch Cancer Institute, Division of Hematology-Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ronald Hoffman
- Tisch Cancer Institute, Division of Hematology-Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Marina Kremyanskaya
- Tisch Cancer Institute, Division of Hematology-Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
14
|
Tupý J. Anemia of inflammatory: does eiderr knowledge mean better diagnosis and treatment? VNITRNI LEKARSTVI 2023; 69:10-18. [PMID: 37072261 DOI: 10.36290/vnl.2023.023] [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: 04/05/2023]
Abstract
Anemia, which is a manifestation of the deterioration of patients' health and performance, is a common concomitant condition in diseases with signs of inflammation activation. This anemia - anemia of inflammation, is caused by disturbances of iron metabolism that lead to iron retention within macrophages, cytokine mediated inhibition of erythropoietin function and erythroid progenitor cell differentiation, and a reduced erytrocyte half-life. Anemia is usually mild to moderate, normocytic and normochromic. It is characterized by low iron circulation, but normal to increased levels of stored ferritin and the hormone hepcidin. The primary therapeutic approach is the treatment of the underlying inflammatory disease. In case of failure, iron supplementation and / or treatment with erythropoietin stimulating agents may be used. Blood transfusions are just an emergency treatment for life-threatening anemia. A new treatment modalities with hepcidin-modifying strategies and stabilizers of hypoxia inducible factors is emerging. However, their therapeutic efficacy needs to be verified and evaluated in clinical trials.
Collapse
|
15
|
De Souza LV, Hoffmann A, Fischer C, Petzer V, Asshoff M, Theurl I, Tymoszuk P, Seifert M, Brigo N, Hilbe R, Demetz E, Von Raffay L, Berger S, Barros-Pinkelnig M, Weiss G. Comparative analysis of oral and intravenous iron therapy in rat models of inflammatory anemia and iron deficiency. Haematologica 2023; 108:135-149. [PMID: 35796011 PMCID: PMC9827174 DOI: 10.3324/haematol.2022.281149] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/29/2022] [Indexed: 02/05/2023] Open
Abstract
Anemia is a major health issue and associated with increased morbidity. Iron deficiency anemia (IDA) is the most prevalent, followed by anemia of chronic disease (ACD). IDA and ACD often co-exist, challenging diagnosis and treatment. While iron supplementation is the first-line therapy for IDA, its optimal route of administration and the efficacy of different repletion strategies in ACD are elusive. Female Lewis rats were injected with group A streptococcal peptidoglycan-polysaccharide (PG-APS) to induce inflammatory arthritis with associated ACD and/or repeatedly phlebotomized and fed with a low iron diet to induce IDA, or a combination thereof (ACD/IDA). Iron was either supplemented by daily oral gavage of ferric maltol or by weekly intravenous (i.v.) injection of ferric carboxymaltose for up to 4 weeks. While both strategies reversed IDA, they remained ineffective to improve hemoglobin (Hb) levels in ACD, although oral iron showed slight amelioration of various erythropoiesis-associated parameters. In contrast, both iron treatments significantly increased Hb in ACD/IDA. In ACD and ACD/IDA animals, i.v. iron administration resulted in iron trapping in liver and splenic macrophages, induction of ferritin expression and increased circulating levels of the iron hormone hepcidin and the inflammatory cytokine interleukin-6, while oral iron supplementation reduced interleukin-6 levels. Thus, oral and i.v. iron resulted in divergent effects on systemic and tissue iron homeostasis and inflammation. Our results indicate that both iron supplements improve Hb in ACD/IDA, but are ineffective in ACD with pronounced inflammation, and that under the latter condition, i.v. iron is trapped in macrophages and may enhance inflammation.
Collapse
Affiliation(s)
- Lara Valente De Souza
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia research, Medical University of Innsbruck
| | - Alexander Hoffmann
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia research, Medical University of Innsbruck
| | - Christine Fischer
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Verena Petzer
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Malte Asshoff
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Igor Theurl
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Markus Seifert
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia research, Medical University of Innsbruck
| | - Natascha Brigo
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Richard Hilbe
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Egon Demetz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Laura Von Raffay
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Sylvia Berger
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Marina Barros-Pinkelnig
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck
| | - Guenter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Austria; Christian Doppler Laboratory for Iron Metabolism and Anemia research, Medical University of Innsbruck.
| |
Collapse
|
16
|
White Z, Elagib KE, Gru AA, Goldfarb AN. Liver kinase B1 (LKB1) in murine erythroid progenitors modulates erythropoietin setpoint in association with maturation control. Blood Cells Mol Dis 2022; 97:102688. [PMID: 35717902 PMCID: PMC9436178 DOI: 10.1016/j.bcmd.2022.102688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/18/2022]
Abstract
Erythropoiesis is a tightly regulated process. It is stimulated by decreased oxygen in circulation, which leads to the secretion of the hormone erythropoietin (Epo) by the kidneys. An additional layer of control involves the coordinated sensing and use of nutrients. Much cellular machinery contributes to sensing and responding to nutrient status in cells, and one key participant is the kinase LKB1. The current study examines the role of LKB1 in erythropoiesis using a murine in vivo and ex vivo conditional knockout system. In vivo analysis showed erythroid loss of LKB1 to be associated with a robust increase in serum Epo and mild reticulocytosis. Despite these abnormalities, no evidence of anemia or hemolysis was found. Further characterization using an ex vivo progenitor culture assay demonstrated accelerated erythroid maturation in the LKB1-deficient cells. Based on pharmacologic evidence, this phenotype appeared to result from impaired AMP-activated protein kinase (AMPK) signaling downstream of LKB1. These findings reveal a role for LKB1 in fine-tuning Epo-driven erythropoiesis in association with maturational control.
Collapse
Affiliation(s)
- Zollie White
- Department of Pathology, University of Virginia School of Medicine, USA
| | | | - Alejandro A Gru
- Department of Pathology, University of Virginia School of Medicine, USA
| | - Adam N Goldfarb
- Department of Pathology, University of Virginia School of Medicine, USA.
| |
Collapse
|
17
|
Khairnar RC, Parihar N, Prabhavalkar KS, Bhatt LK. Emerging targets signaling for inflammation in Parkinson's disease drug discovery. Metab Brain Dis 2022; 37:2143-2161. [PMID: 35536461 DOI: 10.1007/s11011-022-00999-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/29/2022] [Indexed: 10/18/2022]
Abstract
Parkinson's disease (PD) patients not only show motor features such as bradykinesia, tremor, and rigidity but also non-motor features such as anxiety, depression, psychosis, memory loss, attention deficits, fatigue, sexual dysfunction, gastrointestinal issues, and pain. Many pharmacological treatments are available for PD patients; however, these treatments are partially or transiently effective since they only decrease the symptoms. As these therapies are unable to restore dopaminergic neurons and stop the development of Parkinson's disease, therefore, the need for an effective therapeutic approach is required. The current review summarizes novel targets for PD, that can be utilized to identify disease-modifying treatments.
Collapse
Affiliation(s)
- Rhema Chandan Khairnar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Niraj Parihar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Kedar S Prabhavalkar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (West), Mumbai, 400056, India.
| |
Collapse
|
18
|
Li Y, Miller I, Prasad P, George NA, Parrow NL, Fleming RE. Effects of Exogenous Transferrin on the Regulation of Iron Metabolism and Erythropoiesis in Iron Deficiency With or Without Anemia. Front Physiol 2022; 13:893149. [PMID: 35634155 PMCID: PMC9132588 DOI: 10.3389/fphys.2022.893149] [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: 03/10/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Erythropoietic response is controlled not only by erythropoietin but also by iron. In addition to its role in iron delivery, transferrin also functions as a signaling molecule, with effects on both iron homeostasis and erythropoiesis. We investigated hematologic parameters, iron status and expression of key proteins, including the hepatic iron regulatory protein hepcidin and the suppressive erythroid factor Erfe, in mice subject to dietary iron deficiency with and without anemia. The acute effect of iron on these parameters was investigated by administration of exogenous iron-loaded transferrin (holoTf) in each of the mouse models. Serum iron in mice with iron deficiency (ID) is modestly lower with hematologic parameters maintained by utilization of iron stores in mice with ID. As expected, erythropoietin expression and concentration, along with marrow Erfe are unaffected in ID mice. Administration of holoTf restores serum iron and Tf saturation levels to those observed in control mice and results in an increase in hepcidin compared to ID mice not treated with holoTf. The expression of the Bmp signaling molecule Bmp6 is not significantly increased following Tf treatment in ID mice. Thus, the expression level of the gene encoding hepcidin, Hamp1, is increased relative to Bmp6 expression in ID mice following treatment with holoTf, leading us to speculate that Tf saturation may influence Bmp sensitivity. In mice with iron deficiency anemia (IDA), decreased hematologic parameters were accompanied by pronounced decreases in serum and tissue iron concentrations, and an increase in serum erythropoietin. In the absence of exogenous holoTf, the greater serum erythropoietin was not reflected by an increase in marrow Erfe expression. HoloTf administration did not acutely change serum Epo in IDA mice. Marrow Erfe expression was, however, markedly increased in IDA mice following holoTf, plausibly accounting for the lack of an increase in Hamp1 following holoTf treatment in the IDA mice. The increase in Erfe despite no change in erythropoietin suggests that Tf acts to increase erythropoietin sensitivity. These observations underscore the importance of Tf in modulating the erythropoietic response in recovery from iron deficiency anemia, with implications for other stress erythropoiesis conditions.
Collapse
Affiliation(s)
- Yihang Li
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Ian Miller
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Princy Prasad
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Nisha Ajit George
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Nermi L. Parrow
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Robert E. Fleming
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO, United States
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
19
|
Correnti M, Gammella E, Cairo G, Recalcati S. Iron Mining for Erythropoiesis. Int J Mol Sci 2022; 23:ijms23105341. [PMID: 35628152 PMCID: PMC9140467 DOI: 10.3390/ijms23105341] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 01/27/2023] Open
Abstract
Iron is necessary for essential processes in every cell of the body, but the erythropoietic compartment is a privileged iron consumer. In fact, as a necessary component of hemoglobin and myoglobin, iron assures oxygen distribution; therefore, a considerable amount of iron is required daily for hemoglobin synthesis and erythroid cell proliferation. Therefore, a tight link exists between iron metabolism and erythropoiesis. The liver-derived hormone hepcidin, which controls iron homeostasis via its interaction with the iron exporter ferroportin, coordinates erythropoietic activity and iron homeostasis. When erythropoiesis is enhanced, iron availability to the erythron is mainly ensured by inhibiting hepcidin expression, thereby increasing ferroportin-mediated iron export from both duodenal absorptive cells and reticuloendothelial cells that process old and/or damaged red blood cells. Erythroferrone, a factor produced and secreted by erythroid precursors in response to erythropoietin, has been identified and characterized as a suppressor of hepcidin synthesis to allow iron mobilization and facilitate erythropoiesis.
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW HRI is the heme-regulated elF2α kinase that phosphorylates the α-subunit of elF2. Although the role of HRI in inhibiting globin synthesis in erythroid cells is well established, broader roles of HRI in translation have been uncovered recently. This review is to summarize the new discoveries of HRI in stress erythropoiesis and in fetal γ-globin expression. RECENT FINDINGS HRI and activating transcription factor 4 (ATF4) mRNAs are highly expressed in early erythroblasts. Inhibition of protein synthesis by HRI-phosphorylated elF2α (elF2αP) is necessary to maintain protein homeostasis in both the cytoplasm and mitochondria. In addition, HRI-elF2αP specifically enhances translation of ATF4 mRNA leading to the repression of mechanistic target of rapamycin complex 1 (mTORC1) signaling. ATF4-target genes are most highly activated during iron deficiency to maintain mitochondrial function, redox homeostasis, and to enable erythroid differentiation. HRI is therefore a master translation regulator of erythropoiesis sensing intracellular heme concentrations and oxidative stress for effective erythropoiesis. Intriguingly, HRI-elF2αP-ATF4 signaling also inhibits fetal hemoglobin production in human erythroid cells. SUMMARY The primary function of HRI is to maintain protein homeostasis accompanied by the induction of ATF4 to mitigate stress. Role of HRI-ATF4 in γ-globin expression raises the potential of HRI as a therapeutic target for hemoglobinopathy.
Collapse
Affiliation(s)
- Jane-Jane Chen
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shuping Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| |
Collapse
|
21
|
Lanser L, Fuchs D, Kurz K, Weiss G. Physiology and Inflammation Driven Pathophysiology of Iron Homeostasis-Mechanistic Insights into Anemia of Inflammation and Its Treatment. Nutrients 2021; 13:3732. [PMID: 34835988 PMCID: PMC8619077 DOI: 10.3390/nu13113732] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 02/07/2023] Open
Abstract
Anemia is very common in patients with inflammatory disorders. Its prevalence is associated with severity of the underlying disease, and it negatively affects quality of life and cardio-vascular performance of patients. Anemia of inflammation (AI) is caused by disturbances of iron metabolism resulting in iron retention within macrophages, a reduced erythrocyte half-life, and cytokine mediated inhibition of erythropoietin function and erythroid progenitor cell differentiation. AI is mostly mild to moderate, normochromic and normocytic, and characterized by low circulating iron, but normal and increased levels of the storage protein ferritin and the iron hormone hepcidin. The primary therapeutic approach for AI is treatment of the underlying inflammatory disease which mostly results in normalization of hemoglobin levels over time unless other pathologies such as vitamin deficiencies, true iron deficiency on the basis of bleeding episodes, or renal insufficiency are present. If the underlying disease and/or anemia are not resolved, iron supplementation therapy and/or treatment with erythropoietin stimulating agents may be considered whereas blood transfusions are an emergency treatment for life-threatening anemia. New treatments with hepcidin-modifying strategies and stabilizers of hypoxia inducible factors emerge but their therapeutic efficacy for treatment of AI in ill patients needs to be evaluated in clinical trials.
Collapse
Affiliation(s)
- Lukas Lanser
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.L.); (K.K.)
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Katharina Kurz
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.L.); (K.K.)
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria; (L.L.); (K.K.)
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
22
|
Sinha S, Pereira-Reis J, Guerra A, Rivella S, Duarte D. The Role of Iron in Benign and Malignant Hematopoiesis. Antioxid Redox Signal 2021; 35:415-432. [PMID: 33231101 PMCID: PMC8328043 DOI: 10.1089/ars.2020.8155] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/26/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022]
Abstract
Significance: Iron is an essential element required for sustaining a normal healthy life. However, an excess amount of iron in the bloodstream and tissue generates toxic hydroxyl radicals through Fenton reactions. Henceforth, a balance in iron concentration is extremely important to maintain cellular homeostasis in both normal hematopoiesis and erythropoiesis. Iron deficiency or iron overload can impact hematopoiesis and is associated with many hematological diseases. Recent Advances: The mechanisms of action of key iron regulators such as erythroferrone and the discovery of new drugs, such as ACE-536/luspatercept, are of potential interest to treat hematological disorders, such as β-thalassemia. New therapies targeting inflammation-induced ineffective erythropoiesis are also in progress. Furthermore, emerging evidences support differential interactions between iron and its cellular antioxidant responses of hematopoietic and neighboring stromal cells. Both iron and its systemic regulator, such as hepcidin, play a significant role in regulating erythropoiesis. Critical Issues: Significant pre-clinical studies are on the way and new drugs targeting iron metabolism have been recently approved or are undergoing clinical trials to treat pathological conditions with impaired erythropoiesis such as myelodysplastic syndromes or β-thalassemia. Future Directions: Future studies should explore how iron regulates hematopoiesis in both benign and malignant conditions. Antioxid. Redox Signal. 35, 415-432.
Collapse
Affiliation(s)
- Sayantani Sinha
- Division of Hematology, Department of Pediatrics, The Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
| | - Joana Pereira-Reis
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Amaliris Guerra
- Division of Hematology, Department of Pediatrics, The Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
| | - Stefano Rivella
- Division of Hematology, Department of Pediatrics, The Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Cell and Molecular Biology Affinity Group (CAMB), University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
- Penn Center for Musculoskeletal Disorders, The Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
| | - Delfim Duarte
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Department of Onco-Hematology, Instituto Português de Oncologia (IPO), Porto, Portugal
- Unit of Biochemistry, Department of Biomedicine, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
- Porto Comprehensive Cancer Center (P.CCC), Porto, Portugal
| |
Collapse
|
23
|
Porter J, Taher A, Viprakasit V, Kattamis A, Coates TD, Garbowski M, Dürrenberger F, Manolova V, Richard F, Cappellini MD. Oral ferroportin inhibitor vamifeport for improving iron homeostasis and erythropoiesis in β-thalassemia: current evidence and future clinical development. Expert Rev Hematol 2021; 14:633-644. [PMID: 34324404 DOI: 10.1080/17474086.2021.1935854] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
INTRODUCTION In β-thalassemia, imbalanced globin synthesis causes reduced red blood cell survival and ineffective erythropoiesis. Suppressed hepcidin levels increase ferroportin-mediated iron transport in enterocytes, causing increased iron absorption and potentially iron overload. Low hepcidin also stimulates ferroportin-mediated iron release from macrophages, increasing transferrin saturation (TSAT), potentially forming non-transferrin-bound iron, which can be toxic. Modulating the hepcidin-ferroportin axis is an attractive strategy to improve ineffective erythropoiesis and limit the potential tissue damage resulting from iron overload. There are no oral β-thalassemia treatments that consistently ameliorate anemia and prevent iron overload. AREAS COVERED The preclinical and clinical development of vamifeport (VIT-2763), a novel ferroportin inhibitor, was reviewed. PubMed, EMBASE and ClinicalTrials.gov were searched using the search term 'VIT-2763'. EXPERT OPINION Vamifeport is the first oral ferroportin inhibitor in clinical development. In healthy volunteers, vamifeport had comparable safety to placebo, was well tolerated and rapidly decreased iron levels and reduced TSAT, consistent with observations in preclinical models. Data from ongoing/planned Phase II studies are critical to define its potential in β-thalassemia and other conditions associated with iron overabsorption and/or ineffective erythropoiesis. If vamifeport potentially increases hemoglobin and reduces iron-related parameters, it could be a suitable treatment for non-transfusion-dependent and transfusion-dependent β-thalassemia.
Collapse
Affiliation(s)
- John Porter
- Professor of Haematology, Department of Haematology, University College London, Consultant in Haematology, University College London Hospitals and Head of Joint UCLH and Whittington Hospital Red Cell Unit, London, UK
| | - Ali Taher
- Professor of Medicine, Hematology and Oncology, Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Vip Viprakasit
- Professor of Pediatrics, Director, Thalassemia Research Program, Director, SiCORE in Advanced Cell & Gene Therapy Center (SiCORE-ACGT), Division of Hematology and Oncology, Department of Pediatrics & Siriraj Thalassemia Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Antonis Kattamis
- Professor of Pediatric Hematology-Oncology, Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Thomas D Coates
- Section Head, Hematology, Cancer and Blood Disease Institute, Professor of Pediatrics and Pathology, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Maciej Garbowski
- Clinical Research Fellow, Department of Haematology, University College London Cancer Institute, London, UK
| | - Franz Dürrenberger
- Head of Chemical and Preclinical R&D, Vifor (International) AG, Chemical and Preclinical Research and Development, St. Gallen, Switzerland
| | - Vania Manolova
- Head of Biology R&D, Vifor (International) AG, Chemical and Preclinical Research and Development, St. Gallen, Switzerland
| | - Frank Richard
- Clinical Research Director, Vifor Pharma AG, Glattbrugg, Switzerland
| | - M Domenica Cappellini
- Professor of Internal Medicine, Department of Clinical Sciences and Community, University of Milan, Milan, Italy
| |
Collapse
|
24
|
The critical roles of iron during the journey from fetus to adolescent: Developmental aspects of iron homeostasis. Blood Rev 2021; 50:100866. [PMID: 34284901 DOI: 10.1016/j.blre.2021.100866] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/12/2022]
Abstract
Iron is indispensable for human life. However, it is also potentially toxic, since it catalyzes the formation of harmful oxidative radicals in unbound form and may facilitate pathogen growth. Therefore, iron homeostasis needs to be tightly regulated. Rapid growth and development require large amounts of iron, while (especially young) children are vulnerable to infections with iron-dependent pathogens due to an immature immune system. Moreover, unbalanced iron status early in life may have effects on the nervous system, immune system and gut microbiota that persist into adulthood. In this narrative review, we assess the critical roles of iron for growth and development and elaborate how the body adapts to physiologically high iron demands during the journey from fetus to adolescent. As a first step towards the development of clinical guidelines for the management of iron disorders in children, we summarize the unmet needs regarding the developmental aspects of iron homeostasis.
Collapse
|
25
|
Grzywa TM, Nowis D, Golab J. The role of CD71 + erythroid cells in the regulation of the immune response. Pharmacol Ther 2021; 228:107927. [PMID: 34171326 DOI: 10.1016/j.pharmthera.2021.107927] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023]
Abstract
Complex regulation of the immune response is necessary to support effective defense of an organism against hostile invaders and to maintain tolerance to harmless microorganisms and autoantigens. Recent studies revealed previously unappreciated roles of CD71+ erythroid cells (CECs) in regulation of the immune response. CECs physiologically reside in the bone marrow where erythropoiesis takes place. Under stress conditions, CECs are enriched in some organs outside of the bone marrow as a result of extramedullary erythropoiesis. However, the role of CECs goes well beyond the production of erythrocytes. In neonates, increased numbers of CECs contribute to their vulnerability to infectious diseases. On the other side, neonatal CECs suppress activation of immune cells in response to abrupt colonization with commensal microorganisms after delivery. CECs are also enriched in the peripheral blood of pregnant women as well as in the placenta and are responsible for the regulation of feto-maternal tolerance. In patients with cancer, anemia leads to increased frequency of CECs in the peripheral blood contributing to diminished antiviral and antibacterial immunity, as well as to accelerated cancer progression. Moreover, recent studies revealed the role of CECs in HIV and SARS-CoV-2 infections. CECs use a full arsenal of mechanisms to regulate immune response. These cells suppress proinflammatory responses of myeloid cells and T-cell proliferation by the depletion of ʟ-arginine by arginase. Moreover, CECs produce reactive oxygen species to decrease T-cell proliferation. CECs also secrete cytokines, including transforming growth factor β (TGF-β), which promotes T-cell differentiation into regulatory T-cells. Here, we comprehensively describe the role of CECs in orchestrating immune response and indicate some therapeutic approaches that might be used to regulate their effector functions in the treatment of human conditions.
Collapse
Affiliation(s)
- Tomasz M Grzywa
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland; Doctoral School, Medical University of Warsaw, Zwirki and Wigury 61 Street, 02-091 Warsaw, Poland; Laboratory of Experimental Medicine, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland.
| | - Dominika Nowis
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland; Laboratory of Experimental Medicine, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland.
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland; Centre of Preclinical Research, Medical University of Warsaw, Banacha 1b Street, 02-097 Warsaw, Poland.
| |
Collapse
|
26
|
Silvestri L, Nai A. Iron and erythropoiesis: A mutual alliance. Semin Hematol 2021; 58:145-152. [PMID: 34389106 DOI: 10.1053/j.seminhematol.2021.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/26/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023]
Abstract
The large amount of iron required for hemoglobin synthesis keeps iron homeostasis and erythropoiesis inter-connected, both iron levels being affected by increased erythropoiesis, and erythropoiesis regulated by serum iron. The connection between these 2 processes is maintained even when erythropoiesis is ineffective. In the last years great advances in the understanding of the mechanisms of this crosstalk have been achieved thanks to the discovery of 2 essential players: hepcidin, the master regulator of iron homeostasis, and erythroferrone, the long sought erythroid regulator. In addition, how circulating transferrin-bound iron contributes to the crosstalk between the 2 systems has started to be unraveled.
Collapse
Affiliation(s)
- Laura Silvestri
- Regulation of Iron Metabolism Unit-Div. Genetics & Cell Biology-IRCCS San Raffaele Scientific Institute, Milano, Italy; San Raffaele Vita-Salute University, Milano, Italy.
| | - Antonella Nai
- Regulation of Iron Metabolism Unit-Div. Genetics & Cell Biology-IRCCS San Raffaele Scientific Institute, Milano, Italy; San Raffaele Vita-Salute University, Milano, Italy
| |
Collapse
|
27
|
Iron homeostasis during anemia of inflammation: a prospective study in patients with tuberculosis. Blood 2021; 138:1293-1303. [PMID: 33876222 DOI: 10.1182/blood.2020010562] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 04/02/2021] [Indexed: 11/20/2022] Open
Abstract
Anemia of inflammation is a hallmark of tuberculosis. Factors controlling iron metabolism during anemia of inflammation and its resolution are uncertain. Whether iron supplements should be given during anti-tuberculosis treatment to support Hb recovery is unclear. Before and during treatment of tuberculosis, we assessed iron kinetics, and changes in inflammation and iron metabolism indices. In a 26-wk prospective study, Tanzanian adults with tuberculosis (n=18) were studied before treatment and then every two weeks during treatment; oral and intravenous iron tracers were administered before treatment, after intensive phase (8/12 wk) and complete treatment (24 wk); no iron supplements were given. Before treatment, hepcidin and erythroferrone (ERFE) were greatly elevated, erythrocyte iron utilization was high (~80%) and iron absorption was negligible (<1%). During treatment, hepcidin and IL-6 decreased ~70% after only 2 wk (p<0.001); in contrast, ERFE did not significantly decrease until 8 wk (p<0.01). ERFE and IL-6 were the main opposing determinants of hepcidin (p<0.05) and greater ERFE was associated with reticulocytosis and hemoglobin (Hb) repletion (p<0.01). Dilution of baseline tracer concentration was 2.6-fold higher during intensive phase treatment (p<0.01) indicating enhanced erythropoiesis. After treatment completion, iron absorption increased ~20-fold (p<0.001); Hb increased ~25% (p<0.001). In tuberculosis-associated anemia of inflammation, our findings suggest elevated ERFE is unable to suppress hepcidin and iron absorption is negligible. During treatment, as inflammation resolves, ERFE may remain elevated, contributing to hepcidin suppression and Hb repletion. Iron is well-absorbed only after tuberculosis treatment and supplementation should be reserved for patients remaining anemic after treatment. (ClinicalTrials.gov Identifier:NCT02176772).
Collapse
|
28
|
Richard C, Viret S, Cantero Aguilar L, Lefevre C, Leduc M, Faouzi EH, Azar N, Lavazec C, Mayeux P, Verdier F. Myotonic dystrophy kinase-related CDC42-binding kinase α, a new transferrin receptor type 2-binding partner, is a regulator of erythropoiesis. Am J Hematol 2021; 96:480-492. [PMID: 33476437 DOI: 10.1002/ajh.26104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 01/01/2023]
Abstract
Efficient erythropoiesis relies on the expression of the transferrin receptor type 2 (TFR2). In erythroid precursors, TFR2 facilitates the export of the erythropoietin receptor (EPOR) to cell surface, which ensures the survival and proliferation of erythroblasts. Although TFR2 has a crucial role in erythropoiesis regulation, its mechanism of action remains to be clarified. To understand its role better, we aimed at identifying its protein partners by mass-spectrometry after immunoprecipitation in erythroid cells. Here we report the kinase MRCKα (myotonic dystrophy kinase-related CDC42-binding kinase α) as a new partner of both TFR2 and EPOR in erythroblasts. We show that MRCKα is co-expressed with TFR2, and TFR1 during terminal differentiation and regulates the internalization of the two types of transferrin receptors. The knockdown of MRCKα by shRNA in human primary erythroblasts leads to a decreased cell surface expression of both TFR1 and TFR2, an increased cell-surface expression of EPOR, and a delayed differentiation. Additionally, knockout of Mrckα in the murine MEDEP cells also leads to a striking delay in erythropoiesis, showcasing the importance of this kinase in both species. Our data highlight the importance of MRCKα in the regulation of erythropoiesis.
Collapse
Affiliation(s)
- Cyrielle Richard
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Sophie Viret
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Lilia Cantero Aguilar
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Carine Lefevre
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Marjorie Leduc
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
- Plateforme Protéomique 3P5‐Proteom'IC, Université de Paris, Institut Cochin, INSERM, U1016, CNRS UMR8104 Paris France
| | - El Hassan Faouzi
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Nabih Azar
- Unité d'Hémobiothérapie, Hôpital La Pitié Salpêtrière Paris France
| | - Catherine Lavazec
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Patrick Mayeux
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
- Plateforme Protéomique 3P5‐Proteom'IC, Université de Paris, Institut Cochin, INSERM, U1016, CNRS UMR8104 Paris France
| | - Frédérique Verdier
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| |
Collapse
|
29
|
Tackling the unknowns in understanding and management of hospital acquired anemia. Blood Rev 2021; 49:100830. [PMID: 33810899 DOI: 10.1016/j.blre.2021.100830] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/21/2021] [Accepted: 03/21/2021] [Indexed: 01/29/2023]
Abstract
Hospital acquired anemia (HAA) has been a recognized entity for nearly 50 years. Despite multiple hypotheses, a mechanistic understanding is lacking, and targeted interventions have not yet yielded significantly impactful results. Known risk factors include advanced age, multiple co-morbidities, low bone marrow reserve, admission to the intensive care unit, and frequent phlebotomy. However, confounding variables in many studies continues to complicate the identification of additional risk factors. Improved understanding of iron metabolism, erythropoiesis, and the erythroid iron restriction response in the last few decades, as well as the recent demonstration of poor outcomes correlating with increased transfusion have refocused attention on HAA. While retrospective database studies provide ample correlative data between 1) HAA and poor outcomes; 2) reduction of phlebotomy volume and decrease in transfusion requirement; and 3) over-transfusion and increased mortality, no causal link between reduced phlebotomy volume, decreased rates of HAA, and improved mortality or other relevant outcomes have been definitely established. Here, we review the current state of knowledge and provide a summary of potential directions to understand and mitigate HAA. There are at present no clear guidelines on whether and when to evaluate hospitalized patients for underlying causes of anemia. We thus provide a guide for clinicians in general practice toward identifying patients at the highest risk for HAA, decreasing blood loss through phlebotomy to the greatest degree feasible, and evaluating and treating reversible causes of anemia in a targeted population.
Collapse
|
30
|
Correcting β-thalassemia by combined therapies that restrict iron and modulate erythropoietin activity. Blood 2021; 136:1968-1979. [PMID: 32556142 DOI: 10.1182/blood.2019004719] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 05/21/2020] [Indexed: 11/20/2022] Open
Abstract
β-Thalassemia intermedia is a disorder characterized by ineffective erythropoiesis (IE), anemia, splenomegaly, and systemic iron overload. Novel approaches are being explored based on the modulation of pathways that reduce iron absorption (ie, using hepcidin activators like Tmprss6-antisense oligonucleotides [ASOs]) or increase erythropoiesis (by erythropoietin [EPO] administration or modulating the ability of transferrin receptor 2 [Tfr2] to control red blood cell [RBC] synthesis). Targeting Tmprss6 messenger RNA by Tmprss6-ASO was proven to be effective in improving IE and splenomegaly by inducing iron restriction. However, we postulated that combinatorial strategies might be superior to single therapies. Here, we combined Tmprss6-ASO with EPO administration or removal of a single Tfr2 allele in the bone marrow of animals affected by β-thalassemia intermedia (Hbbth3/+). EPO administration alone or removal of a single Tfr2 allele increased hemoglobin levels and RBCs. However, EPO or Tfr2 single-allele deletion alone, respectively, exacerbated or did not improve splenomegaly in β-thalassemic mice. To overcome this issue, we postulated that some level of iron restriction (by targeting Tmprss6) would improve splenomegaly while preserving the beneficial effects on RBC production mediated by EPO or Tfr2 deletion. While administration of Tmprss6-ASO alone improved the anemia, the combination of Tmprss6-ASO + EPO or Tmprss6-ASO + Tfr2 single-allele deletion produced significantly higher hemoglobin levels and reduced splenomegaly. In conclusion, our results clearly indicate that these combinatorial approaches are superior to single treatments in ameliorating IE and anemia in β-thalassemia and could provide guidance to translate some of these approaches into viable therapies.
Collapse
|
31
|
A fully human anti-BMP6 antibody reduces the need for erythropoietin in rodent models of the anemia of chronic disease. Blood 2021; 136:1080-1090. [PMID: 32438400 DOI: 10.1182/blood.2019004653] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/28/2020] [Indexed: 12/16/2022] Open
Abstract
Recombinant erythropoietin (EPO) and iron substitution are a standard of care for treatment of anemias associated with chronic inflammation, including anemia of chronic kidney disease. A black box warning for EPO therapy and concerns about negative side effects related to high-dose iron supplementation as well as the significant proportion of patients becoming EPO resistant over time explains the medical need to define novel strategies to ameliorate anemia of chronic disease (ACD). As hepcidin is central to the iron-restrictive phenotype in ACD, therapeutic approaches targeting hepcidin were recently developed. We herein report the therapeutic effects of a fully human anti-BMP6 antibody (KY1070) either as monotherapy or in combination with Darbepoetin alfa on iron metabolism and anemia resolution in 2 different, well-established, and clinically relevant rodent models of ACD. In addition to counteracting hepcidin-driven iron limitation for erythropoiesis, we found that the combination of KY1070 and recombinant human EPO improved the erythroid response compared with either monotherapy in a qualitative and quantitative manner. Consequently, the combination of KY1070 and Darbepoetin alfa resulted in an EPO-sparing effect. Moreover, we found that suppression of hepcidin via KY1070 modulates ferroportin expression on erythroid precursor cells, thereby lowering potentially toxic-free intracellular iron levels and by accelerating erythroid output as reflected by increased maturation of erythrocyte progenitors. In summary, we conclude that treatment of ACD, as a highly complex disease, becomes more effective by a multifactorial therapeutic approach upon mobilization of endogenous iron deposits and stimulation of erythropoiesis.
Collapse
|
32
|
Iron control of erythroid microtubule cytoskeleton as a potential target in treatment of iron-restricted anemia. Nat Commun 2021; 12:1645. [PMID: 33712594 PMCID: PMC7955080 DOI: 10.1038/s41467-021-21938-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 02/20/2021] [Indexed: 12/17/2022] Open
Abstract
Anemias of chronic disease and inflammation (ACDI) result from restricted iron delivery to erythroid progenitors. The current studies reveal an organellar response in erythroid iron restriction consisting of disassembly of the microtubule cytoskeleton and associated Golgi disruption. Isocitrate supplementation, known to abrogate the erythroid iron restriction response, induces reassembly of microtubules and Golgi in iron deprived progenitors. Ferritin, based on proteomic profiles, regulation by iron and isocitrate, and putative interaction with microtubules, is assessed as a candidate mediator. Knockdown of ferritin heavy chain (FTH1) in iron replete progenitors induces microtubule collapse and erythropoietic blockade; conversely, enforced ferritin expression rescues erythroid differentiation under conditions of iron restriction. Fumarate, a known ferritin inducer, synergizes with isocitrate in reversing molecular and cellular defects of iron restriction and in oral remediation of murine anemia. These findings identify a cytoskeletal component of erythroid iron restriction and demonstrate potential for its therapeutic targeting in ACDI. Debilitating anemias in chronic diseases can result from deficient iron delivery to red cell precursors. Here, the authors show how this deficiency damages the cytoskeletal framework of progenitor cells and identify a targeted strategy for cytoskeletal repair, leading to anemia correction.
Collapse
|
33
|
Grzywa TM, Justyniarska M, Nowis D, Golab J. Tumor Immune Evasion Induced by Dysregulation of Erythroid Progenitor Cells Development. Cancers (Basel) 2021; 13:870. [PMID: 33669537 PMCID: PMC7922079 DOI: 10.3390/cancers13040870] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer cells harness normal cells to facilitate tumor growth and metastasis. Within this complex network of interactions, the establishment and maintenance of immune evasion mechanisms are crucial for cancer progression. The escape from the immune surveillance results from multiple independent mechanisms. Recent studies revealed that besides well-described myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs) or regulatory T-cells (Tregs), erythroid progenitor cells (EPCs) play an important role in the regulation of immune response and tumor progression. EPCs are immature erythroid cells that differentiate into oxygen-transporting red blood cells. They expand in the extramedullary sites, including the spleen, as well as infiltrate tumors. EPCs in cancer produce reactive oxygen species (ROS), transforming growth factor β (TGF-β), interleukin-10 (IL-10) and express programmed death-ligand 1 (PD-L1) and potently suppress T-cells. Thus, EPCs regulate antitumor, antiviral, and antimicrobial immunity, leading to immune suppression. Moreover, EPCs promote tumor growth by the secretion of growth factors, including artemin. The expansion of EPCs in cancer is an effect of the dysregulation of erythropoiesis, leading to the differentiation arrest and enrichment of early-stage EPCs. Therefore, anemia treatment, targeting ineffective erythropoiesis, and the promotion of EPC differentiation are promising strategies to reduce cancer-induced immunosuppression and the tumor-promoting effects of EPCs.
Collapse
Affiliation(s)
- Tomasz M. Grzywa
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (T.M.G.); (M.J.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
- Laboratory of Experimental Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Magdalena Justyniarska
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (T.M.G.); (M.J.)
| | - Dominika Nowis
- Laboratory of Experimental Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (T.M.G.); (M.J.)
| |
Collapse
|
34
|
Mleczko‐Sanecka K, Silvestri L. Cell-type-specific insights into iron regulatory processes. Am J Hematol 2021; 96:110-127. [PMID: 32945012 DOI: 10.1002/ajh.26001] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/20/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022]
Abstract
Despite its essential role in many biological processes, iron is toxic when in excess due to its propensity to generate reactive oxygen species. To prevent diseases associated with iron deficiency or iron loading, iron homeostasis must be tightly controlled. Intracellular iron content is regulated by the Iron Regulatory Element-Iron Regulatory Protein (IRE-IRP) system, whereas systemic iron availability is adjusted to body iron needs chiefly by the hepcidin-ferroportin (FPN) axis. Here, we aimed to review advances in the field that shed light on cell-type-specific regulatory mechanisms that control or modify systemic and local iron balance, and how shifts in cellular iron levels may affect specialized cell functions.
Collapse
Affiliation(s)
| | - Laura Silvestri
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology IRCCS San Raffaele Scientific Institute Milan Italy
- Vita‐Salute San Raffaele University Milan Italy
| |
Collapse
|
35
|
Anaemia of chronic diseases: Pathophysiology, diagnosis and treatment. Med Clin (Barc) 2020; 156:235-242. [PMID: 33358297 DOI: 10.1016/j.medcli.2020.07.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/18/2020] [Accepted: 07/20/2020] [Indexed: 12/15/2022]
Abstract
Anaemia of chronic disease (ACD) is generated by the activation of the immune system by autoantigens, microbial molecules or tumour antigens resulting in the release of cytokines that cause an elevation of serum hepcidin, hypoferraemia, suppression of erythropoiesis, decrease in erythropoietin (EPO) and shortening of the half-life of red blood cells. Anaemia is usually normocytic and normochromic, which is the most prevalent after iron deficiency anaemia, and it is the most frequent in the elderly and in hospitalized patients. If the anaemia is severe, the patient's quality of life deteriorates, and it can have a negative impact on survival. Treatment is aimed at controlling the underlying disease and correcting anaemia. Sometimes intravenous iron and EPO have been used, but the therapeutic future is directed against hepcidin, which is the final target of anaemia.
Collapse
|
36
|
Richard C, Verdier F. Transferrin Receptors in Erythropoiesis. Int J Mol Sci 2020; 21:ijms21249713. [PMID: 33352721 PMCID: PMC7766611 DOI: 10.3390/ijms21249713] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022] Open
Abstract
Erythropoiesis is a highly dynamic process giving rise to red blood cells from hematopoietic stem cells present in the bone marrow. Red blood cells transport oxygen to tissues thanks to the hemoglobin comprised of α- and β-globin chains and of iron-containing hemes. Erythropoiesis is the most iron-consuming process to support hemoglobin production. Iron delivery is mediated via transferrin internalization by the endocytosis of transferrin receptor type 1 (TFR1), one of the most abundant membrane proteins of erythroblasts. A second transferrin receptor—TFR2—associates with the erythropoietin receptor and has been implicated in the regulation of erythropoiesis. In erythroblasts, both transferrin receptors adopt peculiarities such as an erythroid-specific regulation of TFR1 and a trafficking pathway reliant on TFR2 for iron. This review reports both trafficking and signaling functions of these receptors and reassesses the debated role of TFR2 in erythropoiesis in the light of recent findings. Potential therapeutic uses targeting the transferrin-TFR1 axis or TFR2 in hematological disorders are also discussed.
Collapse
Affiliation(s)
- Cyrielle Richard
- Inserm U1016, CNRS UMR8104, Institut Cochin, Université de Paris, 75014 Paris, France;
- Laboratoire d’excellence GR-Ex, Université de Paris, 75014 Paris, France
| | - Frédérique Verdier
- Inserm U1016, CNRS UMR8104, Institut Cochin, Université de Paris, 75014 Paris, France;
- Laboratoire d’excellence GR-Ex, Université de Paris, 75014 Paris, France
- Correspondence:
| |
Collapse
|
37
|
Jing W, Guo X, Qin F, Li Y, Wang G, Bi Y, Jin X, Han L, Dong X, Zhao Y. G-CSF shifts erythropoiesis from bone marrow into spleen in the setting of systemic inflammation. Life Sci Alliance 2020; 4:4/1/e202000737. [PMID: 33234677 PMCID: PMC7723243 DOI: 10.26508/lsa.202000737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 11/08/2020] [Accepted: 11/09/2020] [Indexed: 11/24/2022] Open
Abstract
The anemia of inflammation is related in part to abnormal erythropoiesis in bone marrow. G-CSF regulates granulopoiesis and is increased during systemic inflammation. Here, we have showed that high levels of G-CSF are associated with repression of bone marrow erythropoiesis and expansion of splenic erythropoiesis in Escherichia coli-infected mice and lipopolysaccharide-treated mice. Under lipopolysaccharide-induced systemic inflammatory conditions in mice, G-CSF neutralization with antibody alleviated the blockage of bone marrow erythropoiesis, prevented the enhancement of splenic erythropoiesis, ameliorated splenomegaly, and reduced the brittleness of spleen. We further demonstrated that after lipopolysaccharide treatment, TLR4-knockout mice display low levels of G-CSF, healthy bone marrow erythropoiesis, almost no stress erythropoiesis in the spleen, and normal size and toughness of spleen. In addition, we found HIF-mediated erythropoietin production is essential for splenic erythropoiesis in the setting of G-CSF-induced suppression of bone marrow erythropoiesis. Our findings identify G-CSF as a critical mediator of inflammation-associated erythropoiesis dysfunction in bone marrow and offer insight into the mechanism of G-CSF-induced splenic erythropoiesis. We provide experimentally significant dimension to the biology of G-CSF.
Collapse
Affiliation(s)
- Weiqiang Jing
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xing Guo
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fei Qin
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Li
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ganyu Wang
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuxuan Bi
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xing Jin
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lihui Han
- Department of Immunology, Shandong Key Laboratory of Infection and Immunity, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoyuan Dong
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yunxue Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China .,Department of Immunology, Shandong Key Laboratory of Infection and Immunity, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
38
|
Yuzbasheva EY, Scarcia P, Yuzbashev TV, Messina E, Kosikhina IM, Palmieri L, Shutov AV, Taratynova MO, Amaro RL, Palmieri F, Sineoky SP, Agrimi G. Engineering Yarrowia lipolytica for the selective and high-level production of isocitric acid through manipulation of mitochondrial dicarboxylate-tricarboxylate carriers. Metab Eng 2020; 65:156-166. [PMID: 33161142 DOI: 10.1016/j.ymben.2020.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/14/2020] [Accepted: 11/01/2020] [Indexed: 11/18/2022]
Abstract
During cultivation under nitrogen starvation, Yarrowia lipolytica produces a mixture of citric acid and isocitric acid whose ratio is mainly determined by the carbon source used. We report that mitochondrial succinate-fumarate carrier YlSfc1 controls isocitric acid efflux from mitochondria. YlSfc1 purified and reconstituted into liposomes transports succinate, fumarate, oxaloacetate, isocitrate and α-ketoglutarate. YlSFC1 overexpression determined the inversion of isocitric acid/citric acid ratio towards isocitric acid, resulting in 33.4 ± 1.9 g/L and 43.3 ± 2.8 g/L of ICA production in test-tube cultivation with glucose and glycerol, respectively. These titers represent a 4.0 and 6.3-fold increase compared to the wild type. YlSFC1 gene expression was repressed in the wild type strain grown in glucose-based medium compared to olive oil medium explaining the reason for the preferred citric acid production during Y. lipolytica growth on carbohydrates. Coexpression of YlSFC1 and adenosine monophosphate deaminase YlAMPD genes together with inactivation of citrate mitochondrial carrier YlYHM2 gene enhanced isocitric acid accumulation up to 41.4 ± 4.1 g/L with an isocitric acid/citric acid ratio of 14.3 in a small-scale cultivation with glucose as a carbon source. During large-scale cultivation with glucose pulse-feeding, the engineered strain produced 136.7 ± 2.5 g/L of ICA with a process selectivity of 88.1%, the highest reported titer and selectivity to date. These results represent the first reported isocitric acid secretion by Y. lipolytica as a main organic acid during cultivation on carbohydrate. Moreover, we demonstrate for the first time that the replacement of one mitochondrial transport system for another can be an efficient tool for switching product accumulation.
Collapse
Affiliation(s)
- Evgeniya Y Yuzbasheva
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK; NRC «Kurchatov Institute» - GOSNIIGENETIKA, Kurchatov Genomic Center, 1-st Dorozhny Pr., 1, Moscow, 117545, Russia; BioMediCan Inc., 40471 Encyclopedia Circle, Fremont, 94538, CA, USA.
| | - Pasquale Scarcia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Campus Universitario, Via Orabona 4, 70125, Bari, Italy
| | - Tigran V Yuzbashev
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK; Centre for Synthetic Biology, Imperial College London, London, SW7 2AZ, UK
| | - Eugenia Messina
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Campus Universitario, Via Orabona 4, 70125, Bari, Italy
| | - Iuliia M Kosikhina
- NRC «Kurchatov Institute» - GOSNIIGENETIKA, Kurchatov Genomic Center, 1-st Dorozhny Pr., 1, Moscow, 117545, Russia; NRC «Kurchatov Institute», 1 Kurchatov Square, Moscow, 123182, Russia
| | - Luigi Palmieri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Campus Universitario, Via Orabona 4, 70125, Bari, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Campus Universitario, Via Orabona 4, 70125, Bari, Italy
| | - Artem V Shutov
- NRC «Kurchatov Institute» - GOSNIIGENETIKA, Kurchatov Genomic Center, 1-st Dorozhny Pr., 1, Moscow, 117545, Russia; NRC «Kurchatov Institute», 1 Kurchatov Square, Moscow, 123182, Russia
| | - Maria O Taratynova
- NRC «Kurchatov Institute» - GOSNIIGENETIKA, Kurchatov Genomic Center, 1-st Dorozhny Pr., 1, Moscow, 117545, Russia; NRC «Kurchatov Institute», 1 Kurchatov Square, Moscow, 123182, Russia
| | - Rodrigo Ledesma Amaro
- Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK; Centre for Synthetic Biology, Imperial College London, London, SW7 2AZ, UK
| | - Ferdinando Palmieri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Campus Universitario, Via Orabona 4, 70125, Bari, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Campus Universitario, Via Orabona 4, 70125, Bari, Italy
| | - Sergey P Sineoky
- NRC «Kurchatov Institute» - GOSNIIGENETIKA, Kurchatov Genomic Center, 1-st Dorozhny Pr., 1, Moscow, 117545, Russia; NRC «Kurchatov Institute», 1 Kurchatov Square, Moscow, 123182, Russia
| | - Gennaro Agrimi
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Campus Universitario, Via Orabona 4, 70125, Bari, Italy.
| |
Collapse
|
39
|
Valente de Souza L, Hoffmann A, Weiss G. Impact of bacterial infections on erythropoiesis. Expert Rev Anti Infect Ther 2020; 19:619-633. [PMID: 33092423 DOI: 10.1080/14787210.2021.1841636] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The importance of iron is highlighted by the many complex metabolic pathways in which it is involved. A sufficient supply is essential for the effective production of 200 billion erythrocytes daily, a process called erythropoiesis. AREAS COVERED During infection, the human body can withhold iron from pathogens, mechanism termed nutritional immunity. The subsequent disturbances in iron homeostasis not only impact on immune function and infection control, but also negatively affect erythropoiesis. The complex interplay between iron, immunity, erythropoiesis and infection control on the molecular and clinical level are highlighted in this review. Diagnostic algorithms for correct interpretation and diagnosis of the iron status in the setting of infection are presented. Therapeutic concepts are discussed regarding effects on anemia correction, but also toward their role on the course of infection. EXPERT OPINION In the setting of infection, anemia is often neglected and its impact on the course of diseases is incompletely understood. Clinical expertise can be improved in correct diagnosing of anemia and disturbances of iron homeostasis. Systemic studies are needed to evaluate the impact of specific therapeutic interventions on anemia correction on the course of infection, but also on patients' cardiovascular performance and quality of life.
Collapse
Affiliation(s)
- Lara Valente de Souza
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University ofI nnsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Alexander Hoffmann
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University ofI nnsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University ofI nnsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
40
|
Musio F. Revisiting the treatment of anemia in the setting of chronic kidney disease, hematologic malignancies, and cancer: perspectives with opinion and commentary. Expert Rev Hematol 2020; 13:1175-1188. [PMID: 33028115 DOI: 10.1080/17474086.2020.1830371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 09/27/2020] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Anemia has and will continue to be a central theme in medicine particularly as clinicians are treating a burgeoning population of complex multi-organ system processes. As a result of multiple randomized controlled trials (RCTs), meta-analyses, and societal recommendations overly restrictive paradigms and under-administration of erythropoiesis stimulating agents (ESAs) have likely been followed by clinicians among all specialties. AREAS COVERED A review of anemia in the context of chronic kidney disease, hematologic malignancies, and cancer is presented with focus on the establishment of ESAs as integral in the treatment of anemia. Multiple RCTs and meta-analyses studying the use of ESAs are presented with focus upon their application to clinical practice. A 'compendium' is proffered describing the evolution, establishment, and implications of ESA administration initially among those with CKD with rapid subsequent application to the Hematology-Oncology population of patients. Literature search methodologies have included MEDLINE (1985-2020), PubMed (1996-2020), Cochrane Central Trials (1985-2020), EMBASE (2000-2020), and ClinicalTrials.gov (2000-2020). EXPERT OPINION Upon evaluation of risks and benefits of ESAs focused opinion and commentary is made supporting more liberal use of these agents and strongly suggesting that the current underlying treatment 'pendulum' has perhaps shifted too far to the 'under-treatment' side in many cases.
Collapse
Affiliation(s)
- Franco Musio
- Senior Partner, Nephrology Associates of Northern Virginia , VA, Fairfax, Virginia, USA
- Nephrology Division Chief, Inova Fairfax Hospital, Department of Medicine, Falls Church , Virginia, VA, USA
- Associate Professor of Medicine, Virginia Commonwealth University (Inova Fairfax Hospital Campus) , Richmond, Virginia, USA
| |
Collapse
|
41
|
Bhoopalan SV, Huang LJS, Weiss MJ. Erythropoietin regulation of red blood cell production: from bench to bedside and back. F1000Res 2020; 9:F1000 Faculty Rev-1153. [PMID: 32983414 PMCID: PMC7503180 DOI: 10.12688/f1000research.26648.1] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/04/2020] [Indexed: 12/18/2022] Open
Abstract
More than 50 years of efforts to identify the major cytokine responsible for red blood cell (RBC) production (erythropoiesis) led to the identification of erythropoietin (EPO) in 1977 and its receptor (EPOR) in 1989, followed by three decades of rich scientific discovery. We now know that an elaborate oxygen-sensing mechanism regulates the production of EPO, which in turn promotes the maturation and survival of erythroid progenitors. Engagement of the EPOR by EPO activates three interconnected signaling pathways that drive RBC production via diverse downstream effectors and simultaneously trigger negative feedback loops to suppress signaling activity. Together, the finely tuned mechanisms that drive endogenous EPO production and facilitate its downstream activities have evolved to maintain RBC levels in a narrow physiological range and to respond rapidly to erythropoietic stresses such as hypoxia or blood loss. Examination of these pathways has elucidated the genetics of numerous inherited and acquired disorders associated with deficient or excessive RBC production and generated valuable drugs to treat anemia, including recombinant human EPO and more recently the prolyl hydroxylase inhibitors, which act partly by stimulating endogenous EPO synthesis. Ongoing structure-function studies of the EPOR and its essential partner, tyrosine kinase JAK2, suggest that it may be possible to generate new "designer" drugs that control selected subsets of cytokine receptor activities for therapeutic manipulation of hematopoiesis and treatment of blood cancers.
Collapse
Affiliation(s)
- Senthil Velan Bhoopalan
- Department of Hematology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS #355, Memphis, TN, 38105, USA
| | - Lily Jun-shen Huang
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Mitchell J. Weiss
- Department of Hematology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS #355, Memphis, TN, 38105, USA
| |
Collapse
|
42
|
Araki D, Alvarado LJ, Huntsman HD, Smith RH, Larochelle A. IFN-γ directly inhibits the activity of erythropoietin in human erythroid progenitors. Blood Cells Mol Dis 2020; 85:102488. [PMID: 32889151 DOI: 10.1016/j.bcmd.2020.102488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Daisuke Araki
- National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Luigi J Alvarado
- National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Heather D Huntsman
- National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Richard H Smith
- National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Andre Larochelle
- National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
43
|
Brändle K, Bergmann TC, Raic A, Li Y, Million N, Rehbock C, Barcikowski S, Lee-Thedieck C. Iron Nanoparticle Composite Hydrogels for Studying Effects of Iron Ion Release on Red Blood Cell In Vitro Production. ACS APPLIED BIO MATERIALS 2020; 3:4766-4778. [PMID: 35021724 DOI: 10.1021/acsabm.0c00297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Growing numbers of complex surgical interventions increase the need for blood transfusions, which cannot be fulfilled by the number of donors. Therefore, the interest in producing erythrocytes from their precursors-the hematopoietic stem and progenitor cells (HSPCs)-in laboratories is rising. To enable this, in vitro systems are needed, which allow analysis of the effects of essential factors such as iron on erythroid development. For this purpose, iron ion-releasing systems based on poly(ethylene glycol) (PEG)-iron nanocomposites are developed to assess if gradual iron release improves iron bioavailability during in vitro erythroid differentiation. The nanocomposites are synthesized using surfactant-free pulsed laser ablation of iron directly in the PEG solution. The iron concentrations released from the material are sufficient to influence in vitro erythropoiesis. In this way, the production of erythroid cells cultured on flat PEG-iron nanocomposite hydrogel pads can be enhanced. In contrast, erythroid differentiation is not enhanced in the biomimetic macroporous 3D composite scaffolds, possibly because of local iron overload within the pores of the system. In conclusion, the developed iron nanoparticle-PEG composite hydrogel allows constant iron ion release and thus paves the way (i) to understand the role of iron during erythropoiesis and (ii) toward the development of biomaterials with a controlled iron release for directing erythropoiesis in culture.
Collapse
Affiliation(s)
- Katharina Brändle
- Institute of Functional Interfaces, Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen 76344, Germany.,Institute of Cell Biology and Biophysics, Leibniz University Hannover, Hannover 30419, Germany
| | - Timna C Bergmann
- Institute of Cell Biology and Biophysics, Leibniz University Hannover, Hannover 30419, Germany
| | - Annamarija Raic
- Institute of Functional Interfaces, Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen 76344, Germany.,Institute of Cell Biology and Biophysics, Leibniz University Hannover, Hannover 30419, Germany
| | - Yaya Li
- Technical Chemistry I and Center for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitätsstr. 5-7, Essen 45141, Germany
| | - Nina Million
- Technical Chemistry I and Center for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitätsstr. 5-7, Essen 45141, Germany
| | - Christoph Rehbock
- Technical Chemistry I and Center for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitätsstr. 5-7, Essen 45141, Germany
| | - Stephan Barcikowski
- Technical Chemistry I and Center for Nanointegration Duisburg-Essen (CENIDE), University of Duisburg-Essen, Universitätsstr. 5-7, Essen 45141, Germany
| | - Cornelia Lee-Thedieck
- Institute of Cell Biology and Biophysics, Leibniz University Hannover, Hannover 30419, Germany
| |
Collapse
|
44
|
Guo W, Abou Ghayda R, Schmidt PJ, Fleming MD, Bhasin S. The role of iron in mediating testosterone's effects on erythropoiesis in mice. FASEB J 2020; 34:11672-11684. [PMID: 32667087 DOI: 10.1096/fj.202000920rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 11/11/2022]
Abstract
Testosterone stimulates iron-dependent erythropoiesis and suppresses hepcidin. To clarify the role of iron in mediating testosterone's effects on erythropoiesis, we induced iron deficiency in mice by feeding low iron diet. Iron-replete and iron-deficient mice were treated weekly with testosterone propionate or vehicle for 3 weeks. Testosterone treatment increased red cell count in iron-replete mice, but, surprisingly, testosterone reduced red cell count in iron-deficient mice. Splenic stress erythropoiesis was stimulated in iron-deficient mice relative to iron-replete mice, and further increased by testosterone treatment, as indicated by the increase in red pulp area, the number of nucleated erythroblasts, and expression levels of TfR1, GATA1, and other erythroid genes. Testosterone treatment of iron-deficient mice increased the ratio of early-to-late erythroblasts in the spleen and bone marrow, and serum LDH level, consistent with ineffective erythropoiesis. In iron-deficient mice, erythropoietin levels were higher but erythropoietin-regulated genes were generally downregulated relative to iron-replete mice, suggesting erythropoietin resistance. Conclusion: Testosterone treatment stimulates splenic stress erythropoiesis in iron-replete as well as iron-deficient mice. However, testosterone worsens anemia in iron-deficient mice because of ineffective erythropoiesis possibly due to erythropoietin resistance associated with iron deficiency. Iron plays an important role in mediating testosterone's effects on erythropoiesis.
Collapse
Affiliation(s)
- Wen Guo
- Research Program in Men's Health: Aging and Metabolism, The Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ramy Abou Ghayda
- Research Program in Men's Health: Aging and Metabolism, The Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Paul J Schmidt
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shalender Bhasin
- Research Program in Men's Health: Aging and Metabolism, The Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
45
|
Hypoxia-inducible factor prolyl hydroxylase inhibitor in the treatment of anemia in chronic kidney disease. Curr Opin Nephrol Hypertens 2020; 29:414-422. [DOI: 10.1097/mnh.0000000000000617] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
46
|
Heme-regulated eIF2α kinase in erythropoiesis and hemoglobinopathies. Blood 2020; 134:1697-1707. [PMID: 31554636 DOI: 10.1182/blood.2019001915] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022] Open
Abstract
As essential components of hemoglobin, iron and heme play central roles in terminal erythropoiesis. The impairment of this process in iron/heme deficiency results in microcytic hypochromic anemia, the most prevalent anemia globally. Heme-regulated eIF2α kinase, also known as heme-regulated inhibitor (HRI), is a key heme-binding protein that senses intracellular heme concentrations to balance globin protein synthesis with the amount of heme available for hemoglobin production. HRI is activated during heme deficiency to phosphorylate eIF2α (eIF2αP), which simultaneously inhibits the translation of globin messenger RNAs (mRNAs) and selectively enhances the translation of activating transcription factor 4 (ATF4) mRNA to induce stress response genes. This coordinated translational regulation is a universal hallmark across the eIF2α kinase family under various stress conditions and is termed the integrated stress response (ISR). Inhibition of general protein synthesis by HRI-eIF2αP in erythroblasts is necessary to prevent proteotoxicity and maintain protein homeostasis in the cytoplasm and mitochondria. Additionally, the HRI-eIF2αP-ATF4 pathway represses mechanistic target of rapamycin complex 1 (mTORC1) signaling, specifically in the erythroid lineage as a feedback mechanism of erythropoietin-stimulated erythropoiesis during iron/heme deficiency. Furthermore, ATF4 target genes are most highly activated during iron deficiency to maintain mitochondrial function and redox homeostasis, as well as to enable erythroid differentiation. Thus, heme and translation regulate erythropoiesis through 2 key signaling pathways, ISR and mTORC1, which are coordinated by HRI to circumvent ineffective erythropoiesis (IE). HRI-ISR is also activated to reduce the severity of β-thalassemia intermedia in the Hbbth1/th1 murine model. Recently, HRI has been implicated in the regulation of human fetal hemoglobin production. Therefore, HRI-ISR has emerged as a potential therapeutic target for hemoglobinopathies.
Collapse
|
47
|
|
48
|
Ginzburg YZ. New diagnostic tools for delineating iron status. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:327-336. [PMID: 31808893 PMCID: PMC6913443 DOI: 10.1182/hematology.2019000035] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Recent advances in our understanding of iron metabolism regulation and crosstalk with erythropoiesis have provided insight into the pathophysiology of multiple disease conditions. For instance, the peptide hormone hepcidin is central to the regulation of iron metabolism. Its effect on cellular iron concentration involves binding ferroportin, the main iron export protein, resulting in its internalization and degradation and leading to iron sequestration within ferroportin-expressing cells. Furthermore, hepcidin regulation by erythropoiesis is attributed in large part to a bone marrow-derived hormone erythroferrone. Erythroferrone-induced hepcidin suppression in diseases of expanded hematopoiesis results in iron overload. Conversely, diseases, such as iron refractory iron deficiency anemia and anemia of chronic inflammation, are characterized by aberrantly increased hepcidin, resulting in iron sequestration and decreased circulating iron and eventually leading to iron-restricted erythropoiesis. Lastly, because iron functions in concert with erythropoietin to promote erythroid precursor survival, proliferation, and differentiation, iron deficiency anemia is a consequence not only of decreased hemoglobin synthesis in each cell but also, a decrease in erythropoietin responsiveness in the bone marrow. How to translate this new information to the clinical setting has not been fully elucidated. The purpose of this manuscript is to summarize current standard tools for identifying iron deficiency in anemic patients; explore the tools and context for evaluating novel markers, such as hepcidin, erythroferrone, and markers of the iron restriction response; and assess available evidence for how their use could increase our understanding of health outcomes in clinically challenging cases.
Collapse
|
49
|
Parrow NL, Li Y, Feola M, Guerra A, Casu C, Prasad P, Mammen L, Ali F, Vaicikauskas E, Rivella S, Ginzburg YZ, Fleming RE. Lobe specificity of iron binding to transferrin modulates murine erythropoiesis and iron homeostasis. Blood 2019; 134:1373-1384. [PMID: 31434707 PMCID: PMC6839954 DOI: 10.1182/blood.2018893099] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 08/12/2019] [Indexed: 12/13/2022] Open
Abstract
Transferrin, the major plasma iron-binding molecule, interacts with cell-surface receptors to deliver iron, modulates hepcidin expression, and regulates erythropoiesis. Transferrin binds and releases iron via either or both of 2 homologous lobes (N and C). To test the hypothesis that the specificity of iron occupancy in the N vs C lobe influences transferrin function, we generated mice with mutations to abrogate iron binding in either lobe (TfN-bl or TfC-bl). Mice homozygous for either mutation had hepatocellular iron loading and decreased liver hepcidin expression (relative to iron concentration), although to different magnitudes. Both mouse models demonstrated some aspects of iron-restricted erythropoiesis, including increased zinc protoporphyrin levels, decreased hemoglobin levels, and microcytosis. Moreover, the TfN-bl/N-bl mice demonstrated the anticipated effect of iron restriction on red cell production (ie, no increase in red blood cell [RBC] count despite elevated erythropoietin levels), along with a poor response to exogenous erythropoietin. In contrast, the TfC-bl/C-bl mice had elevated RBC counts and an exaggerated response to exogenous erythropoietin sufficient to ameliorate the anemia. Observations in heterozygous mice further support a role for relative N vs C lobe iron occupancy in transferrin-mediated regulation of iron homeostasis and erythropoiesis.
Collapse
Affiliation(s)
- Nermi L Parrow
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO
| | - Yihang Li
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO
| | - Maria Feola
- Division of Hematology-Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Amaliris Guerra
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA; and
| | - Carla Casu
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA; and
| | - Princy Prasad
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO
| | - Luke Mammen
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO
| | - Faris Ali
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO
| | - Edvinas Vaicikauskas
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO
| | - Stefano Rivella
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA; and
| | - Yelena Z Ginzburg
- Division of Hematology-Oncology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Robert E Fleming
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO
| |
Collapse
|
50
|
Affiliation(s)
- Tomas Ganz
- From the Departments of Medicine and Pathology, David Geffen School of Medicine at UCLA, Los Angeles
| |
Collapse
|