1
|
Sesti-Costa R, Costa FF, Conran N. Role of Macrophages in Sickle Cell Disease Erythrophagocytosis and Erythropoiesis. Int J Mol Sci 2023; 24:ijms24076333. [PMID: 37047304 PMCID: PMC10094208 DOI: 10.3390/ijms24076333] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Sickle cell disease (SCD) is an inherited blood disorder caused by a β-globin gene point mutation that results in the production of sickle hemoglobin that polymerizes upon deoxygenation, causing the sickling of red blood cells (RBCs). RBC deformation initiates a sequence of events leading to multiple complications, such as hemolytic anemia, vaso-occlusion, chronic inflammation, and tissue damage. Macrophages participate in extravascular hemolysis by removing damaged RBCs, hence preventing the release of free hemoglobin and heme, and triggering inflammation. Upon erythrophagocytosis, macrophages metabolize RBC-derived hemoglobin, activating mechanisms responsible for recycling iron, which is then used for the generation of new RBCs to try to compensate for anemia. In the bone marrow, macrophages can create specialized niches, known as erythroblastic islands (EBIs), which regulate erythropoiesis. Anemia and inflammation present in SCD may trigger mechanisms of stress erythropoiesis, intensifying RBC generation by expanding the number of EBIs in the bone marrow and creating new ones in extramedullary sites. In the current review, we discuss the distinct mechanisms that could induce stress erythropoiesis in SCD, potentially shifting the macrophage phenotype to an inflammatory profile, and changing their supporting role necessary for the proliferation and differentiation of erythroid cells in the disease. The knowledge of the soluble factors, cell surface and intracellular molecules expressed by EBI macrophages that contribute to begin and end the RBC’s lifespan, as well as the understanding of their signaling pathways in SCD, may reveal potential targets to control the pathophysiology of the disease.
Collapse
|
2
|
Romano L, Seu KG, Papoin J, Muench DE, Konstantinidis D, Olsson A, Schlum K, Chetal K, Chasis JA, Mohandas N, Barnes BJ, Zheng Y, Grimes HL, Salomonis N, Blanc L, Kalfa TA. Erythroblastic islands foster granulopoiesis in parallel to terminal erythropoiesis. Blood 2022; 140:1621-1634. [PMID: 35862735 PMCID: PMC9707396 DOI: 10.1182/blood.2022015724] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/26/2022] [Indexed: 12/14/2022] Open
Abstract
The erythroblastic island (EBI), composed of a central macrophage surrounded by maturing erythroblasts, is the erythroid precursor niche. Despite numerous studies, its precise composition is still unclear. Using multispectral imaging flow cytometry, in vitro island reconstitution, and single-cell RNA sequencing of adult mouse bone marrow (BM) EBI-component cells enriched by gradient sedimentation, we present evidence that the CD11b+ cells present in the EBIs are neutrophil precursors specifically associated with BM EBI macrophages, indicating that erythro-(myelo)-blastic islands are a site for terminal granulopoiesis and erythropoiesis. We further demonstrate that the balance between these dominant and terminal differentiation programs is dynamically regulated within this BM niche by pathophysiological states that favor granulopoiesis during anemia of inflammation and favor erythropoiesis after erythropoietin stimulation. Finally, by molecular profiling, we reveal the heterogeneity of EBI macrophages by cellular indexing of transcriptome and epitope sequencing of mouse BM EBIs at baseline and after erythropoietin stimulation in vivo and provide a searchable online viewer of these data characterizing the macrophage subsets serving as hematopoietic niches. Taken together, our findings demonstrate that EBIs serve a dual role as niches for terminal erythropoiesis and granulopoiesis and the central macrophages adapt to optimize production of red blood cells or neutrophils.
Collapse
Affiliation(s)
- Laurel Romano
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Katie G Seu
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Julien Papoin
- Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Laboratory of Pediatric Oncology, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY
| | - David E Muench
- Immunology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, San Diego, CA
| | | | | | - Katrina Schlum
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Kashish Chetal
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA
| | - Joel Anne Chasis
- Life Sciences Division, University of California, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Narla Mohandas
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY
| | - Betsy J Barnes
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institutes for Medical Research, Manhasset, NY
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - H Leighton Grimes
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Lionel Blanc
- Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Laboratory of Pediatric Oncology, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY
| | - Theodosia A Kalfa
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
3
|
Lévesque JP, Summers KM, Bisht K, Millard SM, Winkler IG, Pettit AR. Macrophages form erythropoietic niches and regulate iron homeostasis to adapt erythropoiesis in response to infections and inflammation. Exp Hematol 2021; 103:1-14. [PMID: 34500024 DOI: 10.1016/j.exphem.2021.08.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 12/24/2022]
Abstract
It has recently emerged that tissue-resident macrophages are key regulators of several stem cell niches orchestrating tissue formation during development, as well as postnatally, when they also organize the repair and regeneration of many tissues including the hemopoietic tissue. The fact that macrophages are also master regulators and effectors of innate immunity and inflammation allows them to coordinate hematopoietic response to infections, injuries, and inflammation. After recently reviewing the roles of phagocytes and macrophages in regulating normal and pathologic hematopoietic stem cell niches, we now focus on the key roles of macrophages in regulating erythropoiesis and iron homeostasis. We review herein the recent advances in understanding how macrophages at the center of erythroblastic islands form an erythropoietic niche that controls the terminal differentiation and maturation of erythroblasts into reticulocytes; how red pulp macrophages in the spleen control iron recycling and homeostasis; how these macrophages coordinate emergency erythropoiesis in response to blood loss, infections, and inflammation; and how persistent infections or inflammation can lead to anemia of inflammation via macrophages. Finally, we discuss the technical challenges associated with the molecular characterization of erythroid island macrophages and red pulp macrophages.
Collapse
Affiliation(s)
- Jean-Pierre Lévesque
- Mater Research Institute - The University of Queensland, Woolloongabba, QLD, Australia.
| | - Kim M Summers
- Mater Research Institute - The University of Queensland, Woolloongabba, QLD, Australia
| | - Kavita Bisht
- Mater Research Institute - The University of Queensland, Woolloongabba, QLD, Australia
| | - Susan M Millard
- Mater Research Institute - The University of Queensland, Woolloongabba, QLD, Australia
| | - Ingrid G Winkler
- Mater Research Institute - The University of Queensland, Woolloongabba, QLD, Australia
| | - Allison R Pettit
- Mater Research Institute - The University of Queensland, Woolloongabba, QLD, Australia
| |
Collapse
|
4
|
Correa-Gallegos D, Jiang D, Rinkevich Y. Fibroblasts as confederates of the immune system. Immunol Rev 2021; 302:147-162. [PMID: 34036608 DOI: 10.1111/imr.12972] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 12/18/2022]
Abstract
Fibroblastic stromal cells are as diverse, in origin and function, as the niches they fashion in the mammalian body. This cellular variety impacts the spectrum of responses elicited by the immune system. Fibroblast influence on the immune system keeps evolving our perspective on fibroblast roles and functions beyond just a passive structural part of organs. This review discusses the foundations of fibroblastic stromal-immune crosstalk, under the scope of stromal heterogeneity as a basis for tissue-specific tutoring of the immune system. Focusing on the skin as a relevant immunological organ, we detail the complex interactions between distinct fibroblast populations and immune cells that occur during homeostasis, injury repair, scarring, and disease. We further review the relevance of fibroblastic stromal cell heterogeneity and how this heterogeneity is central to regulate the immune system from its inception during embryonic development into adulthood.
Collapse
Affiliation(s)
- Donovan Correa-Gallegos
- Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München, Munich, Germany
| | - Dongsheng Jiang
- Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München, Munich, Germany
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| |
Collapse
|
5
|
Paulson RF, Hariharan S, Little JA. Stress erythropoiesis: definitions and models for its study. Exp Hematol 2020; 89:43-54.e2. [PMID: 32750404 DOI: 10.1016/j.exphem.2020.07.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022]
Abstract
Steady-state erythropoiesis generates new erythrocytes at a constant rate, and it has enormous productive capacity. This production is balanced by the removal of senescent erythrocytes by macrophages in the spleen and liver. Erythroid homeostasis is highly regulated to maintain sufficient erythrocytes for efficient oxygen delivery to the tissues, while avoiding viscosity problems associated with overproduction. However, there are times when this constant production of erythrocytes is inhibited or is inadequate; at these times, erythroid output is increased to compensate for the loss of production. In some cases, increased steady-state erythropoiesis can offset the loss of erythrocytes but, in response to inflammation caused by infection or tissue damage, steady-state erythropoiesis is inhibited. To maintain homeostasis under these conditions, an alternative stress erythropoiesis pathway is activated. Emerging data suggest that the bone morphogenetic protein 4 (BMP4)-dependent stress erythropoiesis pathway is integrated into the inflammatory response and generates a bolus of new erythrocytes that maintain homeostasis until steady-state erythropoiesis can resume. In this perspective, we define the mechanisms that generate new erythrocytes when steady-state erythropoiesis is impaired and discuss experimental models to study human stress erythropoiesis.
Collapse
Affiliation(s)
- Robert F Paulson
- Center for Molecular Immunology and Infectious Disease and the Department of Veterinary and Biomedical Sciences, Penn State University, University Park, PA; Intercollege Graduate Program in Genetics, Penn State University, University Park, PA.
| | - Sneha Hariharan
- Intercollege Graduate Program in Genetics, Penn State University, University Park, PA
| | - Jane A Little
- Department of Medicine, University of North Carolina Comprehensive Sickle Cell Disease Program, Chapel Hill, NC
| |
Collapse
|
6
|
Aglialoro F, Hofsink N, Hofman M, Brandhorst N, van den Akker E. Inside Out Integrin Activation Mediated by PIEZO1 Signaling in Erythroblasts. Front Physiol 2020; 11:958. [PMID: 32848880 PMCID: PMC7411472 DOI: 10.3389/fphys.2020.00958] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022] Open
Abstract
The non-selective mechanosensitive ion channel PIEZO1 controls erythrocyte volume homeostasis. Different missense gain-of-function mutations in PIEZO1 gene have been identified that cause Hereditary Xerocytosis (HX), a rare autosomal dominant haemolytic anemia. PIEZO1 expression is not limited to erythrocytes and expression levels are significantly higher in erythroid precursors, hinting to a role in erythropoiesis. During erythropoiesis, interactions between erythroblasts, central macrophages, and extracellular matrix within erythroblastic islands are important. Integrin α4β1 and α5β1 present on erythroblasts facilitate such interactions in erythroblastic islands. Here we found that chemical activation of PIEZO1 using Yoda1 leads to increased adhesion to VCAM1 and fibronectin in flowing conditions. Integrin α4, α5, and β1 blocking antibodies prevented this PIEZO1-induced adhesion suggesting inside-out activation of integrin on erythroblasts. Blocking the Ca2+ dependent Calpain and PKC pathways by using specific inhibitors also blocked increased erythroid adhesion to VCAM1 and fibronectins. Cleavage of Talin was observed as a result of Calpain and PKC activity. In conclusion, PIEZO1 activation results in inside-out integrin activation, facilitated by calcium-dependent activation of PKC and Calpain. The data introduces novel concepts in Ca2+ signaling during erythropoiesis with ramification on erythroblastic island homeostasis in health and disease like Hereditary Xerocytosis.
Collapse
Affiliation(s)
- Francesca Aglialoro
- Sanquin Research and Landsteiner Laboratory, Department of Haematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Naomi Hofsink
- Sanquin Research and Landsteiner Laboratory, Department of Haematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Menno Hofman
- Sanquin Research and Landsteiner Laboratory, Department of Haematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Nicole Brandhorst
- Sanquin Research and Landsteiner Laboratory, Department of Haematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Emile van den Akker
- Sanquin Research and Landsteiner Laboratory, Department of Haematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
7
|
Matrix Mechanosensation in the Erythroid and Megakaryocytic Lineages. Cells 2020; 9:cells9040894. [PMID: 32268541 PMCID: PMC7226728 DOI: 10.3390/cells9040894] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 03/30/2020] [Accepted: 04/02/2020] [Indexed: 12/21/2022] Open
Abstract
The biomechanical properties of the bone marrow microenvironment emerge from a combination of interactions between various extracellular matrix (ECM) structural proteins and soluble factors. Matrix stiffness directs stem cell fate, and both bone marrow stromal and hematopoietic cells respond to biophysical cues. Within the bone marrow, the megakaryoblasts and erythroblasts are thought to originate from a common progenitor, giving rise to fully mature magakaryocytes (the platelet precursors) and erythrocytes. Erythroid and megakaryocytic progenitors sense and respond to the ECM through cell surface adhesion receptors such as integrins and mechanosensitive ion channels. While hematopoietic stem progenitor cells remain quiescent on stiffer ECM substrates, the maturation of the erythroid and megakaryocytic lineages occurs on softer ECM substrates. This review surveys the major matrix structural proteins that contribute to the overall biomechanical tone of the bone marrow, as well as key integrins and mechanosensitive ion channels identified as ECM sensors in context of megakaryocytosis or erythropoiesis.
Collapse
|
8
|
Tay J, Bisht K, McGirr C, Millard SM, Pettit AR, Winkler IG, Levesque JP. Imaging flow cytometry reveals that granulocyte colony-stimulating factor treatment causes loss of erythroblastic islands in the mouse bone marrow. Exp Hematol 2020; 82:33-42. [DOI: 10.1016/j.exphem.2020.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 11/15/2022]
|
9
|
Ulyanova T, Georgolopoulos G, Papayannopoulou T. Reappraising the role of α5 integrin and the microenvironmental support in stress erythropoiesis. Exp Hematol 2019; 81:16-31.e4. [PMID: 31887343 DOI: 10.1016/j.exphem.2019.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/13/2019] [Accepted: 12/15/2019] [Indexed: 02/06/2023]
Abstract
We previously studied the role of β1 integrin and some of its different α partners relevant to erythropoiesis. Although clear and consistent answers regarding the role of α4β1 (VLA-4) were evident, the role of its companion integrin α5β1 (VLA-5) was clouded by inconsistent outcomes in all prior publications. Furthermore, the functional consequences of integrin deficiencies only in microenvironmental (ME) cells supporting erythroid cell expansion and maturation post stress have never been explored. In the study described here, we created several additional mouse models in the aim of addressing unanswered questions regarding functional consequences of single or combined integrin deficiencies in erythroid cells or only in ME supporting cells. Our novel and expansive data solidified the intrinsic requirement of both α4 and α5 integrins in erythroid cells for their proliferative expansion and maturation in response to stress; α5 integrin alone, deleted either early in all hematopoietic cells or only in erythroid cell, has only a redundant role in proliferative expansion and is dispensable for erythroid maturation. By contrast, α4 integrin, on its own, exerts a dominant effect on timely and optimal erythroid maturation. Deficiency of both α4 and α5 integrins in ME cells, including macrophages, does not negatively influence stress response by normal erythroid cells, in great contrast to the effect of ME cells deficient in all β1 integrins. Collectively the present data offer deeper insight into the coordination of different β1 integrin functional activities in erythroid cells or in ME cells for optimal erythroid stress response.
Collapse
Affiliation(s)
- Tatyana Ulyanova
- Division of Hematology, Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Thalia Papayannopoulou
- Division of Hematology, Department of Medicine, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
10
|
Oliveira M, Laranjeira P, Fortuna M, Bártolo R, Ribeiro A, Santos M, Cortesão E, Marques G, Sarmento‐Ribeiro AB, Vitória H, Ribeiro L, Paiva A. CD43 and CD49d from the B‐Cell Chronic Lymphoproliferative Disorders Diagnostic Panel Are Useful to Detect Erythroid Dysplasia. CYTOMETRY PART B-CLINICAL CYTOMETRY 2019; 96:417-425. [DOI: 10.1002/cyto.b.21792] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 04/23/2019] [Accepted: 05/18/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Melissa Oliveira
- Unidade de Gestão Operacional de Citometria, Serviço de Patologia ClínicaCentro Hospitalar e Universitário de Coimbra Portugal
| | - Paula Laranjeira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculdade de MedicinaUniversidade de Coimbra Portugal
| | - Manuela Fortuna
- Unidade de Gestão Operacional de Citometria, Serviço de Patologia ClínicaCentro Hospitalar e Universitário de Coimbra Portugal
| | - Rui Bártolo
- Unidade de Gestão Operacional de Citometria, Serviço de Patologia ClínicaCentro Hospitalar e Universitário de Coimbra Portugal
| | - André Ribeiro
- Serviço de Hematologia ClínicaCentro Hospitalar e Universitário de Coimbra Portugal
| | - Mónica Santos
- Serviço de HematologiaCentro Hospitalar Viseu‐Tondela Portugal
| | - Emília Cortesão
- Serviço de Hematologia ClínicaCentro Hospitalar e Universitário de Coimbra Portugal
| | - Gilberto Marques
- Serviço de Patologia ClínicaCentro Hospitalar e Universitário de Coimbra Portugal
| | - Ana Bela Sarmento‐Ribeiro
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculdade de MedicinaUniversidade de Coimbra Portugal
- Serviço de Hematologia ClínicaCentro Hospitalar e Universitário de Coimbra Portugal
| | - Helena Vitória
- Serviço de HematologiaCentro Hospitalar Viseu‐Tondela Portugal
| | - Letícia Ribeiro
- Serviço de Hematologia ClínicaCentro Hospitalar e Universitário de Coimbra Portugal
| | - Artur Paiva
- Unidade de Gestão Operacional de Citometria, Serviço de Patologia ClínicaCentro Hospitalar e Universitário de Coimbra Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculdade de MedicinaUniversidade de Coimbra Portugal
- Instituto Politécnico de Coimbra, ESTESC‐Coimbra Health SchoolCiências Biomédicas Laboratoriais Portugal
| |
Collapse
|
11
|
Maea expressed by macrophages, but not erythroblasts, maintains postnatal murine bone marrow erythroblastic islands. Blood 2019; 133:1222-1232. [PMID: 30674470 DOI: 10.1182/blood-2018-11-888180] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/18/2019] [Indexed: 12/11/2022] Open
Abstract
The erythroblastic island (EI), formed by a central macrophage and developing erythroblasts (EBs), was first described decades ago and was recently shown to play an in vivo role in homeostatic and pathological erythropoiesis. The exact molecular mechanisms, however, mediating the interactions between macrophages and EBs remain unclear. Macrophage-EB attacher (Maea) has previously been suggested to mediate homophilic adhesion bounds bridging macrophages and EBs. Maea-deficient mice die perinatally with anemia and defective erythrocyte enucleation, suggesting a critical role in fetal erythropoiesis. Here, we generated conditional knockout mouse models of Maea to assess its cellular and postnatal contributions. Deletion of Maea in macrophages using Csf1r-Cre or CD169-Cre caused severe reductions of bone marrow (BM) macrophages, EBs, and in vivo island formation, whereas its deletion in the erythroid lineage using Epor-Cre had no such phenotype, suggesting a dominant role of Maea in the macrophage for BM erythropoiesis. Interestingly, Maea deletion in spleen macrophages did not alter their numbers or functions. Postnatal Maea deletion using Mx1-Cre or function inhibition using a novel monoclonal antibody also impaired BM erythropoiesis. These results indicate that Maea contributes to adult BM erythropoiesis by regulating the maintenance of macrophages and their interaction with EBs via an as-yet-unidentified EB receptor.
Collapse
|
12
|
Izawa Y, Gu YH, Osada T, Kanazawa M, Hawkins BT, Koziol JA, Papayannopoulou T, Spatz M, Del Zoppo GJ. β1-integrin-matrix interactions modulate cerebral microvessel endothelial cell tight junction expression and permeability. J Cereb Blood Flow Metab 2018; 38:641-658. [PMID: 28787238 PMCID: PMC5888854 DOI: 10.1177/0271678x17722108] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Acutely following focal cerebral ischemia disruption of the microvessel blood-brain barrier allows transit of plasma proteins into the neuropil as edema formation that coincides with loss of microvessel endothelial β1-integrins. We extend previous findings to show that interference with endothelial β1-integrin-matrix adhesion by the monoclonal IgM Ha2/5 increases the permeability of primary cerebral microvascular endothelial cell monolayers through reorganization of claudin-5, occludin, and zonula occludens-1 (ZO-1) from inter-endothelial borders. Interference with β1-integrin-matrix adhesion initiates F-actin conformational changes that coincide with claudin-5 redistribution. β1-integrin-matrix interference simultaneously increases phosphorylation of myosin light chain (MLC), while inhibition of MLC kinase (MLCK) and Rho kinase (ROCK) abolishes the Ha2/5-dependent increased endothelial permeability by 6 h after β1-integrin-matrix interference. These observations are supported by concordant observations in the cortex of a high-quality murine conditional β1-integrin deletion construct. Together they support the hypothesis that detachment of β1-integrins from abluminal matrix ligands increases vascular endothelial permeability through reorganization of tight junction (TJ) proteins via altered F-actin conformation, and indicate that the β1-integrin-MLC signaling pathway is engaged when β1-integrin detachment occurs. These findings provide a novel approach to the research and treatment of cerebral disorders where the breakdown of the blood-brain barrier accounts for their progression and complication.
Collapse
Affiliation(s)
- Yoshikane Izawa
- 1 Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.,2 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Yu-Huan Gu
- 1 Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Takashi Osada
- 1 Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.,2 Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Masato Kanazawa
- 1 Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.,3 Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Brian T Hawkins
- 1 Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.,4 Discovery, Science, & Technology, RTI International, Research Triangle Park, NC, USA
| | - James A Koziol
- 5 Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Thalia Papayannopoulou
- 1 Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Maria Spatz
- 6 Stroke Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Gregory J Del Zoppo
- 1 Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.,7 Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
13
|
|
14
|
Heideveld E, Hampton-O'Neil LA, Cross SJ, van Alphen FPJ, van den Biggelaar M, Toye AM, van den Akker E. Glucocorticoids induce differentiation of monocytes towards macrophages that share functional and phenotypical aspects with erythroblastic island macrophages. Haematologica 2017; 103:395-405. [PMID: 29284682 PMCID: PMC5830394 DOI: 10.3324/haematol.2017.179341] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022] Open
Abstract
The classical central macrophage found in erythroblastic islands plays an important role in erythroblast differentiation, proliferation and enucleation in the bone marrow. Convenient human in vitro models to facilitate the study of erythroid-macrophage interactions are desired. Recently, we demonstrated that cultured monocytes/macrophages enhance in vitro erythropoiesis by supporting hematopoietic stem and progenitor cell survival. Herein, we describe that these specific macrophages also support erythropoiesis. Human monocytes cultured in serum-free media supplemented with stem cell factor, erythropoietin, lipids and dexamethasone differentiate towards macrophages expressing CD16, CD163, CD169, CD206, CXCR4 and the phagocytic TAM-receptor family. Phenotypically, they resemble both human bone marrow and fetal liver resident macrophages. This differentiation is dependent on glucocorticoid receptor activation. Proteomic studies confirm that glucocorticoid receptor activation differentiates monocytes to anti-inflammatory tissue macrophages with a M2 phenotype, termed GC-macrophages. Proteins involved in migration, tissue residence and signal transduction/receptor activity are upregulated whilst lysosome and hydrolase activity GO-categories are downregulated. Functionally, we demonstrate that GC-macrophages are highly mobile and can interact to form clusters with erythroid cells of all differentiation stages and phagocytose the expelled nuclei, recapitulating aspects of erythroblastic islands. In conclusion, glucocorticoid-directed monocyte differentiation to macrophages represents a convenient model system to study erythroid-macrophage interactions.
Collapse
Affiliation(s)
- Esther Heideveld
- Sanquin Research, Department of Hematopoiesis, Amsterdam and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, the Netherlands
| | | | - Stephen J Cross
- Wolfson Bioimaging Facility, School of Medical Sciences, Bristol, UK
| | | | - Maartje van den Biggelaar
- Sanquin Research, Department of Research Facilities, Amsterdam, the Netherlands.,Sanquin Research, Department of Plasma Proteins, Amsterdam, the Netherlands
| | - Ashley M Toye
- Department of Biochemistry, School of Medical Sciences, Bristol, UK.,Bristol Institute for Transfusion Sciences, NHS Blood and Transplant, Filton, Bristol, UK.,National Institute for Health Research (NIHR) Blood and Transplant Research Unit in Red Blood Cell Products, University of Bristol, UK
| | - Emile van den Akker
- Sanquin Research, Department of Hematopoiesis, Amsterdam and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, the Netherlands
| |
Collapse
|
15
|
McKenzie CV, Colonne CK, Yeo JH, Fraser ST. Splenomegaly: Pathophysiological bases and therapeutic options. Int J Biochem Cell Biol 2017; 94:40-43. [PMID: 29191734 DOI: 10.1016/j.biocel.2017.11.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 09/25/2017] [Accepted: 11/26/2017] [Indexed: 01/23/2023]
Abstract
The spleen is the largest immune organ in the human body and is also essential for red blood cell homeostasis and iron recycling. An average human spleen is approximately 10 centimetres in length and weighs 150g. Pathological conditions can result in the spleen weighing in excess of 2000g and extending over 30 centrimetres in length. This remarkable property of the spleen to expand is termed splenomegaly. Splenomegaly can occur as a physiological response to stress or as a chronic process that is often detrimental to the wellbeing of the individual. Here, we will discuss the normal function and physiology of the spleen, the pathophysiological bases of splenomegaly and the commonly available therapeutic options. Additionally we will address experimental systems to determine the regulatory mechanisms underlying splenomegaly.
Collapse
Affiliation(s)
- Campbell V McKenzie
- Laboratory of Blood Cell Development, Discipline of Physiology, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Chanukya K Colonne
- Laboratory of Blood Cell Development, Discipline of Physiology, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Jia Hao Yeo
- Laboratory of Blood Cell Development, Discipline of Physiology, School of Medical Sciences, University of Sydney, Camperdown, Australia; Discipline of Anatomy and Histology, School of Medical Sciences, University of Sydney, Camperdown, Australia
| | - Stuart T Fraser
- Laboratory of Blood Cell Development, Discipline of Physiology, School of Medical Sciences, University of Sydney, Camperdown, Australia; Discipline of Anatomy and Histology, School of Medical Sciences, University of Sydney, Camperdown, Australia; Australian Institute of Nanoscale Science and Technology, University of Sydney, Camperdown, Australia; Bosch Institute, School of Medical Sciences, University of Sydney, Camperdown, Australia.
| |
Collapse
|
16
|
The macrophage contribution to stress erythropoiesis: when less is enough. Blood 2016; 128:1756-65. [PMID: 27543439 DOI: 10.1182/blood-2016-05-714527] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/12/2016] [Indexed: 12/15/2022] Open
Abstract
Although the importance of native bone marrow and spleen macrophages in enhancing baseline and stress erythropoiesis has been emphasized over several decades, their kinetic and phenotypic changes during a variety of stress responses have been unclear. Furthermore, whether monocyte-derived recruited macrophages can functionally substitute for inadequate or functionally impaired native macrophages has been controversial and seem to be not only tissue- but also stress-type dependent. To provide further insight into these issues, we made detailed observations at baseline and post-erythroid stress (E-stress) in 2 mouse models with genetically depressed macrophage numbers and compared them to their controls. We documented that, irrespective of the stress-induced (hemolytic or post-erythropoietin [Epo]) treatment, only native CD11b(lo) splenic macrophages expand dramatically post-stress in normal mice without significant changes in the monocyte-derived CD11b(hi) subset. The latter remained a minority and did not change post-stress in 2 genetic models lacking either Spi-C or VCAM-1 with impaired native macrophage proliferative expansion. Although CD11b(lo) macrophages in these mice were one-fifth of normal at their peak response, surprisingly, their erythroid response was not compromised and was similar to controls. Thus, despite the prior emphasis on numerical macrophage reliance to provide functional rescue from E-stress, our data highlight the importance of previously described non-macrophage-dependent pathways activated under certain stress conditions to compensate for low macrophage numbers.
Collapse
|
17
|
Role of bone marrow macrophages in controlling homeostasis and repair in bone and bone marrow niches. Semin Cell Dev Biol 2016; 61:12-21. [PMID: 27521519 DOI: 10.1016/j.semcdb.2016.08.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 08/09/2016] [Accepted: 08/09/2016] [Indexed: 12/24/2022]
Abstract
Macrophages, named for their phagocytic ability, participate in homeostasis, tissue regeneration and inflammatory responses. Bone and adjacent marrow contain multiple functionally unique resident tissue macrophage subsets which maintain and regulate anatomically distinct niche environments within these interconnected tissues. Three subsets of bone-bone marrow resident tissue macrophages have been characterised; erythroblastic island macrophages, haematopoietic stem cell niche macrophages and osteal macrophages. The role of these macrophages in controlling homeostasis and repair in bone and bone marrow niches is reviewed in detail.
Collapse
|
18
|
Critical requirement of VEGF-C in transition to fetal erythropoiesis. Blood 2016; 128:710-20. [DOI: 10.1182/blood-2015-12-687970] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 06/13/2016] [Indexed: 12/14/2022] Open
Abstract
Key Points
Vegfc is essential for mobilization, maturation, and enucleation of primitive erythroblasts. Vegfc deletion compromises liver colonization by erythro-myeloid progenitors and subsequent macrophage/erythroid expansion.
Collapse
|
19
|
Koury MJ. Tracking erythroid progenitor cells in times of need and times of plenty. Exp Hematol 2016; 44:653-63. [DOI: 10.1016/j.exphem.2015.10.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 09/22/2015] [Accepted: 10/28/2015] [Indexed: 01/01/2023]
|
20
|
Abstract
PURPOSE OF REVIEW The nature and function of macrophages at the center of erythroblastic islands is not fully understood. This review discusses novel findings on the phenotypic and molecular characterization of erythroblastic island macrophages, and their role in regulating normal and pathological erythropoiesis. RECENT FINDINGS The phenotype to prospectively isolate erythroblastic island macrophages from mouse bone marrow has been identified. In-vivo depletion of erythroblastic island macrophages causes blockade of erythroblast maturation and delays erythropoietic recovery following chemical insults. The cytokine granulocyte colony-stimulating factor arrests medullary erythropoiesis by depleting erythroblastic island macrophages from the bone marrow. In-vivo ablation of macrophages improves anemia associated with β-thalassemia and reduces red blood cell counts in the mouse model of polycythemia vera. The role of cell adhesion molecules regulating interactions between erythroblastic island macrophages and erythroblasts has been clarified, and mechanisms of pyrenocyte engulfment by erythroblastic island macrophages have been demonstrated to involve Mer tyrosine kinase receptor. SUMMARY Prospective isolation of mouse erythroblastic island macrophages together with new genetic mouse models to specifically target erythroblastic island macrophages will enable molecular studies to better define their role in controlling erythroblast maturation. These studies have revealed the key role of erythroblastic island macrophages in regulating normal erythropoiesis and could be interesting targets to treat β-thalassemia or polycythemia vera.
Collapse
|
21
|
Zhang RR, Zhu XF. [Relationship between macrophages and erythropoiesis]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2016; 18:94-9. [PMID: 26781420 PMCID: PMC7390087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/20/2015] [Indexed: 08/01/2024]
Abstract
Macrophages have two major roles in regulating the dynamic equilibrium in erythropoiesis, promoting the differentiation and maturation of nucleated red blood cells into reticulocytes and removing old red blood cells. A recent mouse study has demonstrated that the phenotype of macrophages in erythroblastic islands is CD169+ VCAM-1+ ER-HR3+ CD11b+ F4/80+ Ly-6G+. Molecular connections between erythroid progenitor cells and central macrophages help to maintain the function and integrity of erythroblastic islands. New research advances in Kruppel-like factor 1 (KLF1) provide new evidence for the important role of macrophages in erythroblastic islands. Macrophages play an important role in erythropoiesis both in sickness and in health, and provide a potential targeted therapy for diseases such as polycythemia vera and beta-thalassemia in the future.
Collapse
Affiliation(s)
- Ran-Ran Zhang
- Diagnosis and Treatment Center of Pediatric Blood Diseases, Institute of Hematology and Blood Disease Hospital, Pecking Union Medical College, Chinese Academy of Medical Sciences, Tianjin 300020, China.
| | | |
Collapse
|
22
|
Zhang RR, Zhu XF. [Relationship between macrophages and erythropoiesis]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2016; 18:94-99. [PMID: 26781420 PMCID: PMC7390087 DOI: 10.7499/j.issn.1008-8830.2016.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/20/2015] [Indexed: 06/05/2023]
Abstract
Macrophages have two major roles in regulating the dynamic equilibrium in erythropoiesis, promoting the differentiation and maturation of nucleated red blood cells into reticulocytes and removing old red blood cells. A recent mouse study has demonstrated that the phenotype of macrophages in erythroblastic islands is CD169+ VCAM-1+ ER-HR3+ CD11b+ F4/80+ Ly-6G+. Molecular connections between erythroid progenitor cells and central macrophages help to maintain the function and integrity of erythroblastic islands. New research advances in Kruppel-like factor 1 (KLF1) provide new evidence for the important role of macrophages in erythroblastic islands. Macrophages play an important role in erythropoiesis both in sickness and in health, and provide a potential targeted therapy for diseases such as polycythemia vera and beta-thalassemia in the future.
Collapse
Affiliation(s)
- Ran-Ran Zhang
- Diagnosis and Treatment Center of Pediatric Blood Diseases, Institute of Hematology and Blood Disease Hospital, Pecking Union Medical College, Chinese Academy of Medical Sciences, Tianjin 300020, China.
| | | |
Collapse
|
23
|
Rivella S. β-thalassemias: paradigmatic diseases for scientific discoveries and development of innovative therapies. Haematologica 2015; 100:418-30. [PMID: 25828088 DOI: 10.3324/haematol.2014.114827] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
β-thalassemias are monogenic disorders characterized by defective synthesis of the β-globin chain, one of the major components of adult hemoglobin. A large number of mutations in the β-globin gene or its regulatory elements have been associated with β-thalassemias. Due to the complexity of the regulation of the β-globin gene and the role of red cells in many physiological processes, patients can manifest a large spectrum of phenotypes, and clinical requirements vary from patient to patient. It is important to consider the major differences in the light of potential novel therapeutics. This review summarizes the main discoveries and mechanisms associated with the synthesis of β-globin and abnormal erythropoiesis, as well as current and novel therapies.
Collapse
Affiliation(s)
- Stefano Rivella
- Department of Pediatrics Hematology-Oncology Department of Cell and Developmental Biology Weill Cornell Medical College New York, NY, USA
| |
Collapse
|
24
|
Abstract
Macrophages play a critical role in iron homeostasis via their intimate association with developing and dying red cells. Central nurse macrophages promote erythropoiesis in the erythroblastic island niche. These macrophages make physical contact with erythroblasts, enabling signaling and the transfer of growth factors and possibly nutrients to the cells in their care. Human mature red cells have a lifespan of 120 days before they become senescent and again come into contact with macrophages. Phagocytosis of red blood cells is the main source of iron flux in the body, because heme must be recycled from approximately 270 billion hemoglobin molecules in each red cell, and roughly 2 million senescent red cells are recycled each second. Here we will review pathways for iron trafficking found at the macrophage-erythroid axis, with a focus on possible roles for the transport of heme in toto.
Collapse
|
25
|
Macrophages and iron trafficking at the birth and death of red cells. Blood 2015; 125:2893-7. [PMID: 25778532 DOI: 10.1182/blood-2014-12-567776] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/05/2015] [Indexed: 01/25/2023] Open
Abstract
Macrophages play a critical role in iron homeostasis via their intimate association with developing and dying red cells. Central nurse macrophages promote erythropoiesis in the erythroblastic island niche. These macrophages make physical contact with erythroblasts, enabling signaling and the transfer of growth factors and possibly nutrients to the cells in their care. Human mature red cells have a lifespan of 120 days before they become senescent and again come into contact with macrophages. Phagocytosis of red blood cells is the main source of iron flux in the body, because heme must be recycled from approximately 270 billion hemoglobin molecules in each red cell, and roughly 2 million senescent red cells are recycled each second. Here we will review pathways for iron trafficking found at the macrophage-erythroid axis, with a focus on possible roles for the transport of heme in toto.
Collapse
|
26
|
Fraser ST, Midwinter RG, Coupland LA, Kong S, Berger BS, Yeo JH, Andrade OC, Cromer D, Suarna C, Lam M, Maghzal GJ, Chong BH, Parish CR, Stocker R. Heme oxygenase-1 deficiency alters erythroblastic island formation, steady-state erythropoiesis and red blood cell lifespan in mice. Haematologica 2015; 100:601-10. [PMID: 25682599 DOI: 10.3324/haematol.2014.116368] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 02/10/2015] [Indexed: 12/15/2022] Open
Abstract
Heme oxygenase-1 is critical for iron recycling during red blood cell turnover, whereas its impact on steady-state erythropoiesis and red blood cell lifespan is not known. We show here that in 8- to 14-week old mice, heme oxygenase-1 deficiency adversely affects steady-state erythropoiesis in the bone marrow. This is manifested by a decrease in Ter-119(+)-erythroid cells, abnormal adhesion molecule expression on macrophages and erythroid cells, and a greatly diminished ability to form erythroblastic islands. Compared with wild-type animals, red blood cell size and hemoglobin content are decreased, while the number of circulating red blood cells is increased in heme oxygenase-1 deficient mice, overall leading to microcytic anemia. Heme oxygenase-1 deficiency increases oxidative stress in circulating red blood cells and greatly decreases the frequency of macrophages expressing the phosphatidylserine receptor Tim4 in bone marrow, spleen and liver. Heme oxygenase-1 deficiency increases spleen weight and Ter119(+)-erythroid cells in the spleen, although α4β1-integrin expression by these cells and splenic macrophages positive for vascular cell adhesion molecule 1 are both decreased. Red blood cell lifespan is prolonged in heme oxygenase-1 deficient mice compared with wild-type mice. Our findings suggest that while macrophages and relevant receptors required for red blood cell formation and removal are substantially depleted in heme oxygenase-1 deficient mice, the extent of anemia in these mice may be ameliorated by the prolonged lifespan of their oxidatively stressed erythrocytes.
Collapse
Affiliation(s)
- Stuart T Fraser
- Laboratory for Blood Cell Development, School of Medical Sciences (Physiology, Anatomy & Histology), Sydney Medical School, The University of Sydney, Australia
| | - Robyn G Midwinter
- Centre for Vascular Research, School of Medical Sciences (Pathology) and Bosch Institute, The University of Sydney, Australia
| | - Lucy A Coupland
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Stephanie Kong
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Birgit S Berger
- Centre for Vascular Research, School of Medical Sciences (Pathology) and Bosch Institute, The University of Sydney, Australia
| | - Jia Hao Yeo
- Laboratory for Blood Cell Development, School of Medical Sciences (Physiology, Anatomy & Histology), Sydney Medical School, The University of Sydney, Australia
| | - Osvaldo Cooley Andrade
- Laboratory for Blood Cell Development, School of Medical Sciences (Physiology, Anatomy & Histology), Sydney Medical School, The University of Sydney, Australia
| | - Deborah Cromer
- Complex Systems in Biology Group, Centre for Vascular Research, University of New South Wales, Kensington, Australia
| | - Cacang Suarna
- Centre for Vascular Research, School of Medical Sciences (Pathology) and Bosch Institute, The University of Sydney, Australia Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Magda Lam
- Centre for Vascular Research, School of Medical Sciences (Pathology) and Bosch Institute, The University of Sydney, Australia Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Ghassan J Maghzal
- Centre for Vascular Research, School of Medical Sciences (Pathology) and Bosch Institute, The University of Sydney, Australia Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia School of Medical Sciences, Faculty of Medicine, University of New South Wales, Kensington
| | - Beng H Chong
- Department of Medicine, St George Clinical School, University of New South Wales, Kogarah, Australia
| | - Christopher R Parish
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Roland Stocker
- Centre for Vascular Research, School of Medical Sciences (Pathology) and Bosch Institute, The University of Sydney, Australia Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Australia School of Medical Sciences, Faculty of Medicine, University of New South Wales, Kensington
| |
Collapse
|
27
|
|
28
|
Byon JCH, Padilla SM, Papayannopoulou T. Deletion of Dicer in late erythroid cells results in impaired stress erythropoiesis in mice. Exp Hematol 2014; 42:852-6.e1. [PMID: 24971698 DOI: 10.1016/j.exphem.2014.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/13/2014] [Accepted: 06/13/2014] [Indexed: 11/28/2022]
Abstract
MicroRNAs (miRNAs) have been shown to influence erythroid lineage commitment and differentiation; however, our knowledge of miRNA function in terminal erythropoiesis remains limited. To address this issue, we generated a novel animal model, where the miRNA-processing enzyme, Dicer, is selectively inactivated in erythropoietin receptor positive erythroid cells beginning with CFU-e/proerythroblast cells. This results in significant depletion of all miRNAs from the proerythroblast stage onwards, with one exception, miR-451, which is processed by Ago2 in a Dicer-independent manner. We observed that mature Dicer-dependent miRNAs, like miR-451, are dispensable under steady-state conditions, but these mutants have an impaired response to stress erythropoiesis, as demonstrated by a delay in recovery from anemia. This defect was specific to later maturing erythroid cells, as progenitor numbers were unaffected. In addition to generating a novel mouse model to study miRNA function in late erythroid cells, we conclude that miRNAs (both Dicer-dependent and independent) act primarily to regulate the optimal response to stress among late erythroid cells.
Collapse
Affiliation(s)
- John C H Byon
- Division of Hematology, University of Washington, Seattle, WA, USA
| | - Steven M Padilla
- Division of Hematology, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
29
|
Honda S, Arakawa S, Nishida Y, Yamaguchi H, Ishii E, Shimizu S. Ulk1-mediated Atg5-independent macroautophagy mediates elimination of mitochondria from embryonic reticulocytes. Nat Commun 2014; 5:4004. [PMID: 24895007 DOI: 10.1038/ncomms5004] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 04/29/2014] [Indexed: 01/28/2023] Open
Abstract
Macroautophagy is a highly conserved intracellular process responsible for the degradation of subcellular constituents. Macroautophagy was recently suggested to be involved in the removal of mitochondria from reticulocytes during the final stage of erythrocyte differentiation. Although Atg5 and Atg7 are indispensable for macroautophagy, their role in mitochondrial clearance remains controversial. We recently discovered that mammalian cells use conventional Atg5/Atg7-dependent macroautophagy as well as an alternative Unc-51-like kinase 1 (Ulk1)-dependent Atg5/Atg7-independent macroautophagy process. We hypothesized that the latter may be involved in mitochondrial clearance from reticulocytes during erythrocyte differentiation. Here we report that fetal definitive reticulocytes from Ulk1-deficient and Ulk1/Atg5 double-deficient mice retain their mitochondria, whereas the mitochondria are engulfed and digested within autophagic structures in wild-type and Atg5-deficient mice. Mitochondrial retention by Ulk1-deficient reticulocytes is far less marked in primitive and adult definitive reticulocytes. These data indicate that Ulk1-dependent Atg5-independent macroautophagy is the dominant process of mitochondrial clearance from fetal definitive reticulocytes.
Collapse
Affiliation(s)
- Shinya Honda
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Satoko Arakawa
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yuya Nishida
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Hirofumi Yamaguchi
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Eiichi Ishii
- Department of Pediatrics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Shigeomi Shimizu
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| |
Collapse
|
30
|
de Back DZ, Kostova EB, van Kraaij M, van den Berg TK, van Bruggen R. Of macrophages and red blood cells; a complex love story. Front Physiol 2014; 5:9. [PMID: 24523696 PMCID: PMC3906564 DOI: 10.3389/fphys.2014.00009] [Citation(s) in RCA: 197] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 01/06/2014] [Indexed: 12/13/2022] Open
Abstract
Macrophages tightly control the production and clearance of red blood cells (RBC). During steady state hematopoiesis, approximately 1010 RBC are produced per hour within erythroblastic islands in humans. In these erythroblastic islands, resident bone marrow macrophages provide erythroblasts with interactions that are essential for erythroid development. New evidence suggests that not only under homeostasis but also under stress conditions, macrophages play an important role in promoting erythropoiesis. Once RBC have matured, these cells remain in circulation for about 120 days. At the end of their life span, RBC are cleared by macrophages residing in the spleen and the liver. Current theories about the removal of senescent RBC and the essential role of macrophages will be discussed as well as the role of macrophages in facilitating the removal of damaged cellular content from the RBC. In this review we will provide an overview on the role of macrophages in the regulation of RBC production, maintenance and clearance. In addition, we will discuss the interactions between these two cell types during transfer of immune complexes and pathogens from RBC to macrophages.
Collapse
Affiliation(s)
- Djuna Z de Back
- Landsteiner Laboratory, Department of Blood Cell Research, Academic Medical Center, Sanquin Research, University of Amsterdam Amsterdam, Netherlands
| | - Elena B Kostova
- Landsteiner Laboratory, Department of Blood Cell Research, Academic Medical Center, Sanquin Research, University of Amsterdam Amsterdam, Netherlands
| | - Marian van Kraaij
- Landsteiner Laboratory, Department of Blood Cell Research, Academic Medical Center, Sanquin Research, University of Amsterdam Amsterdam, Netherlands
| | - Timo K van den Berg
- Landsteiner Laboratory, Department of Blood Cell Research, Academic Medical Center, Sanquin Research, University of Amsterdam Amsterdam, Netherlands
| | - Robin van Bruggen
- Landsteiner Laboratory, Department of Blood Cell Research, Academic Medical Center, Sanquin Research, University of Amsterdam Amsterdam, Netherlands
| |
Collapse
|
31
|
Ulyanova T, Padilla SM, Papayannopoulou T. Stage-specific functional roles of integrins in murine erythropoiesis. Exp Hematol 2014; 42:404-409.e4. [PMID: 24463276 DOI: 10.1016/j.exphem.2014.01.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 01/14/2014] [Indexed: 10/25/2022]
Abstract
When the erythroid integrins α5β1 and α4β1 were each deleted previously at the stem cell level, they yielded distinct physiologic responses to stress by affecting erythoid expansion and terminal differentiation or only the latter, respectively. To test at what stage of differentiation the integrin effects were exerted, we created mice with α4- or α5-integrin deletions only in erythroid cells and characterized them at homeostasis and after phenylhydrazine-induced hemolytic stress. Unlike our prior data, the phenotype of mice with α5-erythroid deletions was similar to controls, especially after stress. These outcomes seem to reconcile divergent prior views on the role of α5-integrin in erythropoiesis. By contrast, α4 integrins whether deleted early or late have a dominant effect on bone marrow retention of erythroblasts and on terminal erythroid maturation at homeostasis and after stress.
Collapse
Affiliation(s)
- Tatyana Ulyanova
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Steven M Padilla
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Thalia Papayannopoulou
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
32
|
A mouse model of adult-onset anaemia due to erythropoietin deficiency. Nat Commun 2013; 4:1950. [PMID: 23727690 DOI: 10.1038/ncomms2950] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 04/30/2013] [Indexed: 12/14/2022] Open
Abstract
Erythropoietin regulates erythropoiesis in a hypoxia-inducible manner. Here we generate inherited super-anaemic mice (ISAM) as a mouse model of adult-onset anaemia caused by erythropoietin deficiency. ISAM express erythropoietin in the liver but lack erythropoietin production in the kidney. Around weaning age, when the major erythropoietin-producing organ switches from the liver to the kidney, ISAM develop anaemia due to erythropoietin deficiency, which is curable by administration of recombinant erythropoietin. In ISAM severe chronic anaemia enhances transgenic green fluorescent protein and Cre expression driven by the complete erythropoietin-gene regulatory regions, which facilitates efficient labelling of renal erythropoietin-producing cells. We show that the majority of cortical and outer medullary fibroblasts have the innate potential to produce erythropoietin, and also reveal a new set of erythropoietin target genes. ISAM are a useful tool for the evaluation of erythropoiesis-stimulating agents and to trace the dynamics of erythropoietin-producing cells.
Collapse
|
33
|
Yang Y, Wang H, Chang KH, Qu H, Zhang Z, Xiong Q, Qi H, Cui P, Lin Q, Ruan X, Yang Y, Li Y, Shu C, Li Q, Wakeland EK, Yan J, Hu S, Fang X. Transcriptome dynamics during human erythroid differentiation and development. Genomics 2013; 102:431-441. [PMID: 24121002 DOI: 10.1016/j.ygeno.2013.09.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 09/22/2013] [Accepted: 09/30/2013] [Indexed: 10/26/2022]
Abstract
To explore the mechanisms controlling erythroid differentiation and development, we analyzed the genome-wide transcription dynamics occurring during the differentiation of human embryonic stem cells (HESCs) into the erythroid lineage and development of embryonic to adult erythropoiesis using high throughput sequencing technology. HESCs and erythroid cells at three developmental stages: ESER (embryonic), FLER (fetal), and PBER (adult) were analyzed. Our findings revealed that the number of expressed genes decreased during differentiation, whereas the total expression intensity increased. At each of the three transitions (HESCs-ESERs, ESERs-FLERs, and FLERs-PBERs), many differentially expressed genes were observed, which were involved in maintaining pluripotency, early erythroid specification, rapid cell growth, and cell-cell adhesion and interaction. We also discovered dynamic networks and their central nodes in each transition. Our study provides a fundamental basis for further investigation of erythroid differentiation and development, and has implications in using ESERs for transfusion product in clinical settings.
Collapse
Affiliation(s)
- Yadong Yang
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hai Wang
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Kai-Hsin Chang
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Hongzhu Qu
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhaojun Zhang
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Qian Xiong
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Heyuan Qi
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Peng Cui
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Qiang Lin
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiuyan Ruan
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yaran Yang
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yajuan Li
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Chang Shu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Quanzhen Li
- Department of Immunology & Microarray Core Facility, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Edward K Wakeland
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.,Department of Immunology & Microarray Core Facility, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jiangwei Yan
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Songnian Hu
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiangdong Fang
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
34
|
Ulyanova T, Jiang Y, Padilla SM, Papayannopoulou T. Erythroid cells generated in the absence of specific β1-integrin heterodimers accumulate reactive oxygen species at homeostasis and are unable to mount effective antioxidant defenses. Haematologica 2013; 98:1769-77. [PMID: 23812936 DOI: 10.3324/haematol.2013.087577] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
We have previously reported that β1(Δ/Δ) mice have a markedly impaired response to hemolytic stress, but the mechanisms of this were unclear. In the present study we explored in detail quantitative, phenotypic and functional aspects of erythropoiesis at homeostasis in a large number of animals for each of 3 murine models with specific β1 heterodimer integrin deficiencies. We found that, at homeostasis, β1-deficient mice have a modest uncompensated anemia with ineffective erythropoiesis and decreased red blood cell survival. Mice lacking only α4 integrins (α4β1/α4β7) do not share this phenotype. There is an increased tendency for reactive oxygen species accumulation in β1(Δ/Δ) erythroid cells with decreased anti-oxidant defenses at homeostasis which are exaggerated after stress. Furthermore, expansion of erythroid cells in spleen post-stress is dependent on α5β1, likely through mechanisms activating focal adhesion kinase complexes that are distinct from α4β1-mediated responses. In vivo inhibition of focal adhesion kinase activation partially recapitulates the β1(Δ/Δ) stress response. Mice lacking all α4 and β1 integrins (double knockouts) had, at homeostasis, the most severe phenotype with selective impairment of erythroid responses. The fact that integrins participate in mitigating stress in erythroid cells through redox activation of distinct signaling pathways by specific integrin heterodimers is a link that has not been appreciated until now.
Collapse
|
35
|
|
36
|
Spring FA, Griffiths RE, Mankelow TJ, Agnew C, Parsons SF, Chasis JA, Anstee DJ. Tetraspanins CD81 and CD82 facilitate α4β1-mediated adhesion of human erythroblasts to vascular cell adhesion molecule-1. PLoS One 2013; 8:e62654. [PMID: 23704882 PMCID: PMC3660455 DOI: 10.1371/journal.pone.0062654] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 03/25/2013] [Indexed: 11/28/2022] Open
Abstract
The proliferation and terminal differentiation of erythroid progenitors occurs in human bone marrow within erythroblastic islands, specialised structures consisting of a central macrophage surrounded by developing erythroid cells. Many cell-cell and cell-matrix adhesive interactions maintain and regulate the co-ordinated daily production of reticulocytes. Erythroid cells express only one integrin, α4β1, throughout differentiation, and its interactions with both macrophage Vascular Cell Adhesion Molecule-1 and with extracellular matrix fibronectin are critical for erythropoiesis. We observed that proerythroblasts expressed a broad tetraspanin phenotype, and investigated whether any tetraspanin could modulate integrin function. A specific association between α4β1 and CD81, CD82 and CD151 was demonstrated by confocal microscopy and co-immune precipitation. We observed that antibodies to CD81 and CD82 augmented adhesion of proerythroblasts to Vascular Cell Adhesion Molecule-1 but not to the fibronectin spliceoforms FnIII12-IIICS-15 and FnIII12–15. In contrast, different anti-CD151 antibodies augmented or inhibited adhesion of proerythroblasts to Vascular Cell Adhesion Molecule-1 and the fibronectin spliceoform FnIII12-IIICS-15 but not to FnIII12–15. These results strongly suggest that tetraspanins have a functional role in terminal erythropoiesis by modulating interactions of erythroblast α4β1 with both macrophages and extracellular matrix.
Collapse
Affiliation(s)
- Frances A Spring
- Bristol Institute for Transfusion Sciences, Bristol, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
37
|
Pepperell EE, Watt SM. A novel application for a 3-dimensional timelapse assay that distinguishes chemotactic from chemokinetic responses of hematopoietic CD133(+) stem/progenitor cells. Stem Cell Res 2013; 11:707-20. [PMID: 23727446 PMCID: PMC3744817 DOI: 10.1016/j.scr.2013.04.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 03/25/2013] [Accepted: 04/09/2013] [Indexed: 11/29/2022] Open
Abstract
Efficient homing/mobilization of human hematopoietic stem/progenitor cells to/from bone marrow niches enhances their therapeutic efficacy. Additionally, homing is dependent on cell source and may be modulated by prior ex vivo cell expansion. Here, we describe a novel application of a 3-dimensional time-lapse method for assessing trafficking of individual human cord blood CD133+ hematopoietic stem/progenitor cells in vitro, using the key chemokine CXCL12 as a paradigm. This new methodology allows distinction between chemotactic responses (displacement of center of mass and the forward migration index of the cells), and chemokinetic responses such as total cell path traveled in any direction (accumulated distance) and cell velocity in a 3-dimensional matrix. Other key advantages of this novel assay over existing assays include the ability to assess individual cell migration over times comparable to in vivo homing and rapid mobilization assays (18–24 h) and to directly compare the strength or response of individual hematopoietic progenitor cells to different or competing stimuli and small molecule inhibitors in a single assay prior to analyses in vivo. Importantly, using this method, our results demonstrate definitively that CXCL12 regulates the chemotactic responses of human cord blood CD133+ cells, but not their random migration or chemokinesis. Development of a novel 3-dimensional timelapse chemotaxis assay application. Measuring individual CD133+ HSPC trafficking towards chemokines in a 3D matrix. Chemotactic and chemokinetic responses reflecting homin kinetics in vivo. Comparative analysis of inhibitors or expansion on HSPC chemotaxis and chemokinesis. Definitive proof that CXCL12 regulates CD133+ HSPC chemotaxis but not chemokinesis.
Collapse
Affiliation(s)
- Emma E Pepperell
- Stem Cell Research Laboratory, NHS Blood and Transplant, Oxford, UK
| | | |
Collapse
|
38
|
Ramos P, Casu C, Gardenghi S, Breda L, Crielaard BJ, Guy E, Marongiu MF, Gupta R, Levine RL, Abdel-Wahab O, Ebert BL, Van Rooijen N, Ghaffari S, Grady RW, Giardina PJ, Rivella S. Macrophages support pathological erythropoiesis in polycythemia vera and β-thalassemia. Nat Med 2013; 19:437-45. [PMID: 23502961 PMCID: PMC3618568 DOI: 10.1038/nm.3126] [Citation(s) in RCA: 189] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 02/12/2013] [Indexed: 12/18/2022]
Abstract
Regulation of erythropoiesis is achieved by the integration of distinct signals. Among them, macrophages are emerging as erythropoietin-complementary regulators of erythroid development, particularly under stress conditions. We investigated the contribution of macrophages to physiological and pathological conditions of enhanced erythropoiesis. We used mouse models of induced anemia, polycythemia vera and β-thalassemia in which macrophages were chemically depleted. Our data indicate that macrophages contribute decisively to recovery from induced anemia, as well as the pathological progression of polycythemia vera and β-thalassemia, by modulating erythroid proliferation and differentiation. We validated these observations in primary human cultures, showing a direct impact of macrophages on the proliferation and enucleation of erythroblasts from healthy individuals and patients with polycythemia vera or β-thalassemia. The contribution of macrophages to stress and pathological erythropoiesis, which we have termed stress erythropoiesis macrophage-supporting activity, may have therapeutic implications.
Collapse
Affiliation(s)
- Pedro Ramos
- Department of Pediatrics, Division of Hematology-Oncology, Weill Cornell Medical College, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
CD169⁺ macrophages provide a niche promoting erythropoiesis under homeostasis and stress. Nat Med 2013; 19:429-36. [PMID: 23502962 PMCID: PMC3983996 DOI: 10.1038/nm.3057] [Citation(s) in RCA: 333] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 12/07/2012] [Indexed: 12/14/2022]
Abstract
The role of macrophages in erythropoiesis was suggested several decades ago with the description of “erythroblastic islands” in the bone marrow (BM) composed of a central macrophage surrounded by developing erythroblasts. However, the in vivo role of macrophages in erythropoiesis under homeostasis or disease remains unclear. Specific depletion of CD169+ macrophages markedly reduced erythroblasts in the BM but did not result in overt anemia under homeostasis likely due to concomitant alterations in RBC clearance. However, CD169+ macrophage depletion significantly impaired erythropoietic recovery from hemolytic anemia, acute blood loss and myeloablation. Furthermore, macrophage depletion normalized the erythroid compartment in a JAK2V617F-driven murine model of polycythemia vera (PV), suggesting that erythropoiesis in PV, unexpectedly, remains under the control of macrophages in the BM and splenic microenvironments. These data indicate that CD169+ macrophages promote late erythroid maturation and that modulation of the macrophage compartment represents a novel strategy to treat erythropoietic disorders.
Collapse
|
40
|
EPO-mediated expansion of late-stage erythroid progenitors in the bone marrow initiates recovery from sublethal radiation stress. Blood 2012; 120:2501-11. [PMID: 22889760 DOI: 10.1182/blood-2011-11-394304] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Erythropoiesis is a robust process of cellular expansion and maturation occurring in murine bone marrow and spleen. We previously determined that sublethal irradiation, unlike bleeding or hemolysis, depletes almost all marrow and splenic erythroblasts but leaves peripheral erythrocytes intact. To better understand the erythroid stress response, we analyzed progenitor, precursor, and peripheral blood compartments of mice post-4 Gy total body irradiation. Erythroid recovery initiates with rapid expansion of late-stage erythroid progenitors-day 3 burst-forming units and colony-forming units, associated with markedly increased plasma erythropoietin (EPO). Although initial expansion of late-stage erythroid progenitors is dependent on EPO, this cellular compartment becomes sharply down-regulated despite elevated EPO levels. Loss of EPO-responsive progenitors is associated temporally with a wave of maturing erythroid precursors in marrow and with emergence of circulating erythroid progenitors and subsequent reestablishment of splenic erythropoiesis. These circulating progenitors selectively engraft and mature in irradiated spleen after short-term transplantation, supporting the concept that bone marrow erythroid progenitors migrate to spleen. We conclude that sublethal radiation is a unique model of endogenous stress erythropoiesis, with specific injury to the extravascular erythron, expansion and maturation of EPO-responsive late-stage progenitors exclusively in marrow, and subsequent reseeding of extramedullary sites.
Collapse
|
41
|
|
42
|
Ripich T, Jessberger R. SWAP-70 regulates erythropoiesis by controlling α4 integrin. Haematologica 2011; 96:1743-52. [PMID: 21880631 DOI: 10.3324/haematol.2011.050468] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED Background The regulation of normal and stress-induced erythropoiesis is incompletely understood. Integrin-dependent adhesion plays important roles in erythropoiesis, but how integrins are regulated during erythropoiesis remains largely unknown. DESIGN AND METHODS To obtain novel insights into the regulation of erythropoiesis, we used cellular and molecular approaches to analyze the role of SWAP-70 and the control of integrins through SWAP-70. In addition, mice deficient for this protein were investigated under normal and erythropoietic stress conditions. RESULTS We show that SWAP-70, a protein involved in cytoskeletal F-actin rearrangements and integrin regulation in mast cells, is expressed in hematopoietic stem cells and myeloid-erythroid precursors. Although Swap-70(-/-) mice are not anemic, erythroblastic differentiation is perturbed, and SWAP-70 is required for an efficient erythropoietic stress response to acute anemia and for erythropoietic recovery after bone marrow transplantation in irradiated mice. SWAP-70 deficiency impairs colony-forming unit erythroid development, while burst-forming unit erythroid development is normal, and significantly affects development of late erythroblasts in the spleen and bone marrow. The α(4) integrin is constitutively hyper-activated in Swap-70(-/-) colony-forming unit erythroid cells, which hyper-adhere to fibronectin. Blocking α(4) and β(1) integrin chains in vivo restored erythroblastic differentiation and the erythropoietic stress response in Swap-70(-/-) mice. Conclusions Our study reveals that SWAP-70 is a novel regulator of integrin-mediated red blood cell development and stress-induced erythropoiesis.
Collapse
Affiliation(s)
- Tatsiana Ripich
- Institute of Physiological Chemistry, Dresden University of Technology Fiedlerstr. 42, Dresden, Germany
| | | |
Collapse
|
43
|
c-Maf rules the island. Blood 2011; 118:1192-3. [DOI: 10.1182/blood-2011-06-359752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
44
|
|