151
|
The VP1u Receptor Restricts Parvovirus B19 Uptake to Permissive Erythroid Cells. Viruses 2016; 8:v8100265. [PMID: 27690083 PMCID: PMC5086601 DOI: 10.3390/v8100265] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 09/19/2016] [Accepted: 09/20/2016] [Indexed: 12/20/2022] Open
Abstract
Parvovirus B19 (B19V) is a small non-enveloped virus and known as the causative agent for the mild childhood disease erythema infectiosum. B19V has an extraordinary narrow tissue tropism, showing only productive infection in erythroid precursor cells in the bone marrow. We recently found that the viral protein 1 unique region (VP1u) contains an N-terminal receptor-binding domain (RBD), which mediates the uptake of the virus into cells of the erythroid lineage. To further investigate the role of the RBD in connection with a B19V-unrelated capsid, we chemically coupled the VP1u of B19V to the bacteriophage MS2 capsid and tested the internalization capacity of the bioconjugate on permissive cells. In comparison, we studied the cellular uptake and infection of B19V along the erythroid differentiation. The results showed that the MS2-VP1u bioconjugate mimicked the specific internalization of the native B19V into erythroid precursor cells, which further coincides with the restricted infection profile. The successful mimicry of B19V uptake demonstrates that the RBD in the VP1u is sufficient for the endocytosis of the viral capsid. Furthermore, the recombinant VP1u competed with B19V uptake into permissive cells, thus excluding a significant alternative uptake mechanism by other receptors. Strikingly, the VP1u receptor appeared to be expressed only on erythropoietin-dependent erythroid differentiation stages that also provide the necessary intracellular factors for a productive infection. Taken together, these findings suggest that the VP1u binds to a yet-unknown erythroid-specific cellular receptor and thus restricts the virus entry to permissive cells.
Collapse
|
152
|
Polyurethane scaffolds seeded with CD34(+) cells maintain early stem cells whilst also facilitating prolonged egress of haematopoietic progenitors. Sci Rep 2016; 6:32149. [PMID: 27573994 PMCID: PMC5004174 DOI: 10.1038/srep32149] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/03/2016] [Indexed: 12/15/2022] Open
Abstract
We describe a 3D erythroid culture system that utilises a porous polyurethane (PU) scaffold to mimic the compartmentalisation found in the bone marrow. PU scaffolds seeded with peripheral blood CD34+ cells exhibit a remarkable reproducibility of egress, with an increased output when directly compared to human bone scaffolds over 28 days. Immunofluorescence demonstrated the persistence of CD34+ cells within the scaffolds for the entirety of the culture. To characterise scaffold outputs, we designed a flow cytometry panel that utilises surface marker expression observed in standard 2D erythroid and megakaryocyte cultures. This showed that the egress population is comprised of haematopoietic progenitor cells (CD36+GPA−/low). Control cultures conducted in parallel but in the absence of a scaffold were also generally maintained for the longevity of the culture albeit with a higher level of cell death. The harvested scaffold egress can also be expanded and differentiated to the reticulocyte stage. In summary, PU scaffolds can behave as a subtractive compartmentalised culture system retaining and allowing maintenance of the seeded “CD34+ cell” population despite this population decreasing in amount as the culture progresses, whilst also facilitating egress of increasingly differentiated cells.
Collapse
|
153
|
|
154
|
McIver SC, Katsumura KR, Davids E, Liu P, Kang YA, Yang D, Bresnick EH. Exosome complex orchestrates developmental signaling to balance proliferation and differentiation during erythropoiesis. eLife 2016; 5. [PMID: 27543448 PMCID: PMC5040589 DOI: 10.7554/elife.17877] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/18/2016] [Indexed: 12/11/2022] Open
Abstract
Since the highly conserved exosome complex mediates the degradation and processing of multiple classes of RNAs, it almost certainly controls diverse biological processes. How this post-transcriptional RNA-regulatory machine impacts cell fate decisions and differentiation is poorly understood. Previously, we demonstrated that exosome complex subunits confer an erythroid maturation barricade, and the erythroid transcription factor GATA-1 dismantles the barricade by transcriptionally repressing the cognate genes. While dissecting requirements for the maturation barricade in Mus musculus, we discovered that the exosome complex is a vital determinant of a developmental signaling transition that dictates proliferation/amplification versus differentiation. Exosome complex integrity in erythroid precursor cells ensures Kit receptor tyrosine kinase expression and stem cell factor/Kit signaling, while preventing responsiveness to erythropoietin-instigated signals that promote differentiation. Functioning as a gatekeeper of this developmental signaling transition, the exosome complex controls the massive production of erythroid cells that ensures organismal survival in homeostatic and stress contexts. DOI:http://dx.doi.org/10.7554/eLife.17877.001 Red blood cells supply an animal’s tissues with the oxygen they need to survive. These cells circulate for a certain amount of time before they die. To replenish the red blood cells that are lost, first a protein called stem cell factor (SCF) instructs stem cells and precursor cells to proliferate, and a second protein, known as erythropoietin, then signals to these cells to differentiate into mature red blood cells. It is important to maintain this balance between these two processes because too much proliferation can lead to cancer while too much differentiation will exhaust the supply of stem cells. Previous work has shown that a collection of proteins called the exosome complex can block steps leading towards mature red blood cells. The exosome complex controls several processes within cells by modifying or degrading a variety of messenger RNAs, the molecules that serve as intermediates between DNA and protein. However, it was not clear how the exosome complex sets up the differentiation block and whether it is somehow connected to the signaling from SCF and erythropoietin. McIver et al. set out to address this issue by isolating precursor cells with the potential to become red blood cells from mouse fetal livers and experimentally reducing the levels of the exosome complex. The experiments showed that these cells were no longer able to respond when treated with SCF in culture, whereas the control cells responded as normal. Further experiments showed that cells with less of the exosome complex also made less of a protein named Kit. Normally, SCF interacts with Kit to instruct cells to multiply. Lastly, although the experimental cells could no longer respond to these proliferation signals, they could react to erythropoietin, which promotes differentiation. Thus, normal levels of the exosome complex keep the delicate balance between proliferation and differentiation, which is crucial to the development of red blood cells. In future, it will be important to study the exosome complex in living mice and in human cells, and to see whether it also controls other signaling pathways. Furthermore, it is worth exploring whether this new knowledge can help efforts to produce red blood cells on an industrial scale, which could then be used to treat patients with conditions such as anemia. DOI:http://dx.doi.org/10.7554/eLife.17877.002
Collapse
Affiliation(s)
- Skye C McIver
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, United States.,UW-Madison Blood Research Program, University of Wisconsin School of Medicine and Public Health, Madison, United States.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, United States
| | - Koichi R Katsumura
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, United States.,UW-Madison Blood Research Program, University of Wisconsin School of Medicine and Public Health, Madison, United States.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, United States
| | - Elsa Davids
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, United States.,UW-Madison Blood Research Program, University of Wisconsin School of Medicine and Public Health, Madison, United States.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, United States
| | - Peng Liu
- Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, United States.,Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, United States
| | - Yoon-A Kang
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, United States.,UW-Madison Blood Research Program, University of Wisconsin School of Medicine and Public Health, Madison, United States.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, United States
| | - David Yang
- Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, United States
| | - Emery H Bresnick
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, United States.,UW-Madison Blood Research Program, University of Wisconsin School of Medicine and Public Health, Madison, United States.,Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, United States
| |
Collapse
|
155
|
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]
|
156
|
Liang R, Ghaffari S. Advances in understanding the mechanisms of erythropoiesis in homeostasis and disease. Br J Haematol 2016; 174:661-73. [PMID: 27442953 DOI: 10.1111/bjh.14194] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Anaemia or decreased blood haemoglobin is the most common blood disorder often characterized by reduced red blood cell (RBC) numbers. RBCs are produced from differentiation and commitment of haematopoietic stem cells to the erythroid lineage by a process called erythropoiesis. Coordination of erythropoietin receptor signalling with several erythroid transcription factors including GATA1 is essential for this process. A number of additional players that are critical for RBC production have been identified in recent years. Major technological advances, such as the development of RNA interference, genetically modified animals, including zebrafish, and imaging flow cytometry have led to these discoveries; the emergence of -omics approaches in combination with the optimization of ex vivo erythroid cultures have also produced a more comprehensive understanding of erythropoiesis. Here we summarize studies describing novel regulators of erythropoiesis that modulate erythroid cell production in the context of human erythroid disorders involving hypoxia, iron regulation, immune-related molecules, and the transcription factor FOXO3.
Collapse
Affiliation(s)
- Raymond Liang
- Department of Developmental & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Developmental and Stem Cell Biology Multidisciplinary Training Area, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Saghi Ghaffari
- Department of Developmental & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Developmental and Stem Cell Biology Multidisciplinary Training Area, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
157
|
Bouchnita A, Eymard N, Moyo TK, Koury MJ, Volpert V. Bone marrow infiltration by multiple myeloma causes anemia by reversible disruption of erythropoiesis. Am J Hematol 2016; 91:371-8. [PMID: 26749142 DOI: 10.1002/ajh.24291] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 12/29/2015] [Indexed: 12/20/2022]
Abstract
Multiple myeloma (MM) infiltrates bone marrow and causes anemia by disrupting erythropoiesis, but the effects of marrow infiltration on anemia are difficult to quantify. Marrow biopsies of newly diagnosed MM patients were analyzed before and after four 28-day cycles of non-erythrotoxic remission induction chemotherapy. Complete blood cell counts and serum paraprotein concentrations were measured at diagnosis and before each chemotherapy cycle. At diagnosis, marrow area infiltrated by myeloma correlated negatively with hemoglobin, erythrocytes, and marrow erythroid cells. After successful chemotherapy, patients with less than 30% myeloma infiltration at diagnosis had no change in these parameters, whereas patients with more than 30% myeloma infiltration at diagnosis increased all three parameters. Clinical data were used to develop mathematical models of the effects of myeloma infiltration on the marrow niches of terminal erythropoiesis, the erythroblastic islands (EBIs). A hybrid discrete-continuous model of erythropoiesis based on EBI structure/function was extended to sections of marrow containing multiple EBIs. In the model, myeloma cells can kill erythroid cells by physically destroying EBIs and by producing proapoptotic cytokines. Following chemotherapy, changes in serum paraproteins as measures of myeloma cells and changes in erythrocyte numbers as measures of marrow erythroid cells allowed modeling of myeloma cell death and erythroid cell recovery, respectively. Simulations of marrow infiltration by myeloma and treatment with non-erythrotoxic chemotherapy demonstrate that myeloma-mediated destruction and subsequent reestablishment of EBIs and expansion of erythroid cell populations in EBIs following chemotherapy provide explanations for anemia development and its therapy-mediated recovery in MM patients.
Collapse
Affiliation(s)
- Anass Bouchnita
- Institut Camille Jordan, UMR 5208 CNRS, University Lyon 1; Villeurbanne 69622 France
| | - Nathalie Eymard
- Institut Camille Jordan, UMR 5208 CNRS, University Lyon 1; Villeurbanne 69622 France
| | - Tamara K. Moyo
- Division of Hematology/Oncology; Department of Medicine; Vanderbilt University Medical Center; Nashville Tennessee
| | - Mark J. Koury
- Division of Hematology/Oncology; Department of Medicine; Vanderbilt University Medical Center; Nashville Tennessee
| | - Vitaly Volpert
- Institut Camille Jordan, UMR 5208 CNRS, University Lyon 1; Villeurbanne 69622 France
- INRIA Team Dracula, INRIA Antenne Lyon La Doua, Villeurbanne 69603, France, and European Institute of Systems Biology and Medicine; Lyon 69007 France
| |
Collapse
|
158
|
Barminko J, Reinholt B, Baron MH. Development and differentiation of the erythroid lineage in mammals. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 58:18-29. [PMID: 26709231 PMCID: PMC4775370 DOI: 10.1016/j.dci.2015.12.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 12/15/2015] [Accepted: 12/15/2015] [Indexed: 05/02/2023]
Abstract
The red blood cell (RBC) is responsible for performing the highly specialized function of oxygen transport, making it essential for survival during gestation and postnatal life. Establishment of sufficient RBC numbers, therefore, has evolved to be a major priority of the postimplantation embryo. The "primitive" erythroid lineage is the first to be specified in the developing embryo proper. Significant resources are dedicated to producing RBCs throughout gestation. Two transient and morphologically distinct waves of hematopoietic progenitor-derived erythropoiesis are observed in development before hematopoietic stem cells (HSCs) take over to produce "definitive" RBCs in the fetal liver. Toward the end of gestation, HSCs migrate to the bone marrow, which becomes the primary site of RBC production in the adult. Erythropoiesis is regulated at various stages of erythroid cell maturation to ensure sufficient production of RBCs in response to physiological demands. Here, we highlight key aspects of mammalian erythroid development and maturation as well as differences among the primitive and definitive erythroid cell lineages.
Collapse
Affiliation(s)
- Jeffrey Barminko
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brad Reinholt
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Margaret H Baron
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of The Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
159
|
Pichon A, Jeton F, El Hasnaoui-Saadani R, Hagström L, Launay T, Beaudry M, Marchant D, Quidu P, Macarlupu JL, Favret F, Richalet JP, Voituron N. Erythropoietin and the use of a transgenic model of erythropoietin-deficient mice. HYPOXIA 2016; 4:29-39. [PMID: 27800506 PMCID: PMC5085313 DOI: 10.2147/hp.s83540] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Despite its well-known role in red blood cell production, it is now accepted that erythropoietin (Epo) has other physiological functions. Epo and its receptors are expressed in many tissues, such as the brain and heart. The presence of Epo/Epo receptors in these organs suggests other roles than those usually assigned to this protein. Thus, the aim of this review is to describe the effects of Epo deficiency on adaptation to normoxic and hypoxic environments and to suggest a key role of Epo on main physiological adaptive functions. Our original model of Epo-deficient (Epo-TAgh) mice allowed us to improve our knowledge of the possible role of Epo in O2 homeostasis. The use of anemic transgenic mice revealed Epo as a crucial component of adaptation to hypoxia. Epo-TAgh mice survive well in hypoxic conditions despite low hematocrit. Furthermore, Epo plays a key role in neural control of ventilatory acclimatization and response to hypoxia, in deformability of red blood cells, in cerebral and cardiac angiogenesis, and in neuro- and cardioprotection.
Collapse
Affiliation(s)
- Aurélien Pichon
- Laboratory "Hypoxia and Lung" EA 2363, University Paris 13, Sorbonne Paris Cité, Bobigny Cedex; Laboratory of Excellence GR-Ex, Paris; Laboratory MOVE EA 6314, FSS, Poitiers University, Poitiers, France
| | - Florine Jeton
- Laboratory "Hypoxia and Lung" EA 2363, University Paris 13, Sorbonne Paris Cité, Bobigny Cedex; Laboratory of Excellence GR-Ex, Paris
| | | | - Luciana Hagström
- Laboratório Interdisciplinar de Biociências, Universidade de Brasília, Brasília, Brazil
| | - Thierry Launay
- Unité de Biologie Intégrative des Adaptations à l'Exercice, University Paris Saclay and Genopole , University Sorbonne-Paris-Cité, Paris, France
| | - Michèle Beaudry
- Laboratory "Hypoxia and Lung" EA 2363, University Paris 13, Sorbonne Paris Cité, Bobigny Cedex
| | - Dominique Marchant
- Laboratory "Hypoxia and Lung" EA 2363, University Paris 13, Sorbonne Paris Cité, Bobigny Cedex
| | - Patricia Quidu
- Laboratory "Hypoxia and Lung" EA 2363, University Paris 13, Sorbonne Paris Cité, Bobigny Cedex
| | - Jose-Luis Macarlupu
- High Altitude Unit, Laboratories for Research and Development, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Fabrice Favret
- Laboratory "Mitochondrie, Stress Oxydant et Protection Musculaire" EA 3072, University of Strasbourg, Strasbourg, France
| | - Jean-Paul Richalet
- Laboratory "Hypoxia and Lung" EA 2363, University Paris 13, Sorbonne Paris Cité, Bobigny Cedex; Laboratory of Excellence GR-Ex, Paris
| | - Nicolas Voituron
- Laboratory "Hypoxia and Lung" EA 2363, University Paris 13, Sorbonne Paris Cité, Bobigny Cedex; Laboratory of Excellence GR-Ex, Paris
| |
Collapse
|
160
|
Abstract
SCL, a transcription factor of the basic helix-loop-helix family, is a master regulator of hematopoiesis. Scl specifies lateral plate mesoderm to a hematopoietic fate and establishes boundaries by inhibiting the cardiac lineage. A combinatorial interaction between Scl and Vegfa/Flk1 sets in motion the first wave of primitive hematopoiesis. Subsequently, definitive hematopoietic stem cells (HSCs) emerge from the embryo proper via an endothelial-to-hematopoietic transition controlled by Runx1, acting with Scl and Gata2. Past this stage, Scl in steady state HSCs is redundant with Lyl1, a highly homologous factor. However, Scl is haploinsufficient in stress response, when a rare subpopulation of HSCs with very long term repopulating capacity is called into action. SCL activates transcription by recruiting a core complex on DNA that necessarily includes E2A/HEB, GATA1-3, LIM-only proteins LMO1/2, LDB1, and an extended complex comprising ETO2, RUNX1, ERG, or FLI1. These interactions confer multifunctionality to a complex that can control cell proliferation in erythroid progenitors or commitment to terminal differentiation through variations in single component. Ectopic SCL and LMO1/2 expression in immature thymocytes activates of a stem cell gene network and reprogram cells with a finite lifespan into self-renewing preleukemic stem cells (pre-LSCs), an initiating event in T-cell acute lymphoblastic leukemias. Interestingly, fate conversion of fibroblasts to hematoendothelial cells requires not only Scl and Lmo2 but also Gata2, Runx1, and Erg, indicating a necessary collaboration between these transcription factors for hematopoietic reprogramming. Nonetheless, full reprogramming into self-renewing multipotent HSCs may require additional factors and most likely, a permissive microenvironment.
Collapse
Affiliation(s)
- T Hoang
- Laboratory of Hematopoiesis and Leukemia, Institute of Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC, Canada.
| | - J A Lambert
- Laboratory of Hematopoiesis and Leukemia, Institute of Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC, Canada
| | - R Martin
- Laboratory of Hematopoiesis and Leukemia, Institute of Research in Immunology and Cancer (IRIC), University of Montreal, Montreal, QC, Canada
| |
Collapse
|
161
|
Nandakumar SK, Ulirsch JC, Sankaran VG. Advances in understanding erythropoiesis: evolving perspectives. Br J Haematol 2016; 173:206-18. [PMID: 26846448 DOI: 10.1111/bjh.13938] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Red blood cells (RBCs) are generated from haematopoietic stem and progenitor cells (HSPCs) through the step-wise process of differentiation known as erythropoiesis. In this review, we discuss our current understanding of erythropoiesis and highlight recent advances in this field. During embryonic development, erythropoiesis occurs in three distinct waves comprising first, the yolk sac-derived primitive RBCs, followed sequentially by the erythro-myeloid progenitor (EMP) and HSPC-derived definitive RBCs. Recent work has highlighted the complexity and variability that may exist in the hierarchical arrangement of progenitors responsible for erythropoiesis. Using recently defined cell surface markers, it is now possible to enrich for erythroid progenitors and precursors to a much greater extent than has been possible before. While a great deal of knowledge has been gained on erythropoiesis from model organisms, our understanding of this process is currently being refined through human genetic studies. Genes mutated in erythroid disorders can now be identified more rapidly by the use of next-generation sequencing techniques. Genome-wide association studies on erythroid traits in healthy populations have also revealed new modulators of erythropoiesis. All of these recent developments have significant promise not only for increasing our understanding of erythropoiesis, but also for improving our ability to intervene when RBC production is perturbed in disease.
Collapse
Affiliation(s)
- Satish K Nandakumar
- Division of Hematology/Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jacob C Ulirsch
- Division of Hematology/Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vijay G Sankaran
- Division of Hematology/Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
162
|
Heir P, Srikumar T, Bikopoulos G, Bunda S, Poon BP, Lee JE, Raught B, Ohh M. Oxygen-dependent Regulation of Erythropoietin Receptor Turnover and Signaling. J Biol Chem 2016; 291:7357-72. [PMID: 26846855 DOI: 10.1074/jbc.m115.694562] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Indexed: 01/29/2023] Open
Abstract
von Hippel-Lindau (VHL) disease is a rare familial cancer predisposition syndrome caused by a loss or mutation in a single gene,VHL, but it exhibits a wide phenotypic variability that can be categorized into distinct subtypes. The phenotypic variability has been largely argued to be attributable to the extent of deregulation of the α subunit of hypoxia-inducible factor α, a well established target of VHL E3 ubiquitin ligase, ECV (Elongins/Cul2/VHL). Here, we show that erythropoietin receptor (EPOR) is hydroxylated on proline 419 and 426 via prolyl hydroxylase 3. EPOR hydroxylation is required for binding to the β domain of VHL and polyubiquitylation via ECV, leading to increased EPOR turnover. In addition, several type-specific VHL disease-causing mutants, including those that have retained proper binding and regulation of hypoxia-inducible factor α, showed a severe defect in binding prolyl hydroxylated EPOR peptides. These results identify EPOR as the secondbona fidehydroxylation-dependent substrate of VHL that potentially influences oxygen homeostasis and contributes to the complex genotype-phenotype correlation in VHL disease.
Collapse
Affiliation(s)
- Pardeep Heir
- From the Departments of Laboratory Medicine and Pathobiology and
| | - Tharan Srikumar
- the Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada
| | | | - Severa Bunda
- From the Departments of Laboratory Medicine and Pathobiology and
| | - Betty P Poon
- From the Departments of Laboratory Medicine and Pathobiology and Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8 and
| | - Jeffrey E Lee
- From the Departments of Laboratory Medicine and Pathobiology and
| | - Brian Raught
- the Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada
| | - Michael Ohh
- From the Departments of Laboratory Medicine and Pathobiology and Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8 and
| |
Collapse
|
163
|
Dey S, Li X, Teng R, Alnaeeli M, Chen Z, Rogers H, Noguchi CT. Erythropoietin regulates POMC expression via STAT3 and potentiates leptin response. J Mol Endocrinol 2016; 56:55-67. [PMID: 26563310 PMCID: PMC4692057 DOI: 10.1530/jme-15-0171] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/12/2015] [Indexed: 01/02/2023]
Abstract
The arcuate nucleus of the hypothalamus is essential for metabolic homeostasis and responds to leptin by producing several neuropeptides including proopiomelanocortin (POMC). We previously reported that high-dose erythropoietin (Epo) treatment in mice while increasing hematocrit reduced body weight, fat mass, and food intake and increased energy expenditure. Moreover, we showed that mice with Epo receptor (EpoR) restricted to erythroid cells (ΔEpoRE) became obese and exhibited decreased energy expenditure. Epo/EpoR signaling was found to promote hypothalamus POMC expression independently from leptin. Herein we used WT and ΔEpoRE mice and hypothalamus-derived neural culture system to study the signaling pathways activated by Epo in POMC neurons. We show that Epo stimulation activated STAT3 signaling and upregulated POMC expression in WT neural cultures. ΔEpoRE mice hypothalamus showed reduced POMC levels and lower STAT3 phosphorylation, with and without leptin treatment, compared to in vivo and ex vivo WT controls. Collectively, these data show that Epo regulates hypothalamus POMC expression via STAT3 activation, and provide a previously unrecognized link between Epo and leptin response.
Collapse
|
164
|
Kim MK, Shin H, Park KS, Kim H, Park J, Kim K, Nam J, Choo H, Chong Y. Benzimidazole Derivatives as Potent JAK1-Selective Inhibitors. J Med Chem 2015; 58:7596-602. [PMID: 26351728 DOI: 10.1021/acs.jmedchem.5b01263] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The Janus kinase (JAK) family comprises four members (JAK1, JAK2, JAK3, and Tyk2) that play a key role in mediating cytokine receptor signaling. JAK inhibition thus modulates cytokine-mediated effects. In particular, selective inhibition of JAK1 or JAK3 may provide an efficient therapeutic agent for the treatment of inflammatory diseases, with minimized side effects. In this study, as part of our continued efforts to develop a selective JAK1 inhibitor, a series of 1,2-disubstituted benzimidazole-5-carboxamide derivatives was prepared and their inhibitory activities against all four JAK isozymes were evaluated. A clear structure-activity relationship was observed with respect to JAK1 selectivity; this highlighted the importance of hydrogen bond donors at both N(1) and R2 positions located within a specific distance from the benzimidazole core. One of the synthesized compounds, 1-(2-aminoethyl)-2-(piperidin-4-yl)-1H-benzo[d]imidazole-5-carboxamide (5c), showed remarkable JAK1 selectivity (63-fold vs JAK2, 25-fold vs JAK3, and 74-fold vs Tyk2). Molecular docking revealed that the 2-aminoethyl and piperidin-4-yl substituents of 5c function as probes to differentiate the ATP-binding site of JAK1 from that of JAK2, resulting in preferential JAK1 binding. A kinase panel assay confirmed the JAK1 selectivity of 5c, which showed no appreciable inhibitory activity against 26 other protein kinases at 10 μM.
Collapse
Affiliation(s)
- Mi Kyoung Kim
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University , 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| | - Heerim Shin
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University , 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| | - Kwang-su Park
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University , 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| | - Hyungmi Kim
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University , 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| | - Jiseon Park
- R&D Center, Jeil Pharmaceutical Co., Ltd. 7 Cheongganggachangro, Baegam-myun, Cheoin-gu, Yongin-City, Kyunggi-Do 17172, Korea
| | - Kangjeon Kim
- R&D Center, Jeil Pharmaceutical Co., Ltd. 7 Cheongganggachangro, Baegam-myun, Cheoin-gu, Yongin-City, Kyunggi-Do 17172, Korea
| | - Joonwoo Nam
- R&D Center, Jeil Pharmaceutical Co., Ltd. 7 Cheongganggachangro, Baegam-myun, Cheoin-gu, Yongin-City, Kyunggi-Do 17172, Korea
| | - Hyunah Choo
- Center for Neuro-Medicine, Korea Institute of Science and Technology , 5, Hwarang-ro 14-gil, Seoungbuk-gu, Seoul 02792, Korea.,Department of Biological Chemistry, University of Science and Technology , 217 Gajeong-ro, Youseong-gu, Daejeon 34113, Korea
| | - Youhoon Chong
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University , 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Korea
| |
Collapse
|
165
|
Divoky V, Song J, Horvathova M, Kralova B, Votavova H, Prchal JT, Yoon D. Delayed hemoglobin switching and perinatal neocytolysis in mice with gain-of-function erythropoietin receptor. J Mol Med (Berl) 2015; 94:597-608. [PMID: 26706855 DOI: 10.1007/s00109-015-1375-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 11/18/2015] [Accepted: 11/24/2015] [Indexed: 11/26/2022]
Abstract
UNLABELLED Mutations of the truncated cytoplasmic domain of human erythropoietin receptor (EPOR) result in gain-of-function of erythropoietin (EPO) signaling and a dominantly inherited polycythemia, primary familial and congenital polycythemia (PFCP). We interrogated the unexplained transient absence of perinatal polycythemia observed in PFCP patients using an animal model of PFCP to examine its erythropoiesis during embryonic, perinatal, and early postnatal periods. In this model, we replaced the murine EpoR gene (mEpoR) with the wild-type human EPOR (wtHEPOR) or mutant human EPOR gene (mtHEPOR) and previously reported that the gain-of-function mtHEPOR mice become polycythemic at 3~6 weeks of age, but not at birth, similar to the phenotype of PFCP patients. In contrast, wtHEPOR mice had sustained anemia. We report that the mtHEPOR fetuses are polycythemic, but their polycythemia is abrogated in the perinatal period and reappears again at 3 weeks after birth. mtHEPOR fetuses have a delayed switch from primitive to definitive erythropoiesis, augmented erythropoietin signaling, and prolonged Stat5 phosphorylation while the wtHEPOR fetuses are anemic. Our study demonstrates the in vivo effect of excessive EPO/EPOR signaling on developmental erythropoiesis switch and describes that fetal polycythemia in this PFCP model is followed by transient correction of polycythemia in perinatal life associated with low Epo levels and increased exposure of erythrocytes' phosphatidylserine. We suggest that neocytolysis contributes to the observed perinatal correction of polycythemia in mtHEPOR newborns as embryos leaving the hypoxic uterus are exposed to normoxia at birth. KEY MESSAGE Human gain-of-function EPOR (mtHEPOR) causes fetal polycythemia in knock-in mice. Wild-type human EPOR causes fetal anemia in knock-in mouse model. mtHEPOR mice have delayed switch from primitive to definitive erythropoiesis. Polycythemia of mtHEPOR mice is transiently corrected in perinatal life. mtHEPOR newborns have low Epo and increased exposure of erythrocytes' phosphatidylserine.
Collapse
Affiliation(s)
- Vladimir Divoky
- Department of Biology, Faculty of Medicine and Dentistry, Palacky University, 775 15, Olomouc, Czech Republic
| | - Jihyun Song
- Hematology Division, Department of Medicine, University of Utah and VAH, Salt Lake City, UT, 84132, USA
| | - Monika Horvathova
- Department of Biology, Faculty of Medicine and Dentistry, Palacky University, 775 15, Olomouc, Czech Republic
| | - Barbora Kralova
- Department of Biology, Faculty of Medicine and Dentistry, Palacky University, 775 15, Olomouc, Czech Republic
| | - Hana Votavova
- Institute of Hematology and Blood Transfusion, 12820, Prague, Czech Republic
| | - Josef T Prchal
- Hematology Division, Department of Medicine, University of Utah and VAH, Salt Lake City, UT, 84132, USA.
| | - Donghoon Yoon
- Hematology Division, Department of Medicine, University of Utah and VAH, Salt Lake City, UT, 84132, USA
- Myeloma Institute University of Arkansas for Medical Science, Little Rock, AR, USA
| |
Collapse
|
166
|
Dambach DM, Misner D, Brock M, Fullerton A, Proctor W, Maher J, Lee D, Ford K, Diaz D. Safety Lead Optimization and Candidate Identification: Integrating New Technologies into Decision-Making. Chem Res Toxicol 2015; 29:452-72. [DOI: 10.1021/acs.chemrestox.5b00396] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Donna M. Dambach
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Dinah Misner
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Mathew Brock
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Aaron Fullerton
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - William Proctor
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Jonathan Maher
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Dong Lee
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Kevin Ford
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Dolores Diaz
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| |
Collapse
|
167
|
Villafuerte FC. New genetic and physiological factors for excessive erythrocytosis and Chronic Mountain Sickness. J Appl Physiol (1985) 2015; 119:1481-6. [PMID: 26272318 PMCID: PMC4683346 DOI: 10.1152/japplphysiol.00271.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 08/07/2015] [Indexed: 01/03/2023] Open
Abstract
In the last few years, genetic and functional studies have provided important insight on the pathophysiology of excessive erythrocytosis (EE), the main sign of Chronic Mountain Sickness (CMS). The recent finding of the association of the CMS phenotype with a single-nucleotide polymorphism (SNP) in the Sentrin-specific Protease 1 (SENP1) gene, and its differential expression pattern in Andean highlanders with and without CMS, has triggered large interest in high-altitude studies because of the potential role of its gene product in the control of erythropoiesis. The SENP1 gene encodes for a protease that regulates the function of hypoxia-relevant transcription factors such as Hypoxia-Inducible Factor (HIF) and GATA, and thus might have an erythropoietic regulatory role in CMS through the modulation of the expression of erythropoietin (Epo) or Epo receptors. The different physiological patterns in the Epo-EpoR system found among Andeans, even among highlanders with CMS, together with their different degrees of erythropoietic response, might indicate specific underlying genetic backgrounds, which in turn might reflect different levels of adaptation to lifelong high-altitude hypoxia. This minireview discusses recent genetic findings potentially underlying EE and CMS, and their possible physiological mechanisms in Andean highlanders.
Collapse
Affiliation(s)
- Francisco C Villafuerte
- Laboratorio de Fisiología Comparada, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Perú
| |
Collapse
|
168
|
Zafiriou MP, Noack C, Unsöld B, Didie M, Pavlova E, Fischer HJ, Reichardt HM, Bergmann MW, El-Armouche A, Zimmermann WH, Zelarayan LC. Erythropoietin responsive cardiomyogenic cells contribute to heart repair post myocardial infarction. Stem Cells 2015; 32:2480-91. [PMID: 24806289 DOI: 10.1002/stem.1741] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 03/28/2014] [Accepted: 04/04/2014] [Indexed: 11/10/2022]
Abstract
The role of erythropoietin (Epo) in myocardial repair after infarction remains inconclusive. We observed high Epo receptor (EPOR) expression in cardiac progenitor cells (CPCs). Therefore, we aimed to characterize these cells and elucidate their contribution to myocardial regeneration on Epo stimulation. High EPOR expression was detected during murine embryonic heart development followed by a marked decrease until adulthood. EPOR-positive cells in the adult heart were identified in a CPC-enriched cell population and showed coexpression of stem, mesenchymal, endothelial, and cardiomyogenic cell markers. We focused on the population coexpressing early (TBX5, NKX2.5) and definitive (myosin heavy chain [MHC], cardiac Troponin T [cTNT]) cardiomyocyte markers. Epo increased their proliferation and thus were designated as Epo-responsive MHC expressing cells (EMCs). In vitro, EMCs proliferated and partially differentiated toward cardiomyocyte-like cells. Repetitive Epo administration in mice with myocardial infarction (cumulative dose 4 IU/g) resulted in an increase in cardiac EMCs and cTNT-positive cells in the infarcted area. This was further accompanied by a significant preservation of cardiac function when compared with control mice. Our study characterized an EPO-responsive MHC-expressing cell population in the adult heart. Repetitive, moderate-dose Epo treatment enhanced the proliferation of EMCs resulting in preservation of post-ischemic cardiac function.
Collapse
Affiliation(s)
- Maria Patapia Zafiriou
- Institute of Pharmacology, University Medical Center, Georg-August-Universität Göttingen, Göttingen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Ward D, Carter D, Homer M, Marucci L, Gampel A. Mathematical modeling reveals differential effects of erythropoietin on proliferation and lineage commitment of human hematopoietic progenitors in early erythroid culture. Haematologica 2015; 101:286-96. [PMID: 26589912 DOI: 10.3324/haematol.2015.133637] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/18/2015] [Indexed: 02/06/2023] Open
Abstract
Erythropoietin is essential for the production of mature erythroid cells, promoting both proliferation and survival. Whether erythropoietin and other cytokines can influence lineage commitment of hematopoietic stem and progenitor cells is of significant interest. To study lineage restriction of the common myeloid progenitor to the megakaryocyte/erythroid progenitor of peripheral blood CD34(+) cells, we have shown that the cell surface protein CD36 identifies the earliest lineage restricted megakaryocyte/erythroid progenitor. Using this marker and carboxyfluorescein succinimidyl ester to track cell divisions in vitro, we have developed a mathematical model that accurately predicts population dynamics of erythroid culture. Parameters derived from the modeling of cultures without added erythropoietin indicate that the rate of lineage restriction is not affected by erythropoietin. By contrast, megakaryocyte/erythroid progenitor proliferation is sensitive to erythropoietin from the time that CD36 first appears at the cell surface. These results shed new light on the role of erythropoietin in erythropoiesis and provide a powerful tool for further study of hematopoietic progenitor lineage restriction and erythropoiesis.
Collapse
Affiliation(s)
- Daniel Ward
- Department of Engineering Mathematics, Faculty of Engineering, University of Bristol
| | - Deborah Carter
- School of Biochemistry, Faculty of Medical and Veterinary Science, University of Bristol, UK
| | - Martin Homer
- Department of Engineering Mathematics, Faculty of Engineering, University of Bristol
| | - Lucia Marucci
- Department of Engineering Mathematics, Faculty of Engineering, University of Bristol
| | - Alexandra Gampel
- School of Biochemistry, Faculty of Medical and Veterinary Science, University of Bristol, UK
| |
Collapse
|
170
|
Suzuki N, Yamamoto M. Roles of renal erythropoietin-producing (REP) cells in the maintenance of systemic oxygen homeostasis. Pflugers Arch 2015; 468:3-12. [PMID: 26452589 DOI: 10.1007/s00424-015-1740-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 09/27/2015] [Accepted: 09/29/2015] [Indexed: 12/13/2022]
Abstract
Erythropoietic induction is critical for enhancing the efficiency of oxygen delivery during the chronic phase of the systemic hypoxia response. The erythroid growth factor erythropoietin (Epo) triggers the erythropoietic induction through the activation of erythroid genes related to cell survival, differentiation, and iron metabolism. Because Epo is produced in renal Epo-producing (REP) cells in a hypoxia-inducible manner, REP cells serve as a control center for the systemic hypoxia response. In fact, the loss of Epo production in REP cells causes chronic severe anemia in genetically modified mice, and REP cell-specific inactivation of PHD2 (prolyl-hydroxylase domain enzyme 2) results in erythrocytosis via overexpression of the Epo gene due to the constitutive activation of HIF2α (hypoxia-inducible transcription factor 2α). REP cells are located in the interstitial spaces between renal tubules and capillaries, where the oxygen supply is low but oxygen consumption is high, for the highly sensitive detection of decreased oxygen supplies to the body. Under disease conditions, REP cells transform to myofibroblasts and lose their Epo-producing ability. Therefore, elucidation of Epo gene regulation and REP cell features directly contributes to understanding the pathology of chronic kidney disease. To further analyze REP cells, we introduce a newly established mouse line in which REP cells are efficiently labeled with fluorescent protein.
Collapse
Affiliation(s)
- Norio Suzuki
- Division of Interdisciplinary Medical Science, Center for Oxygen Medicine, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, 980-8575, Japan.
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-ku, Sendai, 980-8575, Japan
| |
Collapse
|
171
|
Mohammdai-asl J, Ramezani A, Norozi F, Malehi AS, Asnafi AA, Far MAJ, Mousavi SH, Saki N. MicroRNAs in erythropoiesis and red blood cell disorders. FRONTIERS IN BIOLOGY 2015; 10:321-332. [DOI: 10.1007/s11515-015-1365-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
172
|
Cytokinesis failure in RhoA-deficient mouse erythroblasts involves actomyosin and midbody dysregulation and triggers p53 activation. Blood 2015; 126:1473-82. [PMID: 26228485 DOI: 10.1182/blood-2014-12-616169] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 07/20/2015] [Indexed: 01/06/2023] Open
Abstract
RhoA GTPase has been shown in vitro in cell lines and in vivo in nonmammalian organisms to regulate cell division, particularly during cytokinesis and abscission, when 2 daughter cells partition through coordinated actomyosin and microtubule machineries. To investigate the role of this GTPase in the rapidly proliferating mammalian erythroid lineage, we developed a mouse model with erythroid-specific deletion of RhoA. This model was proved embryonic lethal as a result of severe anemia by embryonic day 16.5 (E16.5). The primitive red blood cells were enlarged, poikilocytic, and frequently multinucleated, but were able to sustain life despite experiencing cytokinesis failure. In contrast, definitive erythropoiesis failed and the mice died by E16.5, with profound reduction of maturing erythroblast populations within the fetal liver. RhoA was required to activate myosin-regulatory light chain and localized at the site of the midbody formation in dividing wild-type erythroblasts. Cytokinesis failure caused by RhoA deficiency resulted in p53 activation and p21-transcriptional upregulation with associated cell-cycle arrest, increased DNA damage, and cell death. Our findings demonstrate the role of RhoA as a critical regulator for efficient erythroblast proliferation and the p53 pathway as a powerful quality control mechanism in erythropoiesis.
Collapse
|
173
|
Abstract
Determining the developmental pathway leading to erythrocytes and being able to isolate their progenitors are crucial to understanding and treating disorders of red cell imbalance such as anemia, myelodysplastic syndrome, and polycythemia vera. Here we show that the human erythrocyte progenitor (hEP) can be prospectively isolated from adult bone marrow. We found three subfractions that possessed different expression patterns of CD105 and CD71 within the previously defined human megakaryocyte/erythrocyte progenitor (hMEP; Lineage(-) CD34(+) CD38(+) IL-3Rα(-) CD45RA(-)) population. Both CD71(-) CD105(-) and CD71(+) CD105(-) MEPs, at least in vitro, still retained bipotency for the megakaryocyte (MegK) and erythrocyte (E) lineages, although the latter subpopulation is skewed in differentiation toward the erythroid lineage. Notably, the proliferative and differentiation output of the CD71(intermediate(int)/+) CD105(+) subset of cells within the MEP population was completely restricted to the erythroid lineage with the loss of MegK potential. CD71(+) CD105(-) MEPs are erythrocyte-biased MEPs (E-MEPs) and CD71(int/+) CD105(+) cells are EPs. These previously unclassified populations may facilitate further understanding of the molecular mechanisms governing human erythroid development and serve as potential therapeutic targets in disorders of the erythroid lineage.
Collapse
|
174
|
Gassmann M, Muckenthaler MU. Adaptation of iron requirement to hypoxic conditions at high altitude. J Appl Physiol (1985) 2015; 119:1432-40. [PMID: 26183475 DOI: 10.1152/japplphysiol.00248.2015] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/10/2015] [Indexed: 12/12/2022] Open
Abstract
Adequate acclimatization time to enable adjustment to hypoxic conditions is one of the most important aspects for mountaineers ascending to high altitude. Accordingly, most reviews emphasize mechanisms that cope with reduced oxygen supply. However, during sojourns to high altitude adjustment to elevated iron demand is equally critical. Thus in this review we focus on the interaction between oxygen and iron homeostasis. We review the role of iron 1) in the oxygen sensing process and erythropoietin (Epo) synthesis, 2) in gene expression control mediated by the hypoxia-inducible factor-2 (HIF-2), and 3) as an oxygen carrier in hemoglobin, myoglobin, and cytochromes. The blood hormone Epo that is abundantly expressed by the kidney under hypoxic conditions stimulates erythropoiesis in the bone marrow, a process requiring high iron levels. To ensure that sufficient iron is provided, Epo-controlled erythroferrone that is expressed in erythroid precursor cells acts in the liver to reduce expression of the iron hormone hepcidin. Consequently, suppression of hepcidin allows for elevated iron release from storage organs and enhanced absorption of dietary iron by enterocytes. As recently observed in sojourners at high altitude, however, iron uptake may be hampered by reduced appetite and gastrointestinal bleeding. Reduced iron availability, as observed in a hypoxic mountaineer, enhances hypoxia-induced pulmonary hypertension and may contribute to other hypoxia-related diseases. Overall, adequate systemic iron availability is an important prerequisite to adjust to high-altitude hypoxia and may have additional implications for disease-related hypoxic conditions.
Collapse
Affiliation(s)
- Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland, and Universidad Peruana Cayetano Heredia, Lima, Peru; and
| | - Martina U Muckenthaler
- Pediatric Oncology, Hematology and Immunology, University Hospital Heidelberg, Molecular Medicine Partnership Unit, University of Heidelberg, Translational Lung Research Center Heidelberg, and German Center for Lung Research, Heidelberg, Germany
| |
Collapse
|
175
|
Suzuki N. Erythropoietin gene expression: developmental-stage specificity, cell-type specificity, and hypoxia inducibility. TOHOKU J EXP MED 2015; 235:233-40. [PMID: 25786542 DOI: 10.1620/tjem.235.233] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Erythrocytes play an essential role in the delivery of oxygen from the lung to every organ; a decrease in erythrocytes (anemia) causes hypoxic stress and tissue damage. To maintain oxygen homeostasis in adult mammals, when the kidney senses hypoxia, it secretes an erythroid growth factor, erythropoietin (Epo), which stimulates erythropoiesis in the bone marrow. Recently, studies using genetically modified mice have shown that the in vivo expression profile of the Epo gene changes dramatically during development. The first Epo-producing cells emerge in the neural crest and neuroepithelium of mid-stage embryos and support primitive erythropoiesis in the yolk sac. Subsequently, Epo from the hepatocytes stimulates erythropoiesis in the fetal liver of later stage embryos in a paracrine manner. In fact, erythroid lineage cells comprise the largest cell population in the fetal liver, and hepatocytes are distributed among the erythroid cell clusters. Adult erythropoiesis in the bone marrow requires Epo that is secreted by renal Epo-producing cells (REP cells). REP cells are widely distributed in the renal cortex and outer medulla. Hypoxia-inducible Epo production both in hepatocytes and REP cells is controlled at the gene transcription level that is mainly mediated by the hypoxia-inducible transcription factor (HIF) pathway. These mouse studies further provide insights into the molecular mechanisms of the cell-type specific, hypoxia-inducible expression of the Epo gene, which involves multiple sets of cis- and trans-regulatory elements.
Collapse
Affiliation(s)
- Norio Suzuki
- Division of Interdisciplinary Medical Science, Center for Oxygen Medicine, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine
| |
Collapse
|
176
|
Abstract
Improved understanding of the oxygen-dependent regulation of erythropoiesis has provided new insights into the pathogenesis of anaemia associated with renal failure and has led to the development of novel therapeutic agents for its treatment. Hypoxia-inducible factor (HIF)-2 is a key regulator of erythropoiesis and iron metabolism. HIF-2 is activated by hypoxic conditions and controls the production of erythropoietin by renal peritubular interstitial fibroblast-like cells and hepatocytes. In anaemia associated with renal disease, erythropoiesis is suppressed due to inadequate erythropoietin production in the kidney, inflammation and iron deficiency; however, pharmacologic agents that activate the HIF axis could provide a physiologic approach to the treatment of renal anaemia by mimicking hypoxia responses that coordinate erythropoiesis with iron metabolism. This Review discusses the functional inter-relationships between erythropoietin, iron and inflammatory mediators under physiologic conditions and in relation to the pathogenesis of renal anaemia, as well as recent insights into the molecular and cellular basis of erythropoietin production in the kidney. It furthermore provides a detailed overview of current clinical experience with pharmacologic activators of HIF signalling as a novel comprehensive and physiologic approach to the treatment of anaemia.
Collapse
|
177
|
Souma T, Suzuki N, Yamamoto M. Renal erythropoietin-producing cells in health and disease. Front Physiol 2015; 6:167. [PMID: 26089800 PMCID: PMC4452800 DOI: 10.3389/fphys.2015.00167] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 05/19/2015] [Indexed: 01/01/2023] Open
Abstract
Erythropoietin (Epo) is an indispensable erythropoietic hormone primarily produced from renal Epo-producing cells (REPs). Epo production in REPs is tightly regulated in a hypoxia-inducible manner to maintain tissue oxygen homeostasis. Insufficient Epo production by REPs causes renal anemia and anemia associated with chronic disorders. Recent studies have broadened our understanding of REPs from prototypic hypoxia-responsive cells to dynamic fibrogenic cells. In chronic kidney disease, REPs are the major source of scar-forming myofibroblasts and actively produce fibrogenic molecules, including inflammatory cytokines. Notably, myofibroblast-transformed REPs (MF-REPs) recover their original physiological properties after resolution of the disease insults, suggesting that renal anemia and fibrosis could be reversible to some extent. Therefore, understanding the plasticity of REPs will lead to the development of novel targeted therapeutics for both renal fibrosis and anemia. This review summarizes the regulatory mechanisms how hypoxia-inducible Epo gene expression is attained in health and disease conditions.
Collapse
Affiliation(s)
- Tomokazu Souma
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine Sendai, Japan ; Division of Interdisciplinary Medical Science, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine Sendai, Japan ; Division of Nephrology and Hypertension, Feinberg School of Medicine, Northwestern University Chicago, IL, USA
| | - Norio Suzuki
- Division of Interdisciplinary Medical Science, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine Sendai, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine Sendai, Japan
| |
Collapse
|
178
|
|
179
|
Hypoxia Signaling Cascade for Erythropoietin Production in Hepatocytes. Mol Cell Biol 2015; 35:2658-72. [PMID: 26012551 DOI: 10.1128/mcb.00161-15] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/22/2015] [Indexed: 01/01/2023] Open
Abstract
Erythropoietin (Epo) is produced in the kidney and liver in a hypoxia-inducible manner via the activation of hypoxia-inducible transcription factors (HIFs) to maintain oxygen homeostasis. Accelerating Epo production in hepatocytes is one plausible therapeutic strategy for treating anemia caused by kidney diseases. To elucidate the regulatory mechanisms of hepatic Epo production, we analyzed mouse lines harboring liver-specific deletions of genes encoding HIF-prolyl-hydroxylase isoforms (PHD1, PHD2, and PHD3) that mediate the inactivation of HIF1α and HIF2α under normal oxygen conditions. The loss of all PHD isoforms results in both polycythemia, which is caused by Epo overproduction, and fatty livers. We found that deleting any combination of two PHD isoforms induces polycythemia without steatosis complications, whereas the deletion of a single isoform induces no apparent phenotype. Polycythemia is prevented by the loss of either HIF2α or the hepatocyte-specific Epo gene enhancer (EpoHE). Chromatin analyses show that the histones around EpoHE dissociate from the nucleosome structure after HIF2α activation. HIF2α also induces the expression of HIF3α, which is involved in the attenuation of Epo production. These results demonstrate that the total amount of PHD activity is more important than the specific function of each isoform for hepatic Epo expression regulated by a PHD-HIF2α-EpoHE cascade in vivo.
Collapse
|
180
|
Matsuyama T, Tanaka T, Tatsumi K, Daijo H, Kai S, Harada H, Fukuda K. Midazolam inhibits the hypoxia-induced up-regulation of erythropoietin in the central nervous system. Eur J Pharmacol 2015; 761:189-98. [PMID: 26001375 DOI: 10.1016/j.ejphar.2015.05.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 05/08/2015] [Accepted: 05/18/2015] [Indexed: 12/29/2022]
Abstract
Erythropoietin (EPO), a regulator of red blood cell production, is endogenously expressed in the central nervous system. It is mainly produced by astrocytes under hypoxic conditions and has proven to have neuroprotective and neurotrophic effects. In the present study, we investigated the effect of midazolam on EPO expression in primary cultured astrocytes and the mouse brain. Midazolam was administered to 6-week-old BALB/c male mice under hypoxic conditions and pregnant C57BL/6N mice under normoxic conditions. Primary cultured astrocytes were also treated with midazolam under hypoxic conditions. The expression of EPO mRNA in mice brains and cultured astrocytes was studied. In addition, the expression of hypoxia-inducible factor (HIF), known as the main regulator of EPO, was evaluated. Midazolam significantly reduced the hypoxia-induced up-regulation of EPO in BALB/c mice brains and primary cultured astrocytes and suppressed EPO expression in the fetal brain. Midazolam did not affect the total amount of HIF proteins but significantly inhibited the nuclear expression of HIF-1α and HIF-2α proteins. These results demonstrated the suppressive effects of midazolam on the hypoxia-induced up-regulation of EPO both in vivo and in vitro.
Collapse
Affiliation(s)
- Tomonori Matsuyama
- Department of Anesthesia, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tomoharu Tanaka
- Department of Anesthesia, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Kenichiro Tatsumi
- Department of Anesthesia, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hiroki Daijo
- Department of Anesthesia, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shinichi Kai
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, MA 02114, USA
| | - Hiroshi Harada
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Kazuhiko Fukuda
- Department of Anesthesia, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
181
|
Tsuboi I, Yamashita T, Nagano M, Kimura K, To'a Salazar G, Ohneda O. Impaired expression of HIF-2α induces compensatory expression of HIF-1α for the recovery from anemia. J Cell Physiol 2015; 230:1534-48. [DOI: 10.1002/jcp.24899] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/12/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Ikki Tsuboi
- Graduate School of Comprehensive Human Sciences; Laboratory of Regenerative Medicine and Stem Cell Biology; University of Tsukuba; Tsukuba Japan
| | - Toshiharu Yamashita
- Graduate School of Comprehensive Human Sciences; Laboratory of Regenerative Medicine and Stem Cell Biology; University of Tsukuba; Tsukuba Japan
| | - Masumi Nagano
- Graduate School of Comprehensive Human Sciences; Laboratory of Regenerative Medicine and Stem Cell Biology; University of Tsukuba; Tsukuba Japan
| | - Kenichi Kimura
- Graduate School of Comprehensive Human Sciences; Laboratory of Regenerative Medicine and Stem Cell Biology; University of Tsukuba; Tsukuba Japan
| | - Georgina To'a Salazar
- Graduate School of Comprehensive Human Sciences; Laboratory of Regenerative Medicine and Stem Cell Biology; University of Tsukuba; Tsukuba Japan
| | - Osamu Ohneda
- Graduate School of Comprehensive Human Sciences; Laboratory of Regenerative Medicine and Stem Cell Biology; University of Tsukuba; Tsukuba Japan
| |
Collapse
|
182
|
Tapia O, Fong LG, Huber MD, Young SG, Gerace L. Nuclear envelope protein Lem2 is required for mouse development and regulates MAP and AKT kinases. PLoS One 2015; 10:e0116196. [PMID: 25790465 PMCID: PMC4366207 DOI: 10.1371/journal.pone.0116196] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 11/21/2014] [Indexed: 12/31/2022] Open
Abstract
The nuclear lamina, along with associated nuclear membrane proteins, is a nexus for regulating signaling in the nucleus. Numerous human diseases arise from mutations in lamina proteins, and experimental models for these disorders have revealed aberrant regulation of various signaling pathways. Previously, we reported that the inner nuclear membrane protein Lem2, which is expressed at high levels in muscle, promotes the differentiation of cultured myoblasts by attenuating ERK signaling. Here, we have analyzed mice harboring a disrupted allele for the Lem2 gene (Lemd2). No gross phenotypic defects were seen in heterozygotes, although muscle regeneration induced by cardiotoxin was delayed. By contrast, homozygous Lemd2 knockout mice died by E11.5. Although many normal morphogenetic hallmarks were observed in E10.5 knockout embryos, most tissues were substantially reduced in size. This was accompanied by activation of multiple MAP kinases (ERK1/2, JNK, p38) and AKT. Knockdown of Lem2 expression in C2C12 myoblasts also led to activation of MAP kinases and AKT. These findings indicate that Lemd2 plays an essential role in mouse embryonic development and that it is involved in regulating several signaling pathways. Since increased MAP kinase and AKT/mTORC signaling is found in other animal models for diseases linked to nuclear lamina proteins, LEMD2 should be considered to be another candidate gene for human disease.
Collapse
Affiliation(s)
- Olga Tapia
- Department of Cell and Molecular Biology, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California 92037, United States of America
| | - Loren G. Fong
- Department of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States of America
| | - Michael D. Huber
- Department of Cell and Molecular Biology, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California 92037, United States of America
| | - Stephen G. Young
- Department of Medicine, University of California Los Angeles, Los Angeles, California 90095, United States of America
- Department of Human Genetics, University of California Los Angeles, Los Angeles, California 90095, United States of America
| | - Larry Gerace
- Department of Cell and Molecular Biology, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California 92037, United States of America
- * E-mail:
| |
Collapse
|
183
|
Suzuki N, Mukai HY, Yamamoto M. In vivo regulation of erythropoiesis by chemically inducible dimerization of the erythropoietin receptor intracellular domain. PLoS One 2015; 10:e0119442. [PMID: 25790231 PMCID: PMC4366189 DOI: 10.1371/journal.pone.0119442] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 01/13/2015] [Indexed: 11/18/2022] Open
Abstract
Erythropoietin (Epo) and its receptor (EpoR) are required for the regulation of erythropoiesis. Epo binds to the EpoR homodimer on the surface of erythroid progenitors and erythroblasts, and positions the intracellular domains of the homodimer to be in close proximity with each other. This conformational change is sufficient for the initiation of Epo-EpoR signal transduction. Here, we established a system of chemically regulated erythropoiesis in transgenic mice expressing a modified EpoR intracellular domain (amino acids 247-406) in which dimerization is induced using a specific compound (chemical inducer of dimerization, CID). Erythropoiesis is reversibly induced by oral administration of the CID to the transgenic mice. Because transgene expression is limited to hematopoietic cells by the Gata1 gene regulatory region, the effect of the CID is limited to erythropoiesis without adverse effects. Additionally, we show that the 160 amino acid sequence is the minimal essential domain of EpoR for intracellular signaling of chemically inducible erythropoiesis in vivo. We propose that the CID-dependent dimerization system combined with the EpoR intracellular domain and the Gata1 gene regulatory region generates a novel peroral strategy for the treatment of anemia.
Collapse
Affiliation(s)
- Norio Suzuki
- Division of Interdisciplinary Medical Science, Center for Oxygen Medicine, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- * E-mail:
| | - Harumi Y. Mukai
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
184
|
Hänggi P, Telezhkin V, Kemp PJ, Schmugge M, Gassmann M, Goede JS, Speer O, Bogdanova A. Functional plasticity of the N-methyl-d-aspartate receptor in differentiating human erythroid precursor cells. Am J Physiol Cell Physiol 2015; 308:C993-C1007. [PMID: 25788577 PMCID: PMC4469746 DOI: 10.1152/ajpcell.00395.2014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/17/2015] [Indexed: 11/22/2022]
Abstract
Calcium signaling is essential to support erythroid proliferation and differentiation. Precise control of the intracellular Ca2+ levels in erythroid precursor cells (EPCs) is afforded by coordinated expression and function of several cation channels, including the recently identified N-methyl-d-aspartate receptor (NMDAR). Here, we characterized the changes in Ca2+ uptake and electric currents mediated by the NMDARs occurring during EPC differentiation using flow cytometry and patch clamp. During erythropoietic maturation, subunit composition and properties of the receptor changed; in proerythroblasts and basophilic erythroblasts, fast deactivating currents with high amplitudes were mediated by the GluN2A subunit-dominated receptors, while at the polychromatic and orthochromatic erythroblast stages, the GluN2C subunit was getting more abundant, overriding the expression of GluN2A. At these stages, the currents mediated by the NMDARs carried the features characteristic of the GluN2C-containing receptors, such as prolonged decay time and lower conductance. Kinetics of this switch in NMDAR properties and abundance varied markedly from donor to donor. Despite this variability, NMDARs were essential for survival of EPCs in any subject tested. Our findings indicate that NMDARs have a dual role during erythropoiesis, supporting survival of polychromatic erythroblasts and contributing to the Ca2+ homeostasis from the orthochromatic erythroblast stage to circulating red blood cells.
Collapse
Affiliation(s)
- Pascal Hänggi
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Division of Hematology University Hospital Zurich, Zurich, Switzerland; University Children's Hospital, Zurich, Switzerland
| | - Vsevolod Telezhkin
- Division of Pathophysiology and Repair, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Paul J Kemp
- Division of Pathophysiology and Repair, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Markus Schmugge
- University Children's Hospital, Zurich, Switzerland; Children's Research Center, Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Max Gassmann
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Jeroen S Goede
- Division of Hematology University Hospital Zurich, Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Oliver Speer
- University Children's Hospital, Zurich, Switzerland; Children's Research Center, Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Anna Bogdanova
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| |
Collapse
|
185
|
Erythropoietin negatively regulates pituitary ACTH secretion. Brain Res 2015; 1608:14-20. [PMID: 25765155 DOI: 10.1016/j.brainres.2015.02.052] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/24/2015] [Accepted: 02/27/2015] [Indexed: 11/21/2022]
Abstract
Erythropoietin (Epo) and Epo-receptor (EpoR) signaling, in addition to its classical role in erythropoiesis, exhibit a protective response in non-hematopoietic tissues. Mice with EpoR expression restricted to only hematopoietic tissues (ΔEpoRE), become obese, have low energy expenditure, and are glucose intolerant and insulin resistant. In the arcuate nucleus of the mouse hypothalamus, EpoR expression co-localizes in proopiomelanocortin (POMC) neurons. In vivo high-dose Epo treatment increases hypothalamus POMC, reduces food intake and fat mass accumulation. Here we report that Epo treatment also decreases plasma concentration of the pituitary derived POMC peptide, adrenocorticotropic hormone (ACTH). Conversely, ΔEpoRE mice show reduced hypothalamus POMC and high plasma concentrations of ACTH. In the pituitary, POMC is synthesized in the corticotroph cells, and here we examine Epo effect on pituitary POMC expression using the AtT-20 mouse corticotroph pituitary cell line. In AtT-20 cells, enzyme immunoassay analysis showed that Epo inhibits ACTH secretion. This effect is post-translational, as Epo treatment did not affect POMC mRNA expression but increased intracellular levels of ACTH peptide. Moreover, Epo reduced the basal intracellular calcium (Ca(2+)) levels, suggesting an effect in the Ca(2+)-signaling pathway. In summary, our studies suggest a novel regulatory pathway of ACTH secretion in the pituitary via EpoR-signaling. The higher plasma ACTH level in ΔEpoRE mice also suggests a possible mechanism of deregulated pituitary function with loss of Epo-signaling.
Collapse
|
186
|
Wiedenmann T, Ehrhardt S, Cerny D, Hildebrand D, Klein S, Heeg K, Kubatzky KF. Erythropoietin acts as an anti-inflammatory signal on murine mast cells. Mol Immunol 2015; 65:68-76. [PMID: 25645506 DOI: 10.1016/j.molimm.2015.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/23/2014] [Accepted: 01/12/2015] [Indexed: 02/01/2023]
Abstract
Recently it was found that the erythropoietin receptor (EpoR) is expressed on innate immune cells, such as dendritic cells and macrophages. We found that murine bone marrow-derived mast cells express the EpoR and that its expression is increased under hypoxic conditions. Interestingly, Epo stimulation of the cells did not activate signal transducer and activator of transcription molecules, nor did we find differences in the expression of typical STAT-dependent genes, the proliferation rate, and the ability to differentiate or to protect the cells from apoptosis. Instead, we demonstrate that stimulation of mast cells with Epo leads to phosphorylation of the receptor tyrosine kinase c-kit. We hypothesize that this is due to the formation of a receptor complex between the EpoR and c-kit. The common beta chain of the IL-3 receptor family, which was described as part of the tissue protective receptor (TPR) on other non-erythroid cells, however is not activated. To investigate whether the EpoR/c-kit complex has tissue protective properties, cells were treated with the Toll-like receptor ligand LPS. Combined Epo and LPS treatment downregulated the inflammatory response of the cells as detected by a decrease in IL-6 and TNF-α secretion.
Collapse
Affiliation(s)
- Tanja Wiedenmann
- Universitätsklinikum Heidelberg, Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Germany.
| | - Stefanie Ehrhardt
- Universitätsklinikum Heidelberg, Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Germany.
| | - Daniela Cerny
- Universitätsklinikum Heidelberg, Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Germany.
| | - Dagmar Hildebrand
- Universitätsklinikum Heidelberg, Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Germany.
| | - Sabrina Klein
- Universitätsklinikum Heidelberg, Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Germany.
| | - Klaus Heeg
- Universitätsklinikum Heidelberg, Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Germany.
| | - Katharina F Kubatzky
- Universitätsklinikum Heidelberg, Zentrum für Infektiologie, Medizinische Mikrobiologie und Hygiene, Germany.
| |
Collapse
|
187
|
Pharmacodynamics, pharmacokinetics, and tolerability of intravenous or subcutaneous GC1113, a novel erythropoiesis-stimulating agent. Clin Drug Investig 2015; 34:373-82. [PMID: 24623104 DOI: 10.1007/s40261-014-0183-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND AND OBJECTIVES GC1113, a hybrid Fc-fused erythropoietin, is a novel erythropoiesis-stimulating agent that is expected to have an extended duration of action. The preclinical data showed that the hemoglobin increase lasted longer following GC1113 administration than it did following the administration of darbepoetin alfa (NESP®). This study aimed to investigate the pharmacodynamic and pharmacokinetic characteristics and tolerability profiles of GC1113 in humans after single intravenous or subcutaneous administration and to compare the results with those for darbepoetin alfa. METHODS A dose-block randomized, placebo- and active-controlled, dose-escalation phase I clinical trial was conducted in 96 healthy volunteers. Blood samples were collected before and up to 672 h after drug administration and the serum erythropoietin concentration following the GC1113 or darbepoetin alfa administration was measured by an ELISA. The reticulocyte counts were measured for pharmacodynamic assessments. Pharmacokinetic and pharmacodynamic parameters were determined using non-compartmental methods. RESULTS The reticulocyte count-time profiles in the intravenous GC1113 3-5 μg/kg groups were comparable with those of the darbepoetin alfa 30 μg group. After subcutaneous administration of GC1113, reticulocyte count peaked later and decreased more slowly than it did following darbepoetin alfa administration. GC1113 (0.3-5 μg/kg intravenous, 1-8 μg/kg subcutaneous) was well-tolerated in the volunteers, and no immunogenicity was observed. CONCLUSION GC1113 was tolerated and effective in the studied dose range; these findings could be applied to further clinical studies with patients.
Collapse
|
188
|
Abstract
The onset of hematopoiesis in mammals is defined by generation of primitive erythrocytes and macrophage progenitors in embryonic yolk sac. Laboratories have met the challenge of transient and swiftly changing specification events from ventral mesoderm through multipotent progenitors and maturing lineage-restricted hematopoietic subtypes, by developing powerful in vitro experimental models to interrogate hematopoietic ontogeny. Most importantly, studies of differentiating embryonic stem cell derivatives in embryoid body and stromal coculture systems have identified crucial roles for transcription factor networks (e.g. Gata1, Runx1, Scl) and signaling pathways (e.g. BMP, VEGF, WNT) in controlling stem and progenitor cell output. These and other relevant pathways have pleiotropic biological effects, and are often associated with early embryonic lethality in knockout mice. Further refinement in subsequent studies has allowed conditional expression of key regulatory genes, and isolation of progenitors via cell surface markers (e.g. FLK1) and reporter-tagged constructs, with the purpose of measuring their primitive and definitive hematopoietic potential. These observations continue to inform attempts to direct the differentiation, and augment the expansion, of progenitors in human cell culture systems that may prove useful in cell replacement therapies for hematopoietic deficiencies. The purpose of this review is to survey the extant literature on the use of differentiating murine embryonic stem cells in culture to model the developmental process of yolk sac hematopoiesis.
Collapse
|
189
|
Shen H, Cavallero S, Estrada KD, Sandovici I, Kumar SR, Makita T, Lien CL, Constancia M, Sucov HM. Extracardiac control of embryonic cardiomyocyte proliferation and ventricular wall expansion. Cardiovasc Res 2015; 105:271-8. [PMID: 25560321 DOI: 10.1093/cvr/cvu269] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AIMS The strategies that control formation of the ventricular wall during heart development are not well understood. In previous studies, we documented IGF2 as a major mitogenic signal that controls ventricular cardiomyocyte proliferation and chamber wall expansion. Our objective in this study was to define the tissue source of IGF2 in heart development and the upstream pathways that control its expression. METHODS AND RESULTS Using a number of mouse genetic tools, we confirm that the critical source of IGF2 is the epicardium. We find that epicardial Igf2 expression is controlled in a biphasic manner, first induced by erythropoietin and then regulated by oxygen and glucose with onset of placental function. Both processes are independently controlled by retinoic acid signalling. CONCLUSIONS Our results demonstrate that ventricular wall cardiomyocyte proliferation is subdivided into distinct regulatory phases. Each involves instructive cues that originate outside the heart and thereby act on the epicardium in an endocrine manner, a mode of regulation that is mostly unknown in embryogenesis.
Collapse
Affiliation(s)
- Hua Shen
- Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, 1425 San Pablo Street, BCC-511, Los Angeles, CA 90033, USA
| | - Susana Cavallero
- Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, 1425 San Pablo Street, BCC-511, Los Angeles, CA 90033, USA
| | - Kristine D Estrada
- Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, 1425 San Pablo Street, BCC-511, Los Angeles, CA 90033, USA
| | - Ionel Sandovici
- MRC Metabolic Diseases Unit, Department of Obstetrics and Gynaecology and NIHR Cambridge Biomedical Research Centre, University of Cambridge Metabolic Research Laboratories, Cambridge, UK Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - S Ram Kumar
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Takako Makita
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Ching-Ling Lien
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Miguel Constancia
- MRC Metabolic Diseases Unit, Department of Obstetrics and Gynaecology and NIHR Cambridge Biomedical Research Centre, University of Cambridge Metabolic Research Laboratories, Cambridge, UK Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Henry M Sucov
- Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, 1425 San Pablo Street, BCC-511, Los Angeles, CA 90033, USA
| |
Collapse
|
190
|
Zhang L, Wang H, Wang T, Jiang N, Yu P, Chong Y, Fu F. Ferulic acid ameliorates nerve injury induced by cerebral ischemia in rats. Exp Ther Med 2014; 9:972-976. [PMID: 25667662 PMCID: PMC4316951 DOI: 10.3892/etm.2014.2157] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 10/23/2014] [Indexed: 01/04/2023] Open
Abstract
This study was designed to investigate the protective effect of ferulic acid (FA) on nerve injury induced by cerebral ischemia. Focal cerebral ischemia was induced by occlusion of the right middle cerebral artery and reperfusion 90 min later in male Sprague-Dawley rats. Daily treatment of the rats with FA was initiated 30 min after the surgery, and was continued for 7 days. The efficacy of FA against nerve injury was assessed by neurological deficit scores as well as pathohistological observation. The expression levels in the brain and level in the peripheral blood of erythropoietin (EPO) and granulocyte colony-stimulating factor (G-CSF) were analyzed by immunohistochemistry and enzyme-linked immunosorbent assay (ELISA), respectively. The results showed that FA attenuated nerve injury of the hippocampus, significantly ameliorated neurological deficits, and increased EPO but not G-CSF expression in the hippocampus and the peripheral blood of ischemic rats. The findings indicate that FA has certain protective effects on the nerve injury of cerebral ischemia, and suggest that promoting EPO in the brain and peripheral blood may be one of the neuroprotective mechanisms of FA.
Collapse
Affiliation(s)
- Leiming Zhang
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Hongsheng Wang
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Tian Wang
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Na Jiang
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Pengfei Yu
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Yating Chong
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Fenghua Fu
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| |
Collapse
|
191
|
Abstract
Transferrin receptor 2 (TFR2) contributes to hepcidin regulation in the liver and associates with erythropoietin receptor in erythroid cells. Nevertheless, TFR2 mutations cause iron overload (hemochromatosis type 3) without overt erythroid abnormalities. To clarify TFR2 erythroid function, we generated a mouse lacking Tfr2 exclusively in the bone marrow (Tfr2(BMKO)). Tfr2(BMKO) mice have normal iron parameters, reduced hepcidin levels, higher hemoglobin and red blood cell counts, and lower mean corpuscular volume than normal control mice, a phenotype that becomes more evident in iron deficiency. In Tfr2(BMKO) mice, the proportion of nucleated erythroid cells in the bone marrow is higher and the apoptosis lower than in controls, irrespective of comparable erythropoietin levels. Induction of moderate iron deficiency increases erythroblasts number, reduces apoptosis, and enhances erythropoietin (Epo) levels in controls, but not in Tfr2(BMKO) mice. Epo-target genes such as Bcl-xL and Epor are highly expressed in the spleen and in isolated erythroblasts from Tfr2(BMKO) mice. Low hepcidin expression in Tfr2(BMKO) is accounted for by erythroid expansion and production of the erythroid regulator erythroferrone. We suggest that Tfr2 is a component of a novel iron-sensing mechanism that adjusts erythrocyte production according to iron availability, likely by modulating the erythroblast Epo sensitivity.
Collapse
|
192
|
Misener R, Fuentes Garí M, Rende M, Velliou E, Panoskaltsis N, Pistikopoulos EN, Mantalaris A. Global superstructure optimisation of red blood cell production in a parallelised hollow fibre bioreactor. Comput Chem Eng 2014. [DOI: 10.1016/j.compchemeng.2014.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
193
|
Schnöder TM, Arreba-Tutusaus P, Griehl I, Bullinger L, Buschbeck M, Lane SW, Döhner K, Plass C, Lipka DB, Heidel FH, Fischer T. Epo-induced erythroid maturation is dependent on Plcγ1 signaling. Cell Death Differ 2014; 22:974-85. [PMID: 25394487 DOI: 10.1038/cdd.2014.186] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 09/18/2014] [Accepted: 10/06/2014] [Indexed: 12/22/2022] Open
Abstract
Erythropoiesis is a tightly regulated process. Development of red blood cells occurs through differentiation of hematopoietic stem cells (HSCs) into more committed progenitors and finally into erythrocytes. Binding of erythropoietin (Epo) to its receptor (EpoR) is required for erythropoiesis as it promotes survival and late maturation of erythroid progenitors. In vivo and in vitro studies have highlighted the requirement of EpoR signaling through Janus kinase 2 (Jak2) tyrosine kinase and Stat5a/b as a central pathway. Here, we demonstrate that phospholipase C gamma 1 (Plcγ1) is activated downstream of EpoR-Jak2 independently of Stat5. Plcγ1-deficient pro-erythroblasts and erythroid progenitors exhibited strong impairment in differentiation and colony-forming potential. In vivo, suppression of Plcγ1 in immunophenotypically defined HSCs (Lin(-)Sca1(+)KIT(+)CD48(-)CD150(+)) severely reduced erythroid development. To identify Plcγ1 effector molecules involved in regulation of erythroid differentiation, we assessed changes occurring at the global transcriptional and DNA methylation level after inactivation of Plcγ1. The top common downstream effector was H2afy2, which encodes for the histone variant macroH2A2 (mH2A2). Inactivation of mH2A2 expression recapitulated the effects of Plcγ1 depletion on erythroid maturation. Taken together, our findings identify Plcγ1 and its downstream target mH2A2, as a 'non-canonical' Epo signaling pathway essential for erythroid differentiation.
Collapse
Affiliation(s)
- T M Schnöder
- Department of Hematology and Oncology, Center for Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - P Arreba-Tutusaus
- Department of Hematology and Oncology, Center for Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - I Griehl
- Department of Hematology and Oncology, Center for Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - L Bullinger
- Internal Medicine III, Department of Hematology/Oncology, University Hospital Ulm, Ulm, Germany
| | - M Buschbeck
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Campus Can Ruti, Badalona, Spain
| | - S W Lane
- Division of Immunology, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Australia
| | - K Döhner
- Internal Medicine III, Department of Hematology/Oncology, University Hospital Ulm, Ulm, Germany
| | - C Plass
- Division of Epigenomics and Cancer Risk Factors (C010), German Cancer Research Center, Heidelberg, Germany
| | - D B Lipka
- 1] Department of Hematology and Oncology, Center for Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany [2] Division of Epigenomics and Cancer Risk Factors (C010), German Cancer Research Center, Heidelberg, Germany
| | - F H Heidel
- Department of Hematology and Oncology, Center for Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - T Fischer
- Department of Hematology and Oncology, Center for Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| |
Collapse
|
194
|
Kit transduced signals counteract erythroid maturation by MAPK-dependent modulation of erythropoietin signaling and apoptosis induction in mouse fetal liver. Cell Death Differ 2014; 22:790-800. [PMID: 25323585 DOI: 10.1038/cdd.2014.172] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 09/08/2014] [Accepted: 09/16/2014] [Indexed: 12/14/2022] Open
Abstract
Signaling by the stem cell factor receptor Kit in hematopoietic stem and progenitor cells is functionally associated with the regulation of cellular proliferation, differentiation and survival. Expression of the receptor is downregulated upon terminal differentiation in most lineages, including red blood cell terminal maturation, suggesting that omission of Kit transduced signals is a prerequisite for the differentiation process to occur. However, the molecular mechanisms by which Kit signaling preserves the undifferentiated state of progenitor cells are not yet characterized in detail. In this study, we generated a mouse model for inducible expression of a Kit receptor carrying an activating mutation and studied its effects on fetal liver hematopoiesis. We found that sustained Kit signaling leads to expansion of erythroid precursors and interferes with terminal maturation beyond the erythroblast stage. Primary KIT(D816V) erythroblasts stimulated to differentiate fail to exit cell cycle and show elevated rates of apoptosis because of insufficient induction of survival factors. They further retain expression of progenitor cell associated factors c-Myc, c-Myb and GATA-2 and inefficiently upregulate erythroid transcription factors GATA-1, Klf1 and Tal1. In KIT(D816V) erythroblasts we found constitutive activation of the mitogen-activated protein kinase (MAPK) pathway, elevated expression of the src kinase family member Lyn and impaired Akt activation in response to erythropoietin. We demonstrate that the block in differentiation is partially rescued by MAPK inhibition, and completely rescued by the multikinase inhibitor Dasatinib. These results show that a crosstalk between Kit and erythropoietin receptor signaling cascades exists and that continuous Kit signaling, partly mediated by the MAPK pathway, interferes with this crosstalk.
Collapse
|
195
|
Miyazaki Y, Taguchi K, Sou K, Watanabe H, Ishima Y, Miyakawa T, Mitsuya H, Fukagawa M, Otagiri M, Maruyama T. Therapeutic Impact of Erythropoietin-Encapsulated Liposomes Targeted to Bone Marrow on Renal Anemia. Mol Pharm 2014; 11:4238-48. [DOI: 10.1021/mp500453a] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yuri Miyazaki
- Department
of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Kazuaki Taguchi
- Faculty
of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
| | - Keitaro Sou
- Center
for Advanced Biomedical Sciences/TWIns, Waseda University, Tokyo 162-8480, Japan
| | - Hiroshi Watanabe
- Department
of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
- Center
for Clinical Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Yu Ishima
- Department
of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Toshikazu Miyakawa
- Department
of Hematology and Infectious Diseases, Kumamoto University Hospital, Kumamoto 860-8556, Japan
| | - Hiroaki Mitsuya
- Department
of Hematology and Infectious Diseases, Kumamoto University Hospital, Kumamoto 860-8556, Japan
| | - Masafumi Fukagawa
- Division
of Nephrology, Endocrinology, and Metabolism, Tokai University School of Medicine, Isehara 259−1193, Japan
| | - Masaki Otagiri
- Department
of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
- Faculty
of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082, Japan
- DDS
Research Institute, Sojo University, Kumamoto 860-0082, Japan
| | - Toru Maruyama
- Department
of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
- Center
for Clinical Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| |
Collapse
|
196
|
Abstract
Erythropoiesis is a vital process governed through various factors. There is extreme unavailability of suitable donor due to rare phenotypic blood groups and other related complications like hemoglobinopathies, polytransfusion patients, and polyimmunization. Looking at the worldwide scarcity of blood, especially in low income countries and the battlefield, mimicking erythropoiesis using ex vivo methods can provide an efficient answer to various problems associated with present donor derived blood supply system. Fortunately, there are many ex vivo erythropoiesis methodologies being developed by various research groups using stem cells as the major source material for large scale blood production. Most of these ex vivo protocols use a cocktail of similar growth factors under overlapping growth conditions. Erythropoietin (EPO) is a key regulator in most ex vivo protocols along with other growth factors such as SCF, IL-3, IGF-1, and Flt-3. Now transfusable units of blood can be produced by using these protocols with their set of own limitations. The present paper focuses on the molecular mechanism and significance of various growth factors in these protocols that shall remain helpful for large scale production.
Collapse
|
197
|
Knight ZA, Schmidt SF, Birsoy K, Tan K, Friedman JM. A critical role for mTORC1 in erythropoiesis and anemia. eLife 2014; 3:e01913. [PMID: 25201874 PMCID: PMC4179304 DOI: 10.7554/elife.01913] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 09/04/2014] [Indexed: 12/31/2022] Open
Abstract
Red blood cells (RBC) must coordinate their rate of growth and proliferation with the availability of nutrients, such as iron, but the signaling mechanisms that link the nutritional state to RBC growth are incompletely understood. We performed a screen for cell types that have high levels of signaling through mTORC1, a protein kinase that couples nutrient availability to cell growth. This screen revealed that reticulocytes show high levels of phosphorylated ribosomal protein S6, a downstream target of mTORC1. We found that mTORC1 activity in RBCs is regulated by dietary iron and that genetic activation or inhibition of mTORC1 results in macrocytic or microcytic anemia, respectively. Finally, ATP competitive mTOR inhibitors reduced RBC proliferation and were lethal after treatment with phenylhydrazine, an inducer of hemolysis. These results identify the mTORC1 pathway as a critical regulator of RBC growth and proliferation and establish that perturbations in this pathway result in anemia. DOI:http://dx.doi.org/10.7554/eLife.01913.001 To multiply and grow, cells need to create more of the molecules—such as proteins—that make up their structure. This only happens if the cell has a good supply of the nutrients used to build the proteins. Red blood cells are particularly sensitive to the supply of nutrients, especially iron, which is a key component of the hemoglobin molecules that enable the cells to transport oxygen around the body. A lack of iron can lead to a shortage of red blood cells and a condition called anemia. People with mild forms of anemia may feel tired or weak, but more severe forms of anemia can cause heart problems and even death. A protein called mTOR forms part of a protein complex that helps alert the cells of many different organisms to the presence of nutrients. mTOR can add phosphate groups to ribosomes—the molecular machines that translate molecules of mRNA to build proteins. In 2012, researchers developed a technique called Phospho-Trap that can isolate these phosphorylated ribosomes from cells. Cells with an activated mTOR complex express more mTOR protein and in turn have more ribosomes that are modified. Examining the mRNA molecules associated with these ribosomes can reveal which proteins are produced in greater amounts in these cells. Previous experiments using Phospho-Trap found the proteins that make up hemoglobin in unexpectedly high amounts in the mouse brain. Now, Knight et al.—and other researchers involved in the 2012 work—have established that the hemoglobin was not coming from the brain cells but from immature red blood cells circulating within the brain. These immature blood cells were found to have a highly active mTOR complex that promotes the production of hemoglobin and new blood cells. Using genetic techniques in mice, Knight et al. found that the mTOR complex can cause anemia if it is underactive or overactive. Underactive mTOR complexes cause a type of anemia that produces small red blood cells and is usually triggered by a lack of iron. This made sense because mTOR is known to regulate both protein production and cell size. Boosting the activity of the mTOR complex leads to a type of anemia in which the cells are much larger than normal, and which is normally associated with inadequate amounts of folate and B12 vitamins. When Knight et al. gave mice a drug that inhibits the mTOR protein, the mice developed anemia that resolved when the treatment stopped. However, mice that were given the mTOR inhibitor at the same time as a drug that destroys red blood cells, all died within days. Clinical trials are currently testing mTOR inhibitors as a possible cancer treatment; however, a common side effect of chemotherapy is that it stops new red blood cells being produced. Knight et al. suggest that the red blood cells of patients in these clinical trials must be closely monitored before deciding whether to continue the treatment further. DOI:http://dx.doi.org/10.7554/eLife.01913.002
Collapse
Affiliation(s)
- Zachary A Knight
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Sarah F Schmidt
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Kivanc Birsoy
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Keith Tan
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| |
Collapse
|
198
|
Wang L, Di L, Noguchi CT. Erythropoietin, a novel versatile player regulating energy metabolism beyond the erythroid system. Int J Biol Sci 2014; 10:921-39. [PMID: 25170305 PMCID: PMC4147225 DOI: 10.7150/ijbs.9518] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 06/04/2014] [Indexed: 12/12/2022] Open
Abstract
Erythropoietin (EPO), the required cytokine for promoting the proliferation and differentiation of erythroid cells to stimulate erythropoiesis, has been reported to act as a pleiotropic cytokine beyond hematopoietic system. The various activities of EPO are determined by the widespread distribution of its cell surface EPO receptor (EpoR) in multiple tissues including endothelial, neural, myoblasts, adipocytes and other cell types. EPO activity has been linked to angiogenesis, neuroprotection, cardioprotection, stress protection, anti-inflammation and especially the energy metabolism regulation that is recently revealed. The investigations of EPO activity in animals and the expression analysis of EpoR provide more insights on the potential of EPO in regulating energy metabolism and homeostasis. The findings of crosstalk between EPO and some important energy sensors and the regulation of EPO in the cellular respiration and mitochondrial function further provide molecular mechanisms for EPO activity in metabolic activity regulation. In this review, we will summarize the roles of EPO in energy metabolism regulation and the activity of EPO in tissues that are tightly associated with energy metabolism. We will also discuss the effects of EPO in regulating oxidative metabolism and mitochondrial function, the interactions between EPO and important energy regulation factors, and the protective role of EPO from stresses that are related to metabolism, providing a brief overview of previously less appreciated EPO biological function in energy metabolism and homeostasis.
Collapse
Affiliation(s)
- Li Wang
- 1. Faculty of Health Sciences, University of Macau, SAR of People's Republic of China
| | - Lijun Di
- 1. Faculty of Health Sciences, University of Macau, SAR of People's Republic of China
| | - Constance Tom Noguchi
- 2. Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, U.S.A
| |
Collapse
|
199
|
Verma R, Su S, McCrann DJ, Green JM, Leu K, Young PR, Schatz PJ, Silva JC, Stokes MP, Wojchowski DM. RHEX, a novel regulator of human erythroid progenitor cell expansion and erythroblast development. ACTA ACUST UNITED AC 2014; 211:1715-22. [PMID: 25092874 PMCID: PMC4144737 DOI: 10.1084/jem.20130624] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
RHEX is a novel target of the human erythropoietin receptor, and modulator of EPO-dependent red cell production. Ligation of erythropoietin (EPO) receptor (EPOR) JAK2 kinase complexes propagates signals within erythroid progenitor cells (EPCs) that are essential for red blood cell production. To reveal hypothesized novel EPOR/JAK2 targets, a phosphotyrosine (PY) phosphoproteomics approach was applied. Beyond known signal transduction factors, 32 new targets of EPO-modulated tyrosine phosphorylation were defined. Molecular adaptors comprised one major set including growth factor receptor-bound protein 2 (GRB2)–associated binding proteins 1–3 (GAB1-3), insulin receptor substrate 2 (IRS2), docking protein 1 (DOK1), Src homology 2 domain containing transforming protein 1 (SHC1), and sprouty homologue 1 (SPRY1) as validating targets, and SPRY2, SH2 domain containing 2A (SH2D2A), and signal transducing adaptor molecule 2 (STAM2) as novel candidate adaptors together with an ORF factor designated as regulator of human erythroid cell expansion (RHEX). RHEX is well conserved in Homo sapiens and primates but absent from mouse, rat, and lower vertebrate genomes. Among tissues and lineages, RHEX was elevated in EPCs, occurred as a plasma membrane protein, was rapidly PY-phosphorylated >20-fold upon EPO exposure, and coimmunoprecipitated with the EPOR. In UT7epo cells, knockdown of RHEX inhibited EPO-dependent growth. This was associated with extracellular signal-regulated kinase 1,2 (ERK1,2) modulation, and RHEX coupling to GRB2. In primary human EPCs, shRNA knockdown studies confirmed RHEX regulation of erythroid progenitor expansion and further revealed roles in promoting the formation of hemoglobinizing erythroblasts. RHEX therefore comprises a new EPO/EPOR target and regulator of human erythroid cell expansion that additionally acts to support late-stage erythroblast development.
Collapse
Affiliation(s)
- Rakesh Verma
- COBRE Center of Excellence in Stem Cell Biology and Regenerative Medicine, Maine Medical Center Research Institute, Scarborough ME, 04074
| | - Su Su
- COBRE Center of Excellence in Stem Cell Biology and Regenerative Medicine, Maine Medical Center Research Institute, Scarborough ME, 04074
| | - Donald J McCrann
- COBRE Center of Excellence in Stem Cell Biology and Regenerative Medicine, Maine Medical Center Research Institute, Scarborough ME, 04074
| | | | | | | | | | | | | | - Don M Wojchowski
- COBRE Center of Excellence in Stem Cell Biology and Regenerative Medicine, Maine Medical Center Research Institute, Scarborough ME, 04074
| |
Collapse
|
200
|
Wan L, Zhang F, He Q, Tsang WP, Lu L, Li Q, Wu Z, Qiu G, Zhou G, Wan C. EPO promotes bone repair through enhanced cartilaginous callus formation and angiogenesis. PLoS One 2014; 9:e102010. [PMID: 25003898 PMCID: PMC4087003 DOI: 10.1371/journal.pone.0102010] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 06/13/2014] [Indexed: 12/13/2022] Open
Abstract
Erythropoietin (EPO)/erythropoietin receptor (EPOR) signaling is involved in the development and regeneration of several non-hematopoietic tissues including the skeleton. EPO is identified as a downstream target of the hypoxia inducible factor-α (HIF-α) pathway. It is shown that EPO exerts a positive role in bone repair, however, the underlying cellular and molecular mechanisms remain unclear. In the present study we show that EPO and EPOR are expressed in the proliferating, pre-hypertrophic and hypertrophic zone of the developing mouse growth plates as well as in the cartilaginous callus of the healing bone. The proliferation rate of chondrocytes is increased under EPO treatment, while this effect is decreased following siRNA mediated knockdown of EPOR in chondrocytes. EPO treatment increases biosynthesis of proteoglycan, accompanied by up-regulation of chondrogenic marker genes including SOX9, SOX5, SOX6, collagen type 2, and aggrecan. The effects are inhibited by knockdown of EPOR. Blockage of the endogenous EPO in chondrocytes also impaired the chondrogenic differentiation. In addition, EPO promotes metatarsal endothelial sprouting in vitro. This coincides with the in vivo data that local delivery of EPO increases vascularity at the mid-stage of bone healing (day 14). In a mouse femoral fracture model, EPO promotes cartilaginous callus formation at days 7 and 14, and enhances bone healing at day 28 indexed by improved X-ray score and micro-CT analysis of microstructure of new bone regenerates, which results in improved biomechanical properties. Our results indicate that EPO enhances chondrogenic and angiogenic responses during bone repair. EPO's function on chondrocyte proliferation and differentiation is at least partially mediated by its receptor EPOR. EPO may serve as a therapeutic agent to facilitate skeletal regeneration.
Collapse
Affiliation(s)
- Lin Wan
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fengjie Zhang
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Qiling He
- Departments of Microbiology and Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Wing Pui Tsang
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Li Lu
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, New Drug Function Research Center, School of Life Science and Biopharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qingnan Li
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, New Drug Function Research Center, School of Life Science and Biopharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhihong Wu
- Department of Orthopaedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Guixing Qiu
- Department of Orthopaedics, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Guangqian Zhou
- The Center for Anti-Ageing and Regenerative Medicine, Medical School, Shenzhen University, Shenzhen, China
| | - Chao Wan
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences Core Laboratory, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
- * E-mail:
| |
Collapse
|