51
|
Fouquet G, Thongsa-Ad U, Lefevre C, Rousseau A, Tanhuad N, Khongkla E, Saengsawang W, Anurathapan U, Hongeng S, Maciel TT, Hermine O, Bhukhai K. Iron-loaded transferrin potentiates erythropoietin effects on erythroblast proliferation and survival: a novel role through transferrin receptors. Exp Hematol 2021; 99:12-20.e3. [PMID: 34077792 DOI: 10.1016/j.exphem.2021.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/16/2021] [Accepted: 05/26/2021] [Indexed: 12/21/2022]
Abstract
Red blood cell production, or erythropoiesis, is a proliferative process that requires tight regulation. Erythropoietin (Epo) is a glycoprotein cytokine that plays a major role in erythropoiesis by triggering erythroid progenitors/precursors of varying sensitivity. The concentration of Epo in bone marrow is hypothesized to be suboptimal, and the survival of erythroid cells has been suggested to depend on Epo sensitivity. However, the key factors that control Epo sensitivity remain unknown. Two types of transferrin receptors (TfRs), TfR1 and TfR2, are known to play a role in iron uptake in erythroid cells. Here, we hypothesized that TfRs may additionally modulate Epo sensitivity during erythropoiesis by modulating Epo receptor (EpoR) signaling. Using an Epo-sensitive UT-7 (UT7/Epo) erythroid cell and human erythroid progenitor cell models, we report that iron-loaded transferrin, that is, holo-transferrin (holo-Tf), synergizes with suboptimal Epo levels to improve erythroid cell survival, proliferation, and differentiation. This is accomplished via the major signaling pathways of erythropoiesis, which include signal transducer and activator of transcription 5 (STAT5), mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK), and phosphoinositide-3-kinase (PI3K)/AKT. Furthermore, we found that this cooperation is improved by, but does not require, the internalization of TfR1. Interestingly, we observed that loss of TfR2 stabilizes EpoR levels and abolishes the beneficial effects of holo-Tf. Overall, these data reveal novel signaling properties of TfRs, which involve the regulation of erythropoiesis through EpoR signaling.
Collapse
Affiliation(s)
- Guillemette Fouquet
- Institut Hospitalo-Universitaire (IHU) Imagine, Université Sorbonne Paris cité, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France; INSERM U1163, Université Paris Descartes, Faculté de Médecine, Hôpital Necker, Paris, France; Laboratory of Excellence GReX, Paris, France
| | | | - Carine Lefevre
- Laboratory of Excellence GReX, Paris, France; INSERM U1016, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Alice Rousseau
- INSERM U1016, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Nopmullee Tanhuad
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Ekkaphot Khongkla
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Witchuda Saengsawang
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Usanarat Anurathapan
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Thiago T Maciel
- Institut Hospitalo-Universitaire (IHU) Imagine, Université Sorbonne Paris cité, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France; INSERM U1163, Université Paris Descartes, Faculté de Médecine, Hôpital Necker, Paris, France; Laboratory of Excellence GReX, Paris, France
| | - Olivier Hermine
- Institut Hospitalo-Universitaire (IHU) Imagine, Université Sorbonne Paris cité, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France; INSERM U1163, Université Paris Descartes, Faculté de Médecine, Hôpital Necker, Paris, France; Laboratory of Excellence GReX, Paris, France; Service d'Hématologie clinique adultes, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France
| | - Kanit Bhukhai
- Institut Hospitalo-Universitaire (IHU) Imagine, Université Sorbonne Paris cité, Assistance Publique-Hôpitaux de Paris, Hôpital Necker, Paris, France; INSERM U1163, Université Paris Descartes, Faculté de Médecine, Hôpital Necker, Paris, France; Laboratory of Excellence GReX, Paris, France; Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
52
|
Correa-Gallegos D, Jiang D, Rinkevich Y. Fibroblasts as confederates of the immune system. Immunol Rev 2021; 302:147-162. [PMID: 34036608 DOI: 10.1111/imr.12972] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 12/18/2022]
Abstract
Fibroblastic stromal cells are as diverse, in origin and function, as the niches they fashion in the mammalian body. This cellular variety impacts the spectrum of responses elicited by the immune system. Fibroblast influence on the immune system keeps evolving our perspective on fibroblast roles and functions beyond just a passive structural part of organs. This review discusses the foundations of fibroblastic stromal-immune crosstalk, under the scope of stromal heterogeneity as a basis for tissue-specific tutoring of the immune system. Focusing on the skin as a relevant immunological organ, we detail the complex interactions between distinct fibroblast populations and immune cells that occur during homeostasis, injury repair, scarring, and disease. We further review the relevance of fibroblastic stromal cell heterogeneity and how this heterogeneity is central to regulate the immune system from its inception during embryonic development into adulthood.
Collapse
Affiliation(s)
- Donovan Correa-Gallegos
- Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München, Munich, Germany
| | - Dongsheng Jiang
- Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München, Munich, Germany
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| |
Collapse
|
53
|
Turnis ME, Kaminska E, Smith KH, Kartchner BJ, Vogel P, Laxton JD, Ashmun RA, Ney PA, Opferman JT. Requirement for antiapoptotic MCL-1 during early erythropoiesis. Blood 2021; 137:1945-1958. [PMID: 33512417 PMCID: PMC8033457 DOI: 10.1182/blood.2020006916] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 12/22/2020] [Indexed: 01/24/2023] Open
Abstract
Although BCL-xL is critical to the survival of mature erythrocytes, it is still unclear whether other antiapoptotic molecules mediate survival during earlier stages of erythropoiesis. Here, we demonstrate that erythroid-specific Mcl1 deletion results in embryonic lethality beyond embryonic day 13.5 as a result of severe anemia caused by a lack of mature red blood cells (RBCs). Mcl1-deleted embryos exhibit stunted growth, ischemic necrosis, and decreased RBCs in the blood. Furthermore, we demonstrate that MCL-1 is only required during early definitive erythropoiesis; during later stages, developing erythrocytes become MCL-1 independent and upregulate the expression of BCL-xL. Functionally, MCL-1 relies upon its ability to prevent apoptosis to promote erythroid development because codeletion of the proapoptotic effectors Bax and Bak can overcome the requirement for MCL-1 expression. Furthermore, ectopic expression of human BCL2 in erythroid progenitors can compensate for Mcl1 deletion, indicating redundancy between these 2 antiapoptotic family members. These data clearly demonstrate a requirement for MCL-1 in promoting survival of early erythroid progenitors.
Collapse
Affiliation(s)
| | | | | | | | | | - Jonathan D Laxton
- Flow Cytometry and Cell Sorting Shared Resource, St Jude Children's Research Hospital, Memphis, TN; and
| | - Richard A Ashmun
- Flow Cytometry and Cell Sorting Shared Resource, St Jude Children's Research Hospital, Memphis, TN; and
| | | | | |
Collapse
|
54
|
Richard C, Viret S, Cantero Aguilar L, Lefevre C, Leduc M, Faouzi EH, Azar N, Lavazec C, Mayeux P, Verdier F. Myotonic dystrophy kinase-related CDC42-binding kinase α, a new transferrin receptor type 2-binding partner, is a regulator of erythropoiesis. Am J Hematol 2021; 96:480-492. [PMID: 33476437 DOI: 10.1002/ajh.26104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 01/01/2023]
Abstract
Efficient erythropoiesis relies on the expression of the transferrin receptor type 2 (TFR2). In erythroid precursors, TFR2 facilitates the export of the erythropoietin receptor (EPOR) to cell surface, which ensures the survival and proliferation of erythroblasts. Although TFR2 has a crucial role in erythropoiesis regulation, its mechanism of action remains to be clarified. To understand its role better, we aimed at identifying its protein partners by mass-spectrometry after immunoprecipitation in erythroid cells. Here we report the kinase MRCKα (myotonic dystrophy kinase-related CDC42-binding kinase α) as a new partner of both TFR2 and EPOR in erythroblasts. We show that MRCKα is co-expressed with TFR2, and TFR1 during terminal differentiation and regulates the internalization of the two types of transferrin receptors. The knockdown of MRCKα by shRNA in human primary erythroblasts leads to a decreased cell surface expression of both TFR1 and TFR2, an increased cell-surface expression of EPOR, and a delayed differentiation. Additionally, knockout of Mrckα in the murine MEDEP cells also leads to a striking delay in erythropoiesis, showcasing the importance of this kinase in both species. Our data highlight the importance of MRCKα in the regulation of erythropoiesis.
Collapse
Affiliation(s)
- Cyrielle Richard
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Sophie Viret
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Lilia Cantero Aguilar
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Carine Lefevre
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Marjorie Leduc
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
- Plateforme Protéomique 3P5‐Proteom'IC, Université de Paris, Institut Cochin, INSERM, U1016, CNRS UMR8104 Paris France
| | - El Hassan Faouzi
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Nabih Azar
- Unité d'Hémobiothérapie, Hôpital La Pitié Salpêtrière Paris France
| | - Catherine Lavazec
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Patrick Mayeux
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
- Plateforme Protéomique 3P5‐Proteom'IC, Université de Paris, Institut Cochin, INSERM, U1016, CNRS UMR8104 Paris France
| | - Frédérique Verdier
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| |
Collapse
|
55
|
Megakaryocyte TGFβ1 partitions erythropoiesis into immature progenitor/stem cells and maturing precursors. Blood 2021; 136:1044-1054. [PMID: 32548608 DOI: 10.1182/blood.2019003276] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 05/01/2020] [Indexed: 01/06/2023] Open
Abstract
Erythropoietin (EPO) provides the major survival signal to maturing erythroid precursors (EPs) and is essential for terminal erythropoiesis. Nonetheless, progenitor cells can irreversibly commit to an erythroid fate well before EPO acts, risking inefficiency if these progenitors are unneeded to maintain red blood cell (RBC) counts. We identified a new modular organization of erythropoiesis and, for the first time, demonstrate that the pre-EPO module is coupled to late EPO-dependent erythropoiesis by megakaryocyte (Mk) signals. Disrupting megakaryocytic transforming growth factor β1 (Tgfb1) disorganized hematopoiesis by expanding the pre-EPO pool of progenitor cells and consequently triggering significant apoptosis of EPO-dependent EPs. Similarly, pharmacologic blockade of TGFβ signaling in normal mice boosted the pre-EPO module, leading to apoptosis of EPO-sensitive EPs. Subsequent treatment with low-dose EPO triggered robust RBC production in both models. This work reveals modular regulation of erythropoiesis and offers a new strategy for overcoming chronic anemias.
Collapse
|
56
|
Swann JW, Koneva LA, Regan-Komito D, Sansom SN, Powrie F, Griseri T. IL-33 promotes anemia during chronic inflammation by inhibiting differentiation of erythroid progenitors. J Exp Med 2021; 217:151849. [PMID: 32520308 PMCID: PMC7478740 DOI: 10.1084/jem.20200164] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/10/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022] Open
Abstract
An important comorbidity of chronic inflammation is anemia, which may be related to dysregulated activity of hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM). Among HSPCs, we found that the receptor for IL-33, ST2, is expressed preferentially and highly on erythroid progenitors. Induction of inflammatory spondyloarthritis in mice increased IL-33 in BM plasma, and IL-33 was required for inflammation-dependent suppression of erythropoiesis in BM. Conversely, administration of IL-33 in healthy mice suppressed erythropoiesis, decreased hemoglobin expression, and caused anemia. Using purified erythroid progenitors in vitro, we show that IL-33 directly inhibited terminal maturation. This effect was dependent on NF-κB activation and associated with altered signaling events downstream of the erythropoietin receptor. Accordingly, IL-33 also suppressed erythropoietin-accelerated erythropoiesis in vivo. These results reveal a role for IL-33 in pathogenesis of anemia during inflammatory disease and define a new target for its treatment.
Collapse
Affiliation(s)
- James W Swann
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Lada A Koneva
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | - Stephen N Sansom
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Thibault Griseri
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
57
|
Menon V, Ghaffari S. Erythroid enucleation: a gateway into a "bloody" world. Exp Hematol 2021; 95:13-22. [PMID: 33440185 PMCID: PMC8147720 DOI: 10.1016/j.exphem.2021.01.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 12/18/2022]
Abstract
Erythropoiesis is an intricate process starting in hematopoietic stem cells and leading to the daily production of 200 billion red blood cells (RBCs). Enucleation is a greatly complex and rate-limiting step during terminal maturation of mammalian RBC production involving expulsion of the nucleus from the orthochromatic erythroblasts, resulting in the formation of reticulocytes. The dynamic enucleation process involves many factors ranging from cytoskeletal proteins to transcription factors to microRNAs. Lack of optimum terminal erythroid maturation and enucleation has been an impediment to optimum RBC production ex vivo. Major efforts in the past two decades have exposed some of the mechanisms that govern the enucleation process. This review focuses in detail on mechanisms implicated in enucleation and discusses the future perspectives of this fascinating process.
Collapse
Affiliation(s)
- Vijay Menon
- Department of Cell, Developmental & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Saghi Ghaffari
- Department of Cell, Developmental & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY; Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
58
|
Miyauchi K, Nakai T, Saito S, Yamamoto T, Sato K, Kato K, Nezu M, Miyazaki M, Ito S, Yamamoto M, Suzuki N. Renal interstitial fibroblasts coproduce erythropoietin and renin under anaemic conditions. EBioMedicine 2021; 64:103209. [PMID: 33508746 PMCID: PMC7841315 DOI: 10.1016/j.ebiom.2021.103209] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 12/08/2020] [Accepted: 01/04/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Erythrocyte mass contributes to maintaining systemic oxygen delivery and blood viscosity, with the latter being one of the determinants of blood pressure. However, the physiological response to blood pressure changes under anaemic conditions remain unknown. METHODS AND FINDINGS We show that anaemia decreases blood pressure in human patients and mouse models. Analyses of pathways related to blood pressure regulation demonstrate that anaemia enhances the expression of the gene encoding the vasopressor substance renin in kidneys. Although kidney juxtaglomerular cells are known to continuously produce renin, renal interstitial fibroblasts are identified in the present study as a novel site of renin induction under anaemic hypotensive conditions in mice and rats. Notably, some renal interstitial fibroblasts are found to simultaneously express renin and the erythroid growth factor erythropoietin in the anaemic mouse kidney. Antihypertensive agents but not hypoxic stimuli induced interstitial renin expression, suggesting that blood pressure reduction triggers interstitial renin induction in anaemic mice. The interstitial renin expression was also detected in injured fibrotic kidneys of the mouse and human, and the renin-expressing interstitial cells in murine fibrotic kidneys were identified as myofibroblasts originating from renal interstitial fibroblasts. Since the elevated expression levels of renin in fibrotic kidneys along with progression of renal fibrosis were well correlated to the systemic blood pressure increase, the renal interstitial renin production seemed to affect systemic blood pressure. INTERPRETATION Renal interstitial fibroblasts function as central controllers of systemic oxygen delivery by producing both renin and erythropoietin. FUNDING Grants-in-Aid from Japan Society for the Promotion of Science (JSPS) KAKENHI (17K19680, 15H04691, and 26111002) and the Takeda Science Foundation.
Collapse
Affiliation(s)
- Kenichiro Miyauchi
- Division of Oxygen Biology, United Centres for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan; Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Taku Nakai
- Division of Oxygen Biology, United Centres for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Sakae Saito
- Tohoku Medical Megabank Organization, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Tae Yamamoto
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Koji Sato
- Division of Oxygen Biology, United Centres for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan; Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Koichiro Kato
- Division of Oxygen Biology, United Centres for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Masahiro Nezu
- Division of Oxygen Biology, United Centres for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan; Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Japan; Tohoku Medical Megabank Organization, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Mariko Miyazaki
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Sadayoshi Ito
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Masayuki Yamamoto
- Tohoku Medical Megabank Organization, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Norio Suzuki
- Division of Oxygen Biology, United Centres for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Seiryo-machi 2-1, Aoba-ku, Sendai, Miyagi 980-8575, Japan.
| |
Collapse
|
59
|
Yamazaki S, Hirano I, Kato K, Yamamoto M, Suzuki N. Defining the functionally sufficient regulatory region and liver-specific roles of the erythropoietin gene by transgene complementation. Life Sci 2021; 269:119075. [PMID: 33465391 DOI: 10.1016/j.lfs.2021.119075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/24/2020] [Accepted: 01/08/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Erythropoietin (EPO) is an essential growth factor for erythroid cells and is mainly secreted from the kidneys and subsidiarily from the livers of adult mammals in an anemia/hypoxia-inducible manner. AIM AND METHOD To elucidate the regulatory mechanisms of stress-inducible and cell type-specific Epo gene transcription, the rate-limiting step of EPO production, we investigated the sufficiency of a 180-kb genomic fragment flanking the mouse Epo gene locus for recapitulating endogenous Epo gene function by a transgene complementation strategy. KEY FINDINGS While Epo gene-deficient mice exhibited lethal anemia in utero with defects in erythroblast proliferation and maturation, Epo-knockout mice integrated with the 180-kb Epo transgene showed normal erythropoiesis throughout life. In the transgene-rescued mice, liver-specific deletion of the transgene by the Cre-loxP recombination system caused neonatal anemia with erythropoietic defects in the liver but not in the spleen, indicating the essential function of hepatic EPO on normal erythropoiesis in the liver, which is the major erythropoietic site in late embryonic and neonatal stages. SIGNIFICANCE These results demonstrate that the 180 kb Epo gene flanking region contains the fully functional Epo gene unit and that EPO from the liver dominantly stimulates hepatic erythropoiesis but contributes less to erythropoiesis in other organs.
Collapse
Affiliation(s)
- Shun Yamazaki
- Division of Oxygen Biology, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan; Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ikuo Hirano
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Koichiro Kato
- Division of Oxygen Biology, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Norio Suzuki
- Division of Oxygen Biology, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
| |
Collapse
|
60
|
Bretz CA, Ramshekar A, Kunz E, Wang H, Hartnett ME. Signaling Through the Erythropoietin Receptor Affects Angiogenesis in Retinovascular Disease. Invest Ophthalmol Vis Sci 2021; 61:23. [PMID: 32785675 PMCID: PMC7441364 DOI: 10.1167/iovs.61.10.23] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Purpose Exogenous erythropoietin (EPO) is being considered for tissue protection and angiogenesis in retinal vascular diseases. However, studies are limited by insufficient tools to address signaling effects through the EPO receptor (EPOR). We used a humanized mouse model of hypoactive EPOR signaling to test the hypothesis that EPOR signaling supports angiogenesis in retinovascular diseases. Methods Humanized Knockin EPOR mice (hWtEPOR) with hypoactive EPOR signaling were compared to littermate wild-type mice (WT). Postnatal day (p)7 mice of each genotype were exposed to 75% oxygen for five days, followed by 21% oxygen in the oxygen-induced retinopathy model (OIR) and compared to room-air (RA)–raised pups. At time points after OIR, pups were sacrificed, and flat-mounted, lectin-stained retinas were analyzed for central avascular area or intravitreal neovascular area (IVNV). Flash-frozen retinas were analyzed for angiogenic protein (Epo, VEGF, p-Stat3) and gene (Vegfa, Kdr, Epo, Hif1α, Hif2α) expression levels. Results In OIR, hWtEPOR mice had increased AVA compared with WT at p8, p12, and p17, but there was no difference in IVNV between hWtEPOR and WT mice at p17. Although VEGF and p-STAT3 proteins were increased in WT at p17 OIR, there were no differences in retinal angiogenic factor expression levels between hWtEPOR and WT OIR at p17 despite similar areas of IVNV. Conclusions Our data support the hypothesis that EPOR signaling was associated with regrowth of vascularization following oxygen-induced capillary dropout and played a role in intravitreal angiogenesis. Additional study of EPOR signaling regulation on other angiogenic factor pathways may be considered.
Collapse
Affiliation(s)
- Colin A Bretz
- John A. Moran Eye Center, University of Utah, Salt Lake City, Utah, United States
| | - Aniket Ramshekar
- John A. Moran Eye Center, University of Utah, Salt Lake City, Utah, United States
| | - Eric Kunz
- John A. Moran Eye Center, University of Utah, Salt Lake City, Utah, United States
| | - Haibo Wang
- John A. Moran Eye Center, University of Utah, Salt Lake City, Utah, United States
| | - M Elizabeth Hartnett
- John A. Moran Eye Center, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
61
|
Kwon N, Thompson EN, Mayday MY, Scanlon V, Lu YC, Krause DS. Current understanding of human megakaryocytic-erythroid progenitors and their fate determinants. Curr Opin Hematol 2021; 28:28-35. [PMID: 33186151 PMCID: PMC7737300 DOI: 10.1097/moh.0000000000000625] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW This review focuses on our current understanding of fate decisions in bipotent megakaryocyte-erythroid progenitors (MEPs). Although extensive research has been carried out over decades, our understanding of how MEP commit to the erythroid versus megakaryocyte fate remains unclear. RECENT FINDINGS We discuss the isolation of primary human MEP, and focus on gene expression patterns, epigenetics, transcription factors and extrinsic factors that have been implicated in MEP fate determination. We conclude with an overview of the open debates in the field of MEP biology. SUMMARY Understanding MEP fate is important because defects in megakaryocyte and erythrocyte development lead to disease states such as anaemia, thrombocytopenia and leukaemia. MEP also represent a model system for studying fundamental principles underlying cell fate decisions of bipotent and pluripotent progenitors, such that discoveries in MEP are broadly applicable to stem/progenitor cell biology.
Collapse
Affiliation(s)
- Nayoung Kwon
- Department of Cell Biology, Yale School of Medicine, 333 Cedar Street, New Haven, CT
- Yale Stem Cell Center, Yale School of Medicine, 333 Cedar Street, New Haven, CT
| | - Evrett N. Thompson
- Department of Cell Biology, Yale School of Medicine, 333 Cedar Street, New Haven, CT
- Yale Stem Cell Center, Yale School of Medicine, 333 Cedar Street, New Haven, CT
| | - Madeline Y. Mayday
- Yale Stem Cell Center, Yale School of Medicine, 333 Cedar Street, New Haven, CT
- Department of Pathology, Yale School of Medicine, 333 Cedar Street, New Haven, CT
| | - Vanessa Scanlon
- Yale Stem Cell Center, Yale School of Medicine, 333 Cedar Street, New Haven, CT
- Department of Laboratory Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT
| | - Yi-Chien Lu
- Yale Stem Cell Center, Yale School of Medicine, 333 Cedar Street, New Haven, CT
- Department of Pathology, Yale School of Medicine, 333 Cedar Street, New Haven, CT
| | - Diane S. Krause
- Department of Cell Biology, Yale School of Medicine, 333 Cedar Street, New Haven, CT
- Yale Stem Cell Center, Yale School of Medicine, 333 Cedar Street, New Haven, CT
- Department of Pathology, Yale School of Medicine, 333 Cedar Street, New Haven, CT
- Department of Laboratory Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT
| |
Collapse
|
62
|
Cellular Basis of Embryonic Hematopoiesis and Its Implications in Prenatal Erythropoiesis. Int J Mol Sci 2020; 21:ijms21249346. [PMID: 33302450 PMCID: PMC7763178 DOI: 10.3390/ijms21249346] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/04/2020] [Accepted: 12/05/2020] [Indexed: 01/02/2023] Open
Abstract
Primitive erythrocytes are the first hematopoietic cells observed during ontogeny and are produced specifically in the yolk sac. Primitive erythrocytes express distinct hemoglobins compared with adult erythrocytes and circulate in the blood in the nucleated form. Hematopoietic stem cells produce adult-type (so-called definitive) erythrocytes. However, hematopoietic stem cells do not appear until the late embryonic/early fetal stage. Recent studies have shown that diverse types of hematopoietic progenitors are present in the yolk sac as well as primitive erythroblasts. Multipotent hematopoietic progenitors that arose in the yolk sac before hematopoietic stem cells emerged likely fill the gap between primitive erythropoiesis and hematopoietic stem-cell-originated definitive erythropoiesis and hematopoiesis. In this review, we discuss the cellular origin of primitive erythropoiesis in the yolk sac and definitive hematopoiesis in the fetal liver. We also describe mechanisms for developmental switches that occur during embryonic and fetal erythropoiesis and hematopoiesis, particularly focusing on recent studies performed in mice.
Collapse
|
63
|
Lin Z, King R, Tang V, Myers G, Balbin-Cuesta G, Friedman A, McGee B, Desch K, Ozel AB, Siemieniak D, Reddy P, Emmer B, Khoriaty R. The Endoplasmic Reticulum Cargo Receptor SURF4 Facilitates Efficient Erythropoietin Secretion. Mol Cell Biol 2020; 40:e00180-20. [PMID: 32989016 PMCID: PMC7652404 DOI: 10.1128/mcb.00180-20] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/20/2020] [Accepted: 09/24/2020] [Indexed: 12/16/2022] Open
Abstract
Erythropoietin (EPO) stimulates erythroid differentiation and maturation. Though the transcriptional regulation of EPO has been well studied, the molecular determinants of EPO secretion remain unknown. Here, we generated a HEK293T reporter cell line that provides a quantifiable and selectable readout of intracellular EPO levels and performed a genome-scale CRISPR screen that identified SURF4 as an important mediator of EPO secretion. Targeting SURF4 with multiple independent single guide RNAs (sgRNAs) resulted in intracellular accumulation and extracellular depletion of EPO. Both of these phenotypes were rescued by expression of SURF4 cDNA. Additionally, we found that disruption of SURF4 resulted in accumulation of EPO in the endoplasmic reticulum (ER) compartment and that SURF4 and EPO physically interact. Furthermore, SURF4 disruption in Hep3B cells also caused a defect in the secretion of endogenous EPO under conditions mimicking hypoxia, ruling out an artifact of heterologous overexpression. This work demonstrates that SURF4 functions as an ER cargo receptor that mediates the efficient secretion of EPO. Our findings also suggest that modulating SURF4 may be an effective treatment for disorders of erythropoiesis that are driven by aberrant EPO levels. Finally, we show that SURF4 overexpression results in increased secretion of EPO, suggesting a new strategy for more efficient production of recombinant EPO.
Collapse
Affiliation(s)
- Zesen Lin
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Richard King
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Vi Tang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Greggory Myers
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ginette Balbin-Cuesta
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan, USA
- Medical Scientist Training Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Ann Friedman
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Beth McGee
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Karl Desch
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Ayse Bilge Ozel
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - David Siemieniak
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Pavan Reddy
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan, USA
- University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
| | - Brian Emmer
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Rami Khoriaty
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan, USA
- University of Michigan Rogel Cancer Center, Ann Arbor, Michigan, USA
| |
Collapse
|
64
|
Noguchi CT. Erythropoietin regulates metabolic response in mice via receptor expression in adipose tissue, brain, and bone. Exp Hematol 2020; 92:32-42. [PMID: 32950599 DOI: 10.1016/j.exphem.2020.09.190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022]
Abstract
Erythropoietin (EPO) acts by binding to erythroid progenitor cells to regulate red blood cell production. While EPO receptor (Epor) expression is highest on erythroid tissue, animal models exhibit EPO activity in nonhematopoietic tissues, mediated, in part, by tissue-specific Epor expression. This review describes the metabolic response in mice to endogenous EPO and EPO treatment associated with glucose metabolism, fat mass accumulation, and inflammation in white adipose tissue and brain during diet-induced obesity and with bone marrow fat and bone remodeling. During high-fat diet-induced obesity, EPO treatment improves glucose tolerance, decreases fat mass accumulation, and shifts white adipose tissue from a pro-inflammatory to an anti-inflammatory state. Fat mass regulation by EPO is sex dimorphic, apparent in males and abrogated by estrogen in females. Cerebral EPO also regulates fat mass and hypothalamus inflammation associated with diet-induced obesity in males and ovariectomized female mice. In bone, EPO contributes to the balance between adipogenesis and osteogenesis in both male and female mice. EPO treatment promotes bone loss mediated via Epor in osteoblasts and reduces bone marrow adipocytes before and independent of change in white adipose tissue fat mass. EPO regulation of bone loss and fat mass is independent of EPO-stimulated erythropoiesis. EPO nonhematopoietic tissue response may relate to the long-term consequences of EPO treatment of anemia in chronic kidney disease and to the alternative treatment of oral hypoxia-inducible factor prolyl hydroxylase inhibitors that increase endogenous EPO production.
Collapse
Affiliation(s)
- Constance Tom Noguchi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
65
|
Bhoopalan SV, Huang LJS, Weiss MJ. Erythropoietin regulation of red blood cell production: from bench to bedside and back. F1000Res 2020; 9:F1000 Faculty Rev-1153. [PMID: 32983414 PMCID: PMC7503180 DOI: 10.12688/f1000research.26648.1] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/04/2020] [Indexed: 12/18/2022] Open
Abstract
More than 50 years of efforts to identify the major cytokine responsible for red blood cell (RBC) production (erythropoiesis) led to the identification of erythropoietin (EPO) in 1977 and its receptor (EPOR) in 1989, followed by three decades of rich scientific discovery. We now know that an elaborate oxygen-sensing mechanism regulates the production of EPO, which in turn promotes the maturation and survival of erythroid progenitors. Engagement of the EPOR by EPO activates three interconnected signaling pathways that drive RBC production via diverse downstream effectors and simultaneously trigger negative feedback loops to suppress signaling activity. Together, the finely tuned mechanisms that drive endogenous EPO production and facilitate its downstream activities have evolved to maintain RBC levels in a narrow physiological range and to respond rapidly to erythropoietic stresses such as hypoxia or blood loss. Examination of these pathways has elucidated the genetics of numerous inherited and acquired disorders associated with deficient or excessive RBC production and generated valuable drugs to treat anemia, including recombinant human EPO and more recently the prolyl hydroxylase inhibitors, which act partly by stimulating endogenous EPO synthesis. Ongoing structure-function studies of the EPOR and its essential partner, tyrosine kinase JAK2, suggest that it may be possible to generate new "designer" drugs that control selected subsets of cytokine receptor activities for therapeutic manipulation of hematopoiesis and treatment of blood cancers.
Collapse
Affiliation(s)
- Senthil Velan Bhoopalan
- Department of Hematology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS #355, Memphis, TN, 38105, USA
| | - Lily Jun-shen Huang
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Mitchell J. Weiss
- Department of Hematology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS #355, Memphis, TN, 38105, USA
| |
Collapse
|
66
|
Hamza E, Metzinger L, Metzinger-Le Meuth V. Uremic Toxins Affect Erythropoiesis during the Course of Chronic Kidney Disease: A Review. Cells 2020; 9:cells9092039. [PMID: 32899941 PMCID: PMC7565991 DOI: 10.3390/cells9092039] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/26/2020] [Accepted: 09/04/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) is a global health problem characterized by progressive kidney failure due to uremic toxicity and the complications that arise from it. Anemia consecutive to CKD is one of its most common complications affecting nearly all patients with end-stage renal disease. Anemia is a potential cause of cardiovascular disease, faster deterioration of renal failure and mortality. Erythropoietin (produced by the kidney) and iron (provided from recycled senescent red cells) deficiencies are the main reasons that contribute to CKD-associated anemia. Indeed, accumulation of uremic toxins in blood impairs erythropoietin synthesis, compromising the growth and differentiation of red blood cells in the bone marrow, leading to a subsequent impairment of erythropoiesis. In this review, we mainly focus on the most representative uremic toxins and their effects on the molecular mechanisms underlying anemia of CKD that have been studied so far. Understanding molecular mechanisms leading to anemia due to uremic toxins could lead to the development of new treatments that will specifically target the pathophysiologic processes of anemia consecutive to CKD, such as the newly marketed erythropoiesis-stimulating agents.
Collapse
Affiliation(s)
- Eya Hamza
- HEMATIM UR 4666, C.U.R.S, Université de Picardie Jules Verne, CEDEX 1, 80025 Amiens, France; (E.H.); (V.M.-L.M.)
| | - Laurent Metzinger
- HEMATIM UR 4666, C.U.R.S, Université de Picardie Jules Verne, CEDEX 1, 80025 Amiens, France; (E.H.); (V.M.-L.M.)
- Correspondence: ; Tel.: +33-2282-5356
| | - Valérie Metzinger-Le Meuth
- HEMATIM UR 4666, C.U.R.S, Université de Picardie Jules Verne, CEDEX 1, 80025 Amiens, France; (E.H.); (V.M.-L.M.)
- INSERM UMRS 1148, Laboratory for Vascular Translational Science (LVTS), UFR SMBH, Université Sorbonne Paris Nord, CEDEX, 93017 Bobigny, France
| |
Collapse
|
67
|
Xu L, Wu F, Yang L, Wang F, Zhang T, Deng X, Zhang X, Yuan X, Yan Y, Li Y, Yang Z, Yu D. miR-144/451 inhibits c-Myc to promote erythroid differentiation. FASEB J 2020; 34:13194-13210. [PMID: 33319407 DOI: 10.1096/fj.202000941r] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 07/01/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022]
Abstract
Ablation of miR-144/451 disrupts homeostasis of erythropoiesis. Myc, a protooncogenic protein, is essential for erythroblast proliferation but commits rapid downregulation during erythroid maturation. How erythroblasts orchestrate maturation processes through coding and non-coding genes is largely unknown. In this study, we use miR-144/451 knockout mice as in vivo model, G1E, MEL erythroblast lines and erythroblasts from fresh mouse fetal livers as in vitro systems to demonstrate that targeted depletion of miR-144/451 blocks erythroid nuclear condensation and enucleation. This is due, at least in part, to the continued high expression of Myc in erythroblasts when miR-144/451 is absent. Specifically, miR-144/451 directly inhibits Myc in erythroblasts. Loss of miR-144/451 locus derepresses, and thus, increases the expression of Myc. Sustained high levels of Myc in miR-144/451-depleted erythroblasts blocks erythroid differentiation. Moreover, Myc reversely regulates the expression of miR-144/451, forming a positive miR-144/451-Myc feedback to ensure the complete shutoff of Myc during erythropoiesis. Given that erythroid-specific transcription factor GATA1 activates miR-144/451 and inactivates Myc, our findings indicate that GATA1-miR-144/451-Myc network safeguards normal erythroid differentiation. Our findings also demonstrate that disruption of the miR-144/451-Myc crosstalk causes anemia, suggesting that miR-144/451 might be a potential therapeutic target in red cell diseases.
Collapse
Affiliation(s)
- Lei Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China.,Central Laboratory, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Fan Wu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Lei Yang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Fangfang Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Tong Zhang
- Xinghua People's Hospital, Yangzhou University, Xinghua, China
| | - Xintao Deng
- Xinghua People's Hospital, Yangzhou University, Xinghua, China
| | - Xiumei Zhang
- Xinghua People's Hospital, Yangzhou University, Xinghua, China
| | - Xiaoling Yuan
- Yangzhou Maternal and Child Care Service Center, Yangzhou University, Yangzhou, China
| | - Ying Yan
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Yaoyao Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China.,Central Laboratory, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Zhangping Yang
- Department of Animal Science & Technology, Yangzhou University College of Animal Science and Technology, Yangzhou, China
| | - Duonan Yu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China.,Central Laboratory, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China.,Xinghua People's Hospital, Yangzhou University, Xinghua, China
| |
Collapse
|
68
|
Garrido-Trigo A, Salas A. Molecular Structure and Function of Janus Kinases: Implications for the Development of Inhibitors. J Crohns Colitis 2020; 14:S713-S724. [PMID: 32083640 PMCID: PMC7395311 DOI: 10.1093/ecco-jcc/jjz206] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cytokines can trigger multiple signalling pathways, including Janus tyrosine kinases [JAK] and signal transducers and activators of transcription [STATS] pathways. JAKs are cytoplasmic proteins that, following the binding of cytokines to their receptors, transduce the signal by phosphorylating STAT proteins which enter the nuclei and rapidly target gene promoters to regulate gene transcription. Due to the critical involvement of JAK proteins in mediating innate and adaptive immune responses, these family of kinases have become desirable pharmacological targets in inflammatory diseases, including ulcerative colitis and Crohn's disease. In this review we provide an overview of the main cytokines that signal through the JAK/STAT pathway and the available in vivo evidence on mutant or deleted JAK proteins, and discuss the implications of pharmacologically targeting this kinase family in the context of inflammatory diseases.
Collapse
Affiliation(s)
- Alba Garrido-Trigo
- Department of Gastroenterology, Institut d’Investigacions Biomèdiques August Pi i Sunyer [IDIBAPS] – CIBEREHD, Barcelona, Spain
| | - Azucena Salas
- Department of Gastroenterology, Institut d’Investigacions Biomèdiques August Pi i Sunyer [IDIBAPS] – CIBEREHD, Barcelona, Spain,Corresponding author: Azucena Salas, PhD, Inflammatory Bowel Disease Unit, Department of Gastroenterology, Institut d’Investigacions Biomèdiques August Pi i Sunyer [IDIBAPS] – CIBEREHD, Rosselló 149-153, Barcelona 08036, Spain.
| |
Collapse
|
69
|
Hacein-Bey-Abina S, Estienne M, Bessoles S, Echchakir H, Pederzoli-Ribeil M, Chiron A, Aldaz-Carroll L, Leducq V, Zhang Y, Souyri M, Louache F, Abina AM. Erythropoietin is a major regulator of thrombopoiesis in thrombopoietin-dependent and -independent contexts. Exp Hematol 2020; 88:15-27. [PMID: 32721504 DOI: 10.1016/j.exphem.2020.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/18/2020] [Accepted: 07/22/2020] [Indexed: 01/09/2023]
Abstract
Thrombopoietin (TPO), through activation of its cognate receptor Mpl, is the major regulator of platelet production. However, residual platelets observed in TPO- and Mpl-loss-of-function (LOF) mice suggest the existence of an additional factor to TPO in platelet production. As erythropoietin (EPO) exhibited both in vitro megakaryocytic potential, in association with other early-acting cytokines, and in vivo platelet activation activity, we sought to investigate its role in this setting. Here, we used multiple LOF models to decipher the reciprocal role of EPO and TPO in the regulation of platelet production in TPO-LOF and Mpl-LOF mice and of platelet size heterogeneity in wild-type mice. We first identified EPO as the major thrombopoietic factor in the absence of the TPO-Mpl pathway. Based on the study of several mouse models we found that the EPO-EPO receptor pathway acts on late-stage megakaryopoiesis and is responsible for large-sized platelet production, while the TPO-Mpl pathway promotes small-sized platelet production. On the basis of our data, EPO might be used for thrombocytopenia supportive therapy in congenital amegakaryocytopoiesis. Furthermore, as a distribution skewed toward large platelets is an independent risk factor and a poor prognosis indicator in atherothrombosis, the characterization of EPO's role in the production of large-sized platelets, if confirmed in humans, may open new perspectives in the understanding of the role of EPO-induced platelets in atherothrombosis.
Collapse
Affiliation(s)
- Salima Hacein-Bey-Abina
- UTCBS CNRS UMR 8258, INSERM U1267, Faculté de Pharmacie de Paris, Université de Paris, Paris, France; Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France.
| | | | - Stéphanie Bessoles
- UTCBS CNRS UMR 8258, INSERM U1267, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | | | - Magali Pederzoli-Ribeil
- UTCBS CNRS UMR 8258, INSERM U1267, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | - Andrada Chiron
- UTCBS CNRS UMR 8258, INSERM U1267, Faculté de Pharmacie de Paris, Université de Paris, Paris, France; Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le-Kremlin-Bicêtre, France
| | - Lydia Aldaz-Carroll
- UTCBS CNRS UMR 8258, INSERM U1267, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | | | - Yanyan Zhang
- INSERM U1170, CNRS GDR 3697 MicroNiT, Institut Gustave Roussy, Villejuif, France
| | - Michèle Souyri
- INSERM UMRS 1131, Institut Universitaire d'Hématologie, Université Paris Diderot, Hôpital Saint Louis, Paris, France
| | - Fawzia Louache
- INSERM U1170, CNRS GDR 3697 MicroNiT, Institut Gustave Roussy, Villejuif, France; Université Paris-Sud, Orsay, France
| | - Amine M Abina
- UTCBS CNRS UMR 8258, INSERM U1267, Faculté de Pharmacie de Paris, Université de Paris, Paris, France; NOKAD, Evry, France.
| |
Collapse
|
70
|
Zhong L, Zhang H, Ding ZF, Li J, Lv JW, Pan ZJ, Xu DX, Yin ZS. Erythropoietin-Induced Autophagy Protects Against Spinal Cord Injury and Improves Neurological Function via the Extracellular-Regulated Protein Kinase Signaling Pathway. Mol Neurobiol 2020; 57:3993-4006. [PMID: 32647973 DOI: 10.1007/s12035-020-01997-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022]
Abstract
The objective of this study was to explore the neuroprotective molecular mechanisms of erythropoietin (EPO) in rats following spinal cord injury (SCI). First, a standard SCI model was established. After drug or saline treatment was administered, locomotor function was evaluated in rats using the Basso, Beattie, and Bresnahan (BBB) locomotor rating scale. H&E, Nissl, and TUNEL staining were performed to assess the ratio of cavities, number of motor neurons, and apoptotic cells in the damaged area. The relative protein and mRNA expressions were examined using western blot and qRT-PCR analyses, and the inflammatory markers, axon special protein, and neuromuscular junctions (NMJs) were detected by immunofluorescence. Both doses of EPO notably improved locomotor function, but high-dose EPO was more effective than low-dose EPO. Moreover, EPO reduced the cavity ratio, cell apoptosis, and motor neuron loss in the damaged area, but enhanced the autophagy level and extracellular-regulated protein kinase (ERK) activity. Treatment with an ERK inhibitor significantly prevented the effect of EPO on SCI, and an activator mimicked the benefits of EPO. Further investigation revealed that EPO promoted SCI-induced autophagy via the ERK signaling pathway. EPO activates autophagy to promote locomotor function recovery in rats with SCI via the ERK signaling pathway.
Collapse
Affiliation(s)
- Lin Zhong
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, China.,Department of Orthopedics, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Hui Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, China
| | - Zheng-Fei Ding
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, China
| | - Jian Li
- Department of Toxicology, School of Public Health, Anhui Medical University, #81 Mei Shan Road, Hefei, 230032, China.,Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Jin-Wei Lv
- Department of Toxicology, School of Public Health, Anhui Medical University, #81 Mei Shan Road, Hefei, 230032, China.,Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Zheng-Jun Pan
- Department of Orthopedics, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - De-Xiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, #81 Mei Shan Road, Hefei, 230032, China. .,Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China.
| | - Zong-Sheng Yin
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, China.
| |
Collapse
|
71
|
Nigra AD, Casale CH, Santander VS. Human erythrocytes: cytoskeleton and its origin. Cell Mol Life Sci 2020; 77:1681-1694. [PMID: 31654099 PMCID: PMC11105037 DOI: 10.1007/s00018-019-03346-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/27/2019] [Accepted: 10/16/2019] [Indexed: 01/02/2023]
Abstract
In the last few years, erythrocytes have emerged as the main determinant of blood rheology. In mammals, these cells are devoid of nuclei and are, therefore, unable to divide. Consequently, all circulating erythrocytes come from erythropoiesis, a process in the bone marrow in which several modifications are induced in the expression of membrane and cytoskeletal proteins, and different vertical and horizontal interactions are established between them. Cytoskeleton components play an important role in this process, which explains why they and the interaction between them have been the focus of much recent research. Moreover, in mature erythrocytes, the cytoskeleton integrity is also essential, because the cytoskeleton confers remarkable deformability and stability on the erythrocytes, thus enabling them to undergo deformation in microcirculation. Defects in the cytoskeleton produce changes in erythrocyte deformability and stability, affecting cell viability and rheological properties. Such abnormalities are seen in different pathologies of special interest, such as different types of anemia, hypertension, and diabetes, among others. This review highlights the main findings in mammalian erythrocytes and their progenitors regarding the presence, conformation and function of the three main components of the cytoskeleton: actin, intermediate filaments, and tubulin.
Collapse
Affiliation(s)
- Ayelén D Nigra
- Departamento de Biología Molecular, Facultad de Ciencias Exactas Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, 5800, Río Cuarto, Córdoba, Argentina
- Departamento de Química Biológica, Facultad de Ciencias Químicas, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), UNC-CONICET, Universidad Nacional de Córdoba, 5000, Córdoba, Argentina
| | - Cesar H Casale
- Departamento de Biología Molecular, Facultad de Ciencias Exactas Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, 5800, Río Cuarto, Córdoba, Argentina
| | - Verónica S Santander
- Departamento de Biología Molecular, Facultad de Ciencias Exactas Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, 5800, Río Cuarto, Córdoba, Argentina.
| |
Collapse
|
72
|
Sibon D, Coman T, Rossignol J, Lamarque M, Kosmider O, Bayard E, Fouquet G, Rignault R, Topçu S, Bonneau P, Bernex F, Dussiot M, Deroy K, Laurent L, Callebert J, Launay JM, Georgin-Lavialle S, Courtois G, Maroteaux L, Vaillancourt C, Fontenay M, Hermine O, Côté F. Enhanced Renewal of Erythroid Progenitors in Myelodysplastic Anemia by Peripheral Serotonin. Cell Rep 2020; 26:3246-3256.e4. [PMID: 30893598 DOI: 10.1016/j.celrep.2019.02.071] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 12/30/2018] [Accepted: 02/20/2019] [Indexed: 10/27/2022] Open
Abstract
Tryptophan as the precursor of several active compounds, including kynurenine and serotonin, is critical for numerous important metabolic functions. Enhanced tryptophan metabolism toward the kynurenine pathway has been associated with myelodysplastic syndromes (MDSs), which are preleukemic clonal diseases characterized by dysplastic bone marrow and cytopenias. Here, we reveal a fundamental role for tryptophan metabolized along the serotonin pathway in normal erythropoiesis and in the physiopathology of MDS-related anemia. We identify, both in human and murine erythroid progenitors, a functional cell-autonomous serotonergic network with pro-survival and proliferative functions. In vivo studies demonstrate that pharmacological increase of serotonin levels using fluoxetine, a common antidepressant, has the potential to become an important therapeutic strategy in low-risk MDS anemia refractory to erythropoietin.
Collapse
Affiliation(s)
- David Sibon
- Institut Imagine, INSERM U1163, CNRS ERL 8254, Université Paris Descartes, Sorbonne Paris-Cité, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Tereza Coman
- Institut Imagine, INSERM U1163, CNRS ERL 8254, Université Paris Descartes, Sorbonne Paris-Cité, Laboratoire d'Excellence GR-Ex, Paris, France; Département d'Hématologie, Gustave Roussy Cancer Campus Grand Paris, 94800 Villejuif, France
| | - Julien Rossignol
- Institut Imagine, INSERM U1163, CNRS ERL 8254, Université Paris Descartes, Sorbonne Paris-Cité, Laboratoire d'Excellence GR-Ex, Paris, France; Département d'Hématologie, Gustave Roussy Cancer Campus Grand Paris, 94800 Villejuif, France
| | - Mathilde Lamarque
- Institut Imagine, INSERM U1163, CNRS ERL 8254, Université Paris Descartes, Sorbonne Paris-Cité, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Olivier Kosmider
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes, Paris, APHP, Service d'Hématologie Biologique, Hôpitaux Universitaires Paris Centre-Cochin, Paris 75014, France
| | - Elisa Bayard
- Institut Imagine, INSERM U1163, CNRS ERL 8254, Université Paris Descartes, Sorbonne Paris-Cité, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Guillemette Fouquet
- Institut Imagine, INSERM U1163, CNRS ERL 8254, Université Paris Descartes, Sorbonne Paris-Cité, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Rachel Rignault
- Institut Imagine, INSERM U1163, CNRS ERL 8254, Université Paris Descartes, Sorbonne Paris-Cité, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Selin Topçu
- Institut Imagine, INSERM U1163, CNRS ERL 8254, Université Paris Descartes, Sorbonne Paris-Cité, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Pierre Bonneau
- Institut Imagine, INSERM U1163, CNRS ERL 8254, Université Paris Descartes, Sorbonne Paris-Cité, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Florence Bernex
- Institut de Recherche en Cancérologie de Montpellier, Montpellier 34298, France; INSERM, U1194, Network of Experimental Histology, BioCampus, CNRS, UMS3426, Montpellier 34094, France
| | - Michael Dussiot
- Institut Imagine, INSERM U1163, CNRS ERL 8254, Université Paris Descartes, Sorbonne Paris-Cité, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Kathy Deroy
- INRS-Institut Armand-Frappier and Center for Interdisciplinary Research on Well-Being, Health, Society and Environment, Montreal, QC H7V 1B7, Canada
| | - Laetitia Laurent
- INRS-Institut Armand-Frappier and Center for Interdisciplinary Research on Well-Being, Health, Society and Environment, Montreal, QC H7V 1B7, Canada
| | - Jacques Callebert
- Service de Biochimie, INSERM U942, Hôpital Lariboisière, 75010 Paris, France
| | - Jean-Marie Launay
- Service de Biochimie, INSERM U942, Hôpital Lariboisière, 75010 Paris, France
| | - Sophie Georgin-Lavialle
- Département de Médecine Interne, Hôpital Tenon, Université Pierre et Marie Curie, AP-HP, 4 rue de la Chine, 75020 Paris, France
| | - Geneviève Courtois
- Institut Imagine, INSERM U1163, CNRS ERL 8254, Université Paris Descartes, Sorbonne Paris-Cité, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Luc Maroteaux
- INSERM UMR-S1270, Sorbonne Universités, Université Pierre et Marie Curie, Institut du Fer à Moulin, 75005 Paris, France
| | - Cathy Vaillancourt
- INRS-Institut Armand-Frappier and Center for Interdisciplinary Research on Well-Being, Health, Society and Environment, Montreal, QC H7V 1B7, Canada
| | - Michaela Fontenay
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes, Paris, APHP, Service d'Hématologie Biologique, Hôpitaux Universitaires Paris Centre-Cochin, Paris 75014, France
| | - Olivier Hermine
- Institut Imagine, INSERM U1163, CNRS ERL 8254, Université Paris Descartes, Sorbonne Paris-Cité, Laboratoire d'Excellence GR-Ex, Paris, France; Department of Hematology, Hôpital Necker AP-HP, 75015 Paris, France
| | - Francine Côté
- Institut Imagine, INSERM U1163, CNRS ERL 8254, Université Paris Descartes, Sorbonne Paris-Cité, Laboratoire d'Excellence GR-Ex, Paris, France.
| |
Collapse
|
73
|
Tani J, Ito Y, Tatemichi S, Yamakami M, Fukui T, Hatano Y, Kakimoto S, Kotani A, Sugimura A, Mihara K, Yamamoto R, Tanaka N, Minami K, Takahashi K, Hirato T. Physicochemical and biological evaluation of JR-131 as a biosimilar to a long-acting erythropoiesis-stimulating agent darbepoetin alfa. PLoS One 2020; 15:e0231830. [PMID: 32302352 PMCID: PMC7164597 DOI: 10.1371/journal.pone.0231830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 04/01/2020] [Indexed: 12/02/2022] Open
Abstract
Renal anemia is predominantly caused by a relative deficiency in erythropoietin (EPO). Conventional treatment for renal anemia includes the use of recombinant human EPO (rhEPO) or a long-acting erythropoiesis-activating agent named darbepoetin alfa, which is a modified rhEPO with a carbohydrate chain structure that differs from native hEPO. We have developed a biosimilar to darbepoetin alfa designated JR-131. Here, we comprehensively compare the physicochemical and biological characteristics of JR-131 to darbepoetin alfa. JR-131 demonstrated similar protein structure to the originator, darbepoetin alfa, by peptide mapping and circular dichroism spectroscopy. Additionally, mass spectroscopic analyses and capillary zone electrophoresis revealed similar glycosylation patterns between the two products. Human bone marrow-derived erythroblasts differentiated and proliferated to form colonies with JR-131 to a similar degree as darbepoetin alfa. Finally, JR-131 stimulated erythropoiesis and improved anemia in rats similarly to darbepoetin alfa. Our data show the similarity in physicochemical and biological properties of JR-131 to those of darbepoetin alfa, and JR-131 therefore represents a biosimilar for use in the treatment of renal anemia.
Collapse
Affiliation(s)
- Junya Tani
- Research Division, JCR Pharmaceuticals Co., Ltd., Kobe, Japan
| | - Yae Ito
- Research Division, JCR Pharmaceuticals Co., Ltd., Kobe, Japan
| | | | - Makoto Yamakami
- Research Division, JCR Pharmaceuticals Co., Ltd., Kobe, Japan
| | - Tsuyoshi Fukui
- Research Division, JCR Pharmaceuticals Co., Ltd., Kobe, Japan
| | - Yukichi Hatano
- Research Division, JCR Pharmaceuticals Co., Ltd., Kobe, Japan
| | - Shinji Kakimoto
- Research Planning Division, JCR Pharmaceuticals Co., Ltd., Kobe, Japan
| | - Ayaka Kotani
- Research Planning Division, JCR Pharmaceuticals Co., Ltd., Kobe, Japan
| | | | - Kazutoshi Mihara
- Research Division, JCR Pharmaceuticals Co., Ltd., Kobe, Japan
- * E-mail: (KM); (KM)
| | - Ryuji Yamamoto
- Research Division, JCR Pharmaceuticals Co., Ltd., Kobe, Japan
| | - Noboru Tanaka
- Research Division, JCR Pharmaceuticals Co., Ltd., Kobe, Japan
| | - Kohtaro Minami
- Research Division, JCR Pharmaceuticals Co., Ltd., Kobe, Japan
- * E-mail: (KM); (KM)
| | | | - Tohru Hirato
- Research Division, JCR Pharmaceuticals Co., Ltd., Kobe, Japan
| |
Collapse
|
74
|
Oztermeli A, Karaca S, Yucel I, Midi A, Sen EI, Ozturk BY. The effect of erythropoietin on rat rotator cuff repair model: An experimental study. J Orthop Surg (Hong Kong) 2020; 27:2309499019856389. [PMID: 31234725 DOI: 10.1177/2309499019856389] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES The aim of this study was to determine whether erythropoietin (EPO) can enhance rotator cuff healing in rats as measured by histological analysis and biomechanical testing. METHODS A total of 72 rats were included in this study. In the control group (n = 24), repair was performed without EPO injection. In the local group (n = 24) EPO was injected in the repair site. In the systemic group (n = 24) EPO was administered as an intraperitoneal injection every day for 10 days after repair. Rats were euthanized on day 10 (n = 12 from each group) and day 28 (n = 12 from each group). Histopathological (n = 6) and biomechanical examinations (n = 6) were done. RESULTS Biomechanical results reveal that the maximum load to failure values of the early control group were statistically lower than those of the early systemic group (p = 0.006). Comparing the the total Bonar values histopathologically reveal that the early systemic group was statistically higher than those of the early local group (p = 0.043). The late control group was statistically higher than those of the late local group (p = 0.003) and the late systemic group (p = 0.034). The late systemic group was statistically higher than those of the late local group (p = 0.003). CONCLUSIONS EPO application had a positive effect biomechanically in the early euthanized group and histopathologically in the late euthanized group.
Collapse
Affiliation(s)
- Ahmet Oztermeli
- 1 Department of Orthopaedics and Traumatology, Fatih Sultan Mehmet Training and Research Hospital, Istanbul, Turkey
| | - Sinan Karaca
- 1 Department of Orthopaedics and Traumatology, Fatih Sultan Mehmet Training and Research Hospital, Istanbul, Turkey
| | - Istemi Yucel
- 1 Department of Orthopaedics and Traumatology, Fatih Sultan Mehmet Training and Research Hospital, Istanbul, Turkey
| | - Ahmet Midi
- 2 Department of Pathology, Bahcesehir University Faculty of Medicine, Istanbul, Turkey
| | - Elif Itir Sen
- 2 Department of Pathology, Bahcesehir University Faculty of Medicine, Istanbul, Turkey
| | - Burak Yagmur Ozturk
- 1 Department of Orthopaedics and Traumatology, Fatih Sultan Mehmet Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
75
|
Held MA, Greenfest-Allen E, Jachimowicz E, Stoeckert CJ, Stokes MP, Wood AW, Wojchowski DM. Phospho-proteomic discovery of novel signal transducers including thioredoxin-interacting protein as mediators of erythropoietin-dependent human erythropoiesis. Exp Hematol 2020; 84:29-44. [PMID: 32259549 DOI: 10.1016/j.exphem.2020.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 01/19/2023]
Abstract
Erythroid cell formation critically depends on signals transduced via erythropoietin (EPO)/EPO receptor (EPOR)/JAK2 complexes. This includes not only core response modules (e.g., JAK2/STAT5, RAS/MEK/ERK), but also specialized effectors (e.g., erythroferrone, ASCT2 glutamine transport, Spi2A). By using phospho-proteomics and a human erythroblastic cell model, we identify 121 new EPO target proteins, together with their EPO-modulated domains and phosphosites. Gene ontology (GO) enrichment for "Molecular Function" identified adaptor proteins as one top EPO target category. This includes a novel EPOR/JAK2-coupled network of actin assemblage modifiers, with adaptors DLG-1, DLG-3, WAS, WASL, and CD2AP as prime components. "Cellular Component" GO analysis further identified 19 new EPO-modulated cytoskeletal targets including the erythroid cytoskeletal targets spectrin A, spectrin B, adducin 2, and glycophorin C. In each, EPO-induced phosphorylation occurred at pY sites and subdomains, which suggests coordinated regulation by EPO of the erythroid cytoskeleton. GO analysis of "Biological Processes" further revealed metabolic regulators as a likewise unexpected EPO target set. Targets included aldolase A, pyruvate dehydrogenase α1, and thioredoxin-interacting protein (TXNIP), with EPO-modulated p-Y sites in each occurring within functional subdomains. In TXNIP, EPO-induced phosphorylation occurred at novel p-T349 and p-S358 sites, and was paralleled by rapid increases in TXNIP levels. In UT7epo-E and primary human stem cell (HSC)-derived erythroid progenitor cells, lentivirus-mediated short hairpin RNA knockdown studies revealed novel pro-erythropoietic roles for TXNIP. Specifically, TXNIP's knockdown sharply inhibited c-KIT expression; compromised EPO dose-dependent erythroblast proliferation and survival; and delayed late-stage erythroblast formation. Overall, new insight is provided into EPO's diverse action mechanisms and TXNIP's contributions to EPO-dependent human erythropoiesis.
Collapse
Affiliation(s)
- Matthew A Held
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH
| | | | - Edward Jachimowicz
- Molecular Medicine Department, Maine Medical Center Research Institute, Scarborough, ME
| | | | | | | | - Don M Wojchowski
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH.
| |
Collapse
|
76
|
Dey S, Cui Z, Gavrilova O, Zhang X, Gassmann M, Noguchi CT. Sex-specific brain erythropoietin regulation of mouse metabolism and hypothalamic inflammation. JCI Insight 2020; 5:134061. [PMID: 32078583 DOI: 10.1172/jci.insight.134061] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/12/2020] [Indexed: 12/17/2022] Open
Abstract
The blood hormone erythropoietin (EPO), upon binding to its receptor (EpoR), modulates high-fat diet-induced (HFD-induced) obesity in mice, improves glucose tolerance, and prevents white adipose tissue inflammation. Transgenic mice with constitutive overexpression of human EPO solely in the brain (Tg21) were used to assess the neuroendocrine EPO effect without increasing the hematocrit. Male Tg21 mice resisted HFD-induced weight gain; showed lower serum adrenocorticotropic hormone, corticosterone, and C-reactive protein levels; and prevented myeloid cell recruitment to the hypothalamus compared with WT male mice. HFD-induced hypothalamic inflammation (HI) and microglial activation were higher in male mice, and Tg21 male mice exhibited a lower increase in HI than WT male mice. Physiological EPO function in the brain also showed sexual dimorphism in regulating HFD response. Female estrogen production blocked reduced weight gain and HI. Targeted deletion of EpoR gene expression in neuronal cells worsened HFD-induced glucose intolerance in both male and female mice but increased weight gain and HI in the hypothalamus in male mice only. Both male and female Tg21 mice kept on normal chow and HFD showed significantly improved glycemic control. Our data indicate that cerebral EPO regulates weight gain and HI in a sex-dependent response, distinct from EPO regulation of glycemic control, and independent of erythropoietic EPO response.
Collapse
Affiliation(s)
| | | | | | - Xiaojie Zhang
- Laboratory of Animal Sciences Section, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Max Gassmann
- Institute of Veterinary Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Universidad Peruana Cayetano Heredia, Lima, Peru
| | | |
Collapse
|
77
|
Erythropoietin Receptor Signaling Supports Retinal Function after Vascular Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:630-641. [PMID: 32093902 DOI: 10.1016/j.ajpath.2019.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/30/2019] [Accepted: 11/19/2019] [Indexed: 12/14/2022]
Abstract
The investigation of erythropoietin (EPO) has expanded to include potential nonhematopoietic roles in neural and retinal diseases, including diabetic retinopathy. However, it remains unclear how EPO functions to support the neural retina. Transgenic mice with hypoactive EPO receptor (EPOR) signaling (hWtEPOR) were compared with littermate control mice (WT) to test the role of EPOR signaling under normal conditions and after vascular injury and regrowth into the retina. Although retinal function tested with OptoMotry and electroretinography was comparable to adult (8-week-old) littermate WT mice, hWtEPOR mice had thinner inner and outer plexiform layers and a greater number of amacrine cells. Injury and repair caused by the oxygen-induced retinopathy model reduced visual acuity thresholds, reduced electroretinography amplitudes, and thinned the outer plexiform and inner nuclear layers of both WT and hWtEPOR 8-week-old mice. In hWtEPOR compared with WT mice, scotopic a-wave amplitudes were reduced by injury, despite no change in outer nuclear layer thickness; and peripheral rod, but not cone number, was reduced. Scotopic b-waves were reduced in injured hWtEPOR mice compared with WT, and rod bipolar cell ectopic neurites were increased in both genotypes after injury, suggesting a potential reparative process to preserve connectivity and the b-wave. Normal EPOR signaling appeared important because ectopic neurites and b-waves were lower in the hWtEPOR than WT injured mice.
Collapse
|
78
|
Held MA, Greenfest-Allen E, Su S, Stoeckert CJ, Stokes MP, Wojchowski DM. Phospho-PTM proteomic discovery of novel EPO- modulated kinases and phosphatases, including PTPN18 as a positive regulator of EPOR/JAK2 Signaling. Cell Signal 2020; 69:109554. [PMID: 32027948 DOI: 10.1016/j.cellsig.2020.109554] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 02/07/2023]
Abstract
The formation of erythroid progenitor cells depends sharply upon erythropoietin (EPO), its cell surface receptor (erythropoietin receptor, EPOR), and Janus kinase 2 (JAK2). Clinically, recombinant human EPO (rhEPO) additionally is an important anti-anemia agent for chronic kidney disease (CKD), myelodysplastic syndrome (MDS) and chemotherapy, but induces hypertension, and can exert certain pro-tumorigenic effects. Cellular signals transduced by EPOR/JAK2 complexes, and the nature of EPO-modulated signal transduction factors, therefore are of significant interest. By employing phospho-tyrosine post-translational modification (p-Y PTM) proteomics and human EPO- dependent UT7epo cells, we have identified 22 novel kinases and phosphatases as novel EPO targets, together with their specific sites of p-Y modification. New kinases modified due to EPO include membrane palmitoylated protein 1 (MPP1) and guanylate kinase 1 (GUK1) guanylate kinases, together with the cytoskeleton remodeling kinases, pseudopodium enriched atypical kinase 1 (PEAK1) and AP2 associated kinase 1 (AAK1). Novel EPO- modified phosphatases include protein tyrosine phosphatase receptor type A (PTPRA), phosphohistidine phosphatase 1 (PHPT1), tensin 2 (TENC1), ubiquitin associated and SH3 domain containing B (UBASH3B) and protein tyrosine phosphatase non-receptor type 18 (PTPN18). Based on PTPN18's high expression in hematopoietic progenitors, its novel connection to JAK kinase signaling, and a unique EPO- regulated PTPN18-pY389 motif which is modulated by JAK2 inhibitors, PTPN18's actions in UT7epo cells were investigated. Upon ectopic expression, wt-PTPN18 promoted EPO dose-dependent cell proliferation, and survival. Mechanistically, PTPN18 sustained the EPO- induced activation of not only mitogen-activated protein kinases 1 and 3 (ERK1/2), AKT serine/threonine kinase 1-3 (AKT), and signal transducer and activator of transcription 5A and 5B (STAT5), but also JAK2. Each effect further proved to depend upon PTPN18's EPO- modulated (p)Y389 site. In analyses of the EPOR and the associated adaptor protein RHEX (regulator of hemoglobinization and erythroid cell expansion), wt-PTPN18 increased high molecular weight EPOR forms, while sharply inhibiting the EPO-induced phosphorylation of RHEX-pY141. Each effect likewise depended upon PTPN18-Y389. PTPN18 thus promotes signals for EPO-dependent hematopoietic cell growth, and may represent a new druggable target for myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Matthew A Held
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH, 03824, United States of America
| | - Emily Greenfest-Allen
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, United States of America
| | - Su Su
- Molecular Medicine Department, Maine Medical Center Research Institute, Scarborough, ME, 04074, United States of America
| | - Christian J Stoeckert
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, United States of America
| | - Matthew P Stokes
- Proteomics Division, Cell Signaling Technology, Danvers, MA, 01923., United States of America
| | - Don M Wojchowski
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH, 03824, United States of America.
| |
Collapse
|
79
|
Suresh S, Rajvanshi PK, Noguchi CT. The Many Facets of Erythropoietin Physiologic and Metabolic Response. Front Physiol 2020; 10:1534. [PMID: 32038269 PMCID: PMC6984352 DOI: 10.3389/fphys.2019.01534] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/05/2019] [Indexed: 12/30/2022] Open
Abstract
In mammals, erythropoietin (EPO), produced in the kidney, is essential for bone marrow erythropoiesis, and hypoxia induction of EPO production provides for the important erythropoietic response to ischemic stress, such as during blood loss and at high altitude. Erythropoietin acts by binding to its cell surface receptor which is expressed at the highest level on erythroid progenitor cells to promote cell survival, proliferation, and differentiation in production of mature red blood cells. In addition to bone marrow erythropoiesis, EPO causes multi-tissue responses associated with erythropoietin receptor (EPOR) expression in non-erythroid cells such neural cells, endothelial cells, and skeletal muscle myoblasts. Animal and cell models of ischemic stress have been useful in elucidating the potential benefit of EPO affecting maintenance and repair of several non-hematopoietic organs including brain, heart and skeletal muscle. Metabolic and glucose homeostasis are affected by endogenous EPO and erythropoietin administration affect, in part via EPOR expression in white adipose tissue. In diet-induced obese mice, EPO is protective for white adipose tissue inflammation and gives rise to a gender specific response in weight control associated with white fat mass accumulation. Erythropoietin regulation of fat mass is masked in female mice due to estrogen production. EPOR is also expressed in bone marrow stromal cells (BMSC) and EPO administration in mice results in reduced bone independent of the increase in hematocrit. Concomitant reduction in bone marrow adipocytes and bone morphogenic protein suggests that high EPO inhibits adipogenesis and osteogenesis. These multi-tissue responses underscore the pleiotropic potential of the EPO response and may contribute to various physiological manifestations accompanying anemia or ischemic response and pharmacological uses of EPO.
Collapse
Affiliation(s)
- Sukanya Suresh
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Praveen Kumar Rajvanshi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Constance T Noguchi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
80
|
Bermudez D, Azad P, Figueroa-Mujíca R, Vizcardo-Galindo G, Corante N, Guerra-Giraldez C, Haddad GG, Villafuerte FC. Increased hypoxic proliferative response and gene expression in erythroid progenitor cells of Andean highlanders with chronic mountain sickness. Am J Physiol Regul Integr Comp Physiol 2020; 318:R49-R56. [PMID: 31617751 PMCID: PMC6985794 DOI: 10.1152/ajpregu.00250.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 12/20/2022]
Abstract
Excessive erythrocytosis (EE) is the main sign of chronic mountain sickness (CMS), a maladaptive clinical syndrome prevalent in Andean and other high-altitude populations worldwide. The pathophysiological mechanism of EE is still controversial, as physiological variability of systemic respiratory, cardiovascular, and hormonal responses to chronic hypoxemia complicates the identification of underlying causes. Induced pluripotent stem cells derived from CMS highlanders showed increased expression of genes relevant to the regulation of erythropoiesis, angiogenesis, cardiovascular, and steroid-hormone function that appear to explain the exaggerated erythropoietic response. However, the cellular response to hypoxia in native CMS cells is yet unknown. This study had three related aims: to determine the hypoxic proliferation of native erythroid progenitor burst-forming unit-erythroid (BFU-E) cells derived from CMS and non-CMS peripheral blood mononuclear cells; to examine their sentrin-specific protease 1 (SENP1), GATA-binding factor 1 (GATA1), erythropoietin (EPO), and EPO receptor (EPOR) expression; and to investigate the functional upstream role of SENP1 in native progenitor differentiation into erythroid precursors. Native CMS BFU-E colonies showed increased proliferation under hypoxic conditions compared with non-CMS cells, together with an upregulated expression of SENP1, GATA1, EPOR; and no difference in EPO expression. Knock-down of the SENP1 gene abolished the augmented proliferative response. Thus, we demonstrate that native CMS progenitor cells produce a larger proportion of erythroid precursors under hypoxia and that SENP1 is essential for proliferation. Our findings suggest a significant intrinsic component for developing EE in CMS highlanders at the cellular and gene expression level that could be further enhanced by systemic factors such as alterations in respiratory control, or differential hormonal patterns.
Collapse
Affiliation(s)
- Daniela Bermudez
- Laboratorio de Fisiología Comparada, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Priti Azad
- Division of Respiratory Medicine, Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Rómulo Figueroa-Mujíca
- Laboratorio de Fisiología Comparada, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Gustavo Vizcardo-Galindo
- Laboratorio de Fisiología Comparada, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Noemí Corante
- Laboratorio de Fisiología Comparada, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Cristina Guerra-Giraldez
- Laboratorio de Inflamación Cerebral, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Gabriel G Haddad
- Division of Respiratory Medicine, Department of Pediatrics, University of California, San Diego, La Jolla, California
- Department of Neurosciences, University of California San Diego, La Jolla, California
- Rady Children's Hospital, San Diego, La Jolla, California
| | - Francisco C Villafuerte
- Laboratorio de Fisiología Comparada, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
- Instituto de Investigaciones de la Altura (IIA), Universidad Peruana Cayetano Heredia, Lima, Peru
| |
Collapse
|
81
|
Diaz-Canestro C, Haider T, Lundby C, Montero D. Relationship between plasma volume and essential blood constituents in patients with heart failure and preserved ejection fraction. Clin Physiol Funct Imaging 2019; 40:131-138. [PMID: 31823430 DOI: 10.1111/cpf.12614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 12/04/2019] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Notwithstanding recent progress on molecular mechanisms underlying heart failure with preserved ejection fraction (HFpEF), multiple pathophysiological aspects of this condition including the basis of anaemia and other haematological disorders remain unresolved. In this study, we sought to determine the relationship of plasma volume (PV), a plausible confounding factor for the concentration of solutes in blood, with key haematological markers in HFpEF patients. METHODS Total circulating PV was determined with high precision, automated carbon monoxide rebreathing in 24 stable HFpEF patients (70 ± 8 years, left ventricular ejection fraction = 55±5%) and 18 healthy age- and sex-matched control (HC) individuals. Linear regression analyses were performed to determine the association of PV with a comprehensive set of haematological variables. RESULTS Haematocrit (40·1 ± 4·9 versus 43·6 ± 2·7%, P = 0·004) and haemoglobin concentration (131 ± 16 versus 142 ± 7 g l-1 , P = 0·003) were reduced in HFpEF patients compared with HC individuals. In regression analyses, PV was negatively associated with haematocrit (r = -0·45, P = 0·029) and haemoglobin concentration (r = -0·44, P = 0·030) in HFpEF patients, whereas these variables were not associated with PV in HC individuals (P≥0·198). Regarding blood electrolytes, PV was negatively associated with K+ (r = -0·43, P = 0·036) and Ca2+ (r = -0·44, P = 0·032) in HFpEF patients but not in HC individuals (P≥0·734). None of the above associations were detected in HFpEF patients when using ideal instead of measured PV. CONCLUSION The blood concentration of routine markers of anaemia and electrolyte balance is specifically and linearly associated with PV in HFpEF patients. Excess or deficit of circulating PV may confound clinical diagnosis in this population.
Collapse
Affiliation(s)
- Candela Diaz-Canestro
- Faculty of Kinesiology, Cumming School of Medicine, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - Thomas Haider
- University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Carsten Lundby
- Inland Norway University of Applied Sciences, Lillehammer, Norway
| | - David Montero
- Faculty of Kinesiology, Cumming School of Medicine, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
82
|
Federici G, Varricchio L, Martelli F, Falchi M, Picconi O, Francescangeli F, Contavalli P, Girelli G, Tafuri A, Petricoin EF, Mazzarini M, Zeuner A, Migliaccio AR. Phosphoproteomic Landscaping Identifies Non-canonical cKIT Signaling in Polycythemia Vera Erythroid Progenitors. Front Oncol 2019; 9:1245. [PMID: 31824842 PMCID: PMC6883719 DOI: 10.3389/fonc.2019.01245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/29/2019] [Indexed: 01/08/2023] Open
Abstract
Although stem cell factor (SCF)/cKIT interaction plays key functions in erythropoiesis, cKIT signaling in human erythroid cells is still poorly defined. To provide new insights into cKIT-mediated erythroid expansion in development and disease, we performed phosphoproteomic profiling of primary erythroid progenitors from adult blood (AB), cord blood (CB), and Polycythemia Vera (PV) at steady-state and upon SCF stimulation. While AB and CB, respectively, activated transient or sustained canonical cKIT-signaling, PV showed a non-canonical signaling including increased mTOR and ERK1 and decreased DEPTOR. Accordingly, screening of FDA-approved compounds showed increased PV sensitivity to JAK, cKIT, and MEK inhibitors. Moreover, differently from AB and CB, in PV the mature 145kDa-cKIT constitutively associated with the tetraspanin CD63 and was not endocytosed upon SCF stimulation, contributing to unrestrained cKIT signaling. These results identify a clinically exploitable variegation of cKIT signaling/metabolism that may contribute to the great erythroid output occurring during development and in PV.
Collapse
Affiliation(s)
| | - Lilian Varricchio
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Fabrizio Martelli
- National Center for Preclinical and Clinical Research and Evaluation of Pharmaceutical Drugs, Istituto Superiore di Sanità, Rome, Italy
| | - Mario Falchi
- National HIV/AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | - Orietta Picconi
- National HIV/AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | | | - Paola Contavalli
- Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gabriella Girelli
- Immunohematology and Transfusion Medicine Unit, "La Sapienza" University of Rome, Rome, Italy
| | - Agostino Tafuri
- Sant'Andrea Hospital-La Sapienza, Department of Clinic and Molecular Medicine "La Sapienza" University of Rome, Rome, Italy
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, United States
| | - Maria Mazzarini
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, Bologna, Italy
| | - Ann Zeuner
- Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Anna Rita Migliaccio
- Myeloproliferative Neoplasm Research Consortium, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
83
|
Malik N, Dunn KM, Cassels J, Hay J, Estell C, Sansom OJ, Michie AM. mTORC1 activity is essential for erythropoiesis and B cell lineage commitment. Sci Rep 2019; 9:16917. [PMID: 31729420 PMCID: PMC6858379 DOI: 10.1038/s41598-019-53141-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/26/2019] [Indexed: 12/17/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a serine/threonine protein kinase that mediates phosphoinositide-3-kinase (PI3K)/AKT signalling. This pathway is involved in a plethora of cellular functions including protein and lipid synthesis, cell migration, cell proliferation and apoptosis. In this study, we proposed to delineate the role of mTORC1 in haemopoietic lineage commitment using knock out (KO) mouse and cell line models. Mx1-cre and Vav-cre expression systems were used to specifically target Raptorfl/fl (mTORC1), either in all tissues upon poly(I:C) inoculation, or specifically in haemopoietic stem cells, respectively. Assessment of the role of mTORC1 during the early stages of development in Vav-cre+Raptorfl/fl mice, revealed that these mice do not survive post birth due to aberrations in erythropoiesis resulting from an arrest in development at the megakaryocyte-erythrocyte progenitor stage. Furthermore, Raptor-deficient mice exhibited a block in B cell lineage commitment. The essential role of Raptor (mTORC1) in erythrocyte and B lineage commitment was confirmed in adult Mx1-cre+Raptorfl/fl mice upon cre-recombinase induction. These studies were supported by results showing that the expression of key lineage commitment regulators, GATA1, GATA2 and PAX5 were dysregulated in the absence of mTORC1-mediated signals. The regulatory role of mTOR during erythropoiesis was confirmed in vitro by demonstrating a reduction of K562 cell differentiation towards RBCs in the presence of established mTOR inhibitors. While mTORC1 plays a fundamental role in promoting RBC development, we showed that mTORC2 has an opposing role, as Rictor-deficient progenitor cells exhibited an elevation in RBC colony formation ex vivo. Collectively, our data demonstrate a critical role played by mTORC1 in regulating the haemopoietic cell lineage commitment.
Collapse
Affiliation(s)
- Natasha Malik
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Karen M Dunn
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jennifer Cassels
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jodie Hay
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Christopher Estell
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Owen J Sansom
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, UK
| | - Alison M Michie
- Institute of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
84
|
Sato K, Kumagai N, Suzuki N. Alteration of the DNA Methylation Signature of Renal Erythropoietin-Producing Cells Governs the Sensitivity to Drugs Targeting the Hypoxia-Response Pathway in Kidney Disease Progression. Front Genet 2019; 10:1134. [PMID: 31798631 PMCID: PMC6863978 DOI: 10.3389/fgene.2019.01134] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/18/2019] [Indexed: 12/14/2022] Open
Abstract
Chronic kidney disease (CKD) affects more than 10% of the population worldwide and burdens citizens with heavy medical expenses in many countries. Because a vital erythroid growth factor, erythropoietin (EPO), is secreted from renal interstitial fibroblasts [renal EPO-producing (REP) cells], anemia arises as a major complication of CKD. We determined that hypoxia-inducible factor 2α (HIF2α), which is inactivated by HIF-prolyl hydroxylase domain-containing proteins (PHDs) in an oxygen-dependent manner, tightly regulates EPO production in REP cells at the gene transcription level to maintain oxygen homeostasis. HIF2α-mediated disassembly of the nucleosome in the EPO gene is also involved in hypoxia-inducible EPO production. In renal anemia patients, anemic and pathological hypoxia is ineffective toward EPO induction due to the inappropriate over-activation of PHDs in REP cells transformed into myofibroblasts (MF-REP cells) due to kidney damage. Accordingly, PHD inhibitory compounds are being developed for the treatment of renal anemia. However, our studies have demonstrated that the promoter regions of the genes encoding EPO and HIF2α are highly methylated in MF-REP cells, and the expression of these genes is epigenetically silenced with CKD progression. This finding notably indicates that the efficacy of PHD inhibitors depends on the CKD stage of each patient. In addition, a strategy for harvesting renal cells, including REP cells from the urine of patients, is proposed to identify plausible biomarkers for CKD and to develop personalized precision medicine against CKD by a non-invasive strategy.
Collapse
Affiliation(s)
- Koji Sato
- Division of Oxygen Biology, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.,Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naonori Kumagai
- Department of Pediatrics, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Norio Suzuki
- Division of Oxygen Biology, United Centers for Advanced Research and Translational Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
85
|
Sciesielski LK, Kirschner KM. ExActa HIF prolyl hydroxylase inhibitors-The new lifestyle drug? Acta Physiol (Oxf) 2019; 227:e13370. [PMID: 31465609 DOI: 10.1111/apha.13370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/23/2019] [Accepted: 08/23/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Lina K. Sciesielski
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität BerlinHumboldt‐Universität zu Berlin, and Berlin Institute of HealthDepartment of Neonatology Berlin Germany
| | - Karin M. Kirschner
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität BerlinHumboldt‐Universität zu Berlin, and Berlin Institute of HealthInstitute of Vegetative Physiology Berlin Germany
| |
Collapse
|
86
|
Zhang J, Liu Y, Han X, Mei Y, Yang J, Zhang ZJ, Lu X, Ji P. Rats provide a superior model of human stress erythropoiesis. Exp Hematol 2019; 78:21-34.e3. [PMID: 31562902 DOI: 10.1016/j.exphem.2019.09.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/28/2019] [Accepted: 09/16/2019] [Indexed: 11/15/2022]
Abstract
Mouse models are widely used to study human erythropoiesis in vivo. One important caveat using mouse models is that mice often develop significant extramedullary erythropoiesis with anemia, which could mask important phenotypes. To overcome this drawback in mice, here we established in vitro and in vivo rat models for the studies of stress erythropoiesis. Using flow cytometry-based assays, we can monitor terminal erythropoiesis in rats during fetal and adult erythropoiesis under steady state and stress conditions. We used this system to test rat erythropoiesis under phenylhydrazine (PHZ)-induced hemolytic stress. In contrast to mice, rats did not have an increased proportion of early-stage erythroid precursors during terminal differentiation in the spleen or bone marrow. This could be explained by the abundant bone marrow spaces in rats that allow sufficient erythroid proliferation under stress. Consistently, the extent of splenomegaly in rats after PHZ treatment was significantly lower than that in mice. The level of BMP4, which was significantly increased in mouse spleen after PHZ treatment, remained unchanged in rat spleen. We further demonstrated that the bone marrow c-Kit positive progenitor population underwent a phenotype shift and became more CD71 positive and erythroid skewed with the expression of maturing erythroid markers under stress in rats and humans. In contrast, the phenotype shift to an erythroid-skewed progenitor population in mice occurred mainly in the spleen. Our study establishes rat in vitro and in vivo erythropoiesis models that are more appropriate and superior for the study of human stress erythropoiesis than mouse models.
Collapse
Affiliation(s)
- Jingxin Zhang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL; School of Life Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Yijie Liu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Xu Han
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Yang Mei
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Jing Yang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Zheng J Zhang
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Xinyan Lu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Peng Ji
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL.
| |
Collapse
|
87
|
She J, Wu Y, Lou B, Lodd E, Klems A, Schmoehl F, Yuan Z, Noble FL, Kroll J. Genetic compensation by epob in pronephros development in epoa mutant zebrafish. Cell Cycle 2019; 18:2683-2696. [PMID: 31451030 DOI: 10.1080/15384101.2019.1656019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Zebrafish erythropoietin a (epoa) is a well characterized regulator of red blood cell formation. Recent morpholino mediated knockdown data have also identified epoa being essential for physiological pronephros development in zebrafish, which is driven by blocking apoptosis in developing kidneys. Yet, zebrafish mutants for epoa have not been described so far. In order to compare a transient knockdown vs. permanent knockout for epoa in zebrafish on pronephros development, we used CRISPR/Cas9 technology to generate epoa knockout zebrafish mutants and we performed structural and functional studies on pronephros development. In contrast to epoa morphants, epoa-/- zebrafish mutants showed normal pronephros structure; however, a previously uncharacterized gene in zebrafish, named epob, was identified and upregulated in epoa-/- mutants. epob knockdown altered pronephros development, which was further aggravated in epoa-/- mutants. Likewise, epoa and epob morphants regulated similar and differential gene signatures related to kidney development in zebrafish. In conclusion, stable loss of epoa during embryonic development can be compensated by epob leading to phenotypical discrepancies in epoa knockdown and knockout zebrafish embryos.
Collapse
Affiliation(s)
- Jianqing She
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University , Xi'an , People's Republic of China.,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University , Mannheim , Germany
| | - Yue Wu
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Bowen Lou
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University , Xi'an , People's Republic of China.,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University , Mannheim , Germany
| | - Elisabeth Lodd
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University , Mannheim , Germany
| | - Alina Klems
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO) & Institute of Toxicology and Genetics (ITG), Karlsruhe Institute of Technology (KIT) , Karlsruhe , Germany
| | - Felix Schmoehl
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University , Mannheim , Germany
| | - Zuyi Yuan
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University , Xi'an , People's Republic of China
| | - Ferdinand Le Noble
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO) & Institute of Toxicology and Genetics (ITG), Karlsruhe Institute of Technology (KIT) , Karlsruhe , Germany
| | - Jens Kroll
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University , Mannheim , Germany
| |
Collapse
|
88
|
Bao EL, Cheng AN, Sankaran VG. The genetics of human hematopoiesis and its disruption in disease. EMBO Mol Med 2019; 11:e10316. [PMID: 31313878 PMCID: PMC6685084 DOI: 10.15252/emmm.201910316] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/25/2022] Open
Abstract
Hematopoiesis, or the process of blood cell production, is a paradigm of multi-lineage cellular differentiation that has been extensively studied, yet in many aspects remains incompletely understood. Nearly all clinically measured hematopoietic traits exhibit extensive variation and are highly heritable, underscoring the importance of genetic variation in these processes. This review explores how human genetics have illuminated our understanding of hematopoiesis in health and disease. The study of rare mutations in blood and immune disorders has elucidated novel roles for regulators of hematopoiesis and uncovered numerous important molecular pathways, as seen through examples such as Diamond-Blackfan anemia and the GATA2 deficiency syndromes. Additionally, population studies of common genetic variation have revealed mechanisms by which human hematopoiesis can be modulated. We discuss advances in functionally characterizing common variants associated with blood cell traits and discuss therapeutic insights, such as the discovery of BCL11A as a modulator of fetal hemoglobin expression. Finally, as genetic techniques continue to evolve, we discuss the prospects, challenges, and unanswered questions that lie ahead in this burgeoning field.
Collapse
Affiliation(s)
- Erik L Bao
- Division of Hematology/OncologyBoston Children's HospitalHarvard Medical SchoolBostonMAUSA
- Department of Pediatric OncologyDana‐Farber Cancer InstituteHarvard Medical SchoolBostonMAUSA
- Broad Institute of MIT and HarvardCambridgeMAUSA
- Harvard‐MIT Health Sciences and TechnologyHarvard Medical SchoolBostonMAUSA
| | - Aaron N Cheng
- Division of Hematology/OncologyBoston Children's HospitalHarvard Medical SchoolBostonMAUSA
- Department of Pediatric OncologyDana‐Farber Cancer InstituteHarvard Medical SchoolBostonMAUSA
- Broad Institute of MIT and HarvardCambridgeMAUSA
| | - Vijay G Sankaran
- Division of Hematology/OncologyBoston Children's HospitalHarvard Medical SchoolBostonMAUSA
- Department of Pediatric OncologyDana‐Farber Cancer InstituteHarvard Medical SchoolBostonMAUSA
- Broad Institute of MIT and HarvardCambridgeMAUSA
- Harvard Stem Cell InstituteCambridgeMAUSA
| |
Collapse
|
89
|
Erythroblast island macrophages: recent discovery and future perspectives. BLOOD SCIENCE 2019; 1:61-64. [PMID: 35402789 PMCID: PMC8974950 DOI: 10.1097/bs9.0000000000000017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 11/26/2022] Open
Abstract
Erythroblastic island (EBI), composed of a central macrophage surrounded by developing erythroid cells, is a structure found in hematopoietic tissues such as fetal liver and bone marrow. It is the first described hematopoietic niche that predominantly supports erythropoiesis. Although it is well accepted that EBIs and EBI macrophage play important roles during erythropoiesis, the mechanisms by which they support erythropoiesis remain largely unclear due to our inability to identify and isolate EBI macrophages. Earlier efforts to identify surface markers for EBI macrophages have focused on the adhesion molecules which are involved in macrophage's interaction with erythroblasts. These include EMP, Vcam1, CD169, CD163, and αV integrin. Findings from these earlier studies suggested that combination of Vcam1, CD169, and mouse macrophage surface marker F4/80 can be used to define mouse EBI macrophage. We found that not all F4/80+Vcam1+CD169+ macrophages are EBI macrophages. Instead, we discovered that EBI macrophages are characterized by the expression of Epor in both mouse and man. RNA-seq analyses of the newly identified EBI macrophages revealed that EBI macrophages have involved specialized function in supporting erythropoiesis. Our findings provide foundation for future studies. Here we will review current knowledge of EBI macrophages and discuss future perspectives.
Collapse
|
90
|
Suresh S, de Castro LF, Dey S, Robey PG, Noguchi CT. Erythropoietin modulates bone marrow stromal cell differentiation. Bone Res 2019; 7:21. [PMID: 31666996 PMCID: PMC6804931 DOI: 10.1038/s41413-019-0060-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 04/01/2019] [Accepted: 05/17/2019] [Indexed: 01/08/2023] Open
Abstract
Erythropoietin is essential for bone marrow erythropoiesis and erythropoietin receptor on non-erythroid cells including bone marrow stromal cells suggests systemic effects of erythropoietin. Tg6 mice with chronic erythropoietin overexpression have a high hematocrit, reduced trabecular and cortical bone and bone marrow adipocytes, and decreased bone morphogenic protein 2 driven ectopic bone and adipocyte formation. Erythropoietin treatment (1 200 IU·kg–1) for 10 days similarly exhibit increased hematocrit, reduced bone and bone marrow adipocytes without increased osteoclasts, and reduced bone morphogenic protein signaling in the bone marrow. Interestingly, endogenous erythropoietin is required for normal differentiation of bone marrow stromal cells to osteoblasts and bone marrow adipocytes. ΔEpoRE mice with erythroid restricted erythropoietin receptor exhibit reduced trabecular bone, increased bone marrow adipocytes, and decreased bone morphogenic protein 2 ectopic bone formation. Erythropoietin treated ΔEpoRE mice achieved hematocrit similar to wild-type mice without reduced bone, suggesting that bone reduction with erythropoietin treatment is associated with non-erythropoietic erythropoietin response. Bone marrow stromal cells from wild-type, Tg6, and ΔEpoRE-mice were transplanted into immunodeficient mice to assess development into a bone/marrow organ. Like endogenous bone formation, Tg6 bone marrow cells exhibited reduced differentiation to bone and adipocytes indicating that high erythropoietin inhibits osteogenesis and adipogenesis, while ΔEpoRE bone marrow cells formed ectopic bones with reduced trabecular regions and increased adipocytes, indicating that loss of erythropoietin signaling favors adipogenesis at the expense of osteogenesis. In summary, endogenous erythropoietin signaling regulates bone marrow stromal cell fate and aberrant erythropoietin levels result in their impaired differentiation.
Collapse
Affiliation(s)
- Sukanya Suresh
- 1Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Luis Fernandez de Castro
- 2Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892 USA
| | - Soumyadeep Dey
- 1Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Pamela G Robey
- 2Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892 USA
| | - Constance Tom Noguchi
- 1Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| |
Collapse
|
91
|
Comparison of the Pharmacokinetic-Pharmacodynamic Relationships of Two Darbepoetin Alfa Formulations in Healthy Male Volunteers. BioDrugs 2019; 33:101-112. [PMID: 30506495 PMCID: PMC6373390 DOI: 10.1007/s40259-018-0323-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective This study compared the pharmacokinetic (PK), pharmacodynamic (PD), and safety properties of the test (CJ-40001) and reference (NESP®) versions of darbepoetin alfa following a single subcutaneous (SC) or intravenous (IV) administration in healthy male subjects. Methods A single-blind, randomized, single-dose, two-period, two-intervention crossover study was conducted, with two separate parts consisting of SC or IV administration. In each period, either a test or reference product was administered via the SC or IV route. Serial blood samples for PK analysis and the reticulocyte, hematocrit, hemoglobin, and red blood cell counts for PD analysis were collected for up to 360 or 264 h after SC or IV administration, respectively. The PK and PD parameters were calculated using non-compartmental methods. The 90% confidence intervals of the geometric mean ratios for the PK and PD parameters between the two interventions were estimated. Safety and anti-drug antibody profile assessments were performed. Results The mean darbepoetin alfa concentration–time profiles were comparable between the two products for SC and IV administration. Additionally, the PD and safety profiles were similar between the two products. Anti-drug antibody reactivity was negative for all samples from both intervention groups for SC and IV administration. The time-matched serum darbepoetin alfa concentration and the PD markers presented a counter-clockwise hysteresis, which suggests a time delay between the exposure and response. Conclusion The test and reference darbepoetin alfa formulations had similar PK, PD, and safety profiles. Thus, it is expected that the two formulations are able to be used interchangeably in clinical settings. ClinicalTrials.gov Identifier: NCT03542916.
Collapse
|
92
|
Hirano I, Suzuki N. The Neural Crest as the First Production Site of the Erythroid Growth Factor Erythropoietin. Front Cell Dev Biol 2019; 7:105. [PMID: 31245372 PMCID: PMC6581680 DOI: 10.3389/fcell.2019.00105] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/28/2019] [Indexed: 12/25/2022] Open
Abstract
While the neural crest is considered the fourth germ layer that originates a variety of tissues during mammalian development, we recently discovered that some neural crest cells and neuroepithelial cells play a unique role in secreting a vital hematopoietic hormone, erythropoietin (EPO), in mouse embryos. EPO production by the neural crest is transient in mid-stage embryos but essential for the first erythropoiesis in the yolk sac and for sufficient oxygen supply in the whole embryo growing in utero. The site of EPO production shifts from the neural crest to the liver in late embryonic stages, followed by interstitial fibroblasts of the kidneys in adults. Interestingly, the transition of EPO production sites synchronizes with the transition of erythropoietic sites during mouse development from the yolk sac and the fetal liver to the bone marrow. EPO produced by the neural crest and the neuroepithelium is first stored around the production sites and delivered to the yolk sac as an endocrine hormone for erythropoiesis after heartbeat activation. The fact that EPO is produced by some human cell lines derived from neuroblastoma, which mainly originates from the neural crest, provides evidence that the neural crest secretes EPO for primitive erythropoiesis not only in mouse but also in human embryos. Here, we introduce and discuss recent progress in studies on the mechanism of EPO production by the neural crest and its roles in erythropoietic development and in the fate of EPO-producing neural crest cells.
Collapse
Affiliation(s)
- Ikuo Hirano
- Department of Molecular Hematology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Norio Suzuki
- Division of Oxygen Biology, United Centers for Advanced Research and Translational Medicine, Graduate School of Medicine, Tohoku University, Sendai, Japan
| |
Collapse
|
93
|
Mayers S, Moço PD, Maqbool T, Silva PN, Kilkenny DM, Audet J. Establishment of an erythroid progenitor cell line capable of enucleation achieved with an inducible c-Myc vector. BMC Biotechnol 2019; 19:21. [PMID: 30987611 PMCID: PMC6466758 DOI: 10.1186/s12896-019-0515-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 04/05/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND A robust scalable method for producing enucleated red blood cells (RBCs) is not only a process to produce packed RBC units for transfusion but a potential platform to produce modified RBCs with applications in advanced cellular therapy. Current strategies for producing RBCs have shortcomings in the limited self-renewal capacity of progenitor cells, or difficulties in effectively enucleating erythroid cell lines. We explored a new method to produce RBCs by inducibly expressing c-Myc in primary erythroid progenitor cells and evaluated the proliferative and maturation potential of these modified cells. RESULTS Primary erythroid progenitor cells were genetically modified with an inducible gene transfer vector expressing a single transcription factor, c-Myc, and all the gene elements required to achieve dox-inducible expression. Genetically modified cells had enhanced proliferative potential compared to control cells, resulting in exponential growth for at least 6 weeks. Inducibly proliferating erythroid (IPE) cells were isolated with surface receptors similar to colony forming unit-erythroid (CFU-Es), and after removal of ectopic c-Myc expression cells hemoglobinized, decreased in cell size to that of native RBCs, and enucleated achieving cultures with 17% enucleated cells. Experiments with IPE cells at various levels of ectopic c-Myc expression provided insight into differentiation dynamics of the modified cells, and an optimized two-stage differentiation strategy was shown to promote greater expansion and maturation. CONCLUSIONS Genetic engineering of adult erythroid progenitor cells with an inducible c-Myc vector established an erythroid progenitor cell line that could produce RBCs, demonstrating the potential of this approach to produce large quantities of RBCs and modified RBC products.
Collapse
Affiliation(s)
- Steven Mayers
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto, Toronto, Canada
| | - Pablo Diego Moço
- Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto, Toronto, Canada
| | - Talha Maqbool
- Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto, Toronto, Canada
| | - Pamuditha N Silva
- Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto, Toronto, Canada
| | - Dawn M Kilkenny
- Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto, Toronto, Canada
| | - Julie Audet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada. .,Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto, Toronto, Canada.
| |
Collapse
|
94
|
Kunze R, Marti HH. Angioneurins - Key regulators of blood-brain barrier integrity during hypoxic and ischemic brain injury. Prog Neurobiol 2019; 178:101611. [PMID: 30970273 DOI: 10.1016/j.pneurobio.2019.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022]
Abstract
The loss of blood-brain barrier (BBB) integrity leading to vasogenic edema and brain swelling is a common feature of hypoxic/ischemic brain diseases such as stroke, but is also central to the etiology of other CNS disorders. In the past decades, numerous proteins, belonging to the family of angioneurins, have gained increasing attention as potential therapeutic targets for ischemic stroke, but also other CNS diseases attributed to BBB dysfunction. Angioneurins encompass mediators that affect both neuronal and vascular function. Recently, increasing evidence has been accumulated that certain angioneurins critically determine disease progression and outcome in stroke among others through multifaceted effects on the compromised BBB. Here, we will give a concise overview about the family of angioneurins. We further describe the most important cellular and molecular components that contribute to structural integrity and low permeability of the BBB under steady-state conditions. We then discuss BBB alterations in ischemic stroke, and highlight underlying cellular and molecular mechanisms. For the most prominent angioneurin family members including vascular endothelial growth factors, angiopoietins, platelet-derived growth factors and erythropoietin, we will summarize current scientific literature from experimental studies in animal models, and if available from clinical trials, on the following points: (i) spatiotemporal expression of these factors in the healthy and hypoxic/ischemic CNS, (ii) impact of loss- or gain-of-function during cerebral hypoxia/ischemia for BBB integrity and beyond, and (iii) potential underlying molecular mechanisms. Moreover, we will highlight novel therapeutic strategies based on the activation of endogenous angioneurins that might improve BBB dysfuntion during ischemic stroke.
Collapse
Affiliation(s)
- Reiner Kunze
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany.
| | - Hugo H Marti
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany
| |
Collapse
|
95
|
Yang F, Huang Y, Chen X, Liu L, Liao D, Zhang H, Huang G, Liu W, Zhu X, Wang W, Lobo CA, Yazdanbakhsh K, An X, Ju Z. Deletion of a flippase subunit Tmem30a in hematopoietic cells impairs mouse fetal liver erythropoiesis. Haematologica 2019; 104:1984-1994. [PMID: 30819915 PMCID: PMC6886424 DOI: 10.3324/haematol.2018.203992] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 02/27/2019] [Indexed: 01/01/2023] Open
Abstract
Transmembrane protein 30A (Tmem30a) is the β-subunit of P4-ATPases which function as flippase that transports aminophospholipids such as phosphatidylserine from the outer to the inner leaflets of the plasma membrane to maintain asymmetric distribution of phospholipids. It has been documented that deficiency of Tmem30a led to exposure of phosphatidylserine. However, the role of Tmem30a in vivo remains largely unknown. Here we found that Vav-Cre-driven conditional deletion of Tmem30a in hematopoietic cells led to embryonic lethality due to severe anemia by embryonic day 16.5. The numbers of erythroid colonies and erythroid cells were decreased in the Tmem30a deficient fetal liver. This was accompanied by increased apoptosis of erythroid cells. Confocal microscopy analysis revealed an increase of localization of erythropoietin receptor to areas of membrane raft microdomains in response to erythropoietin stimulation in Ter119−erythroid progenitors, which was impaired in Tmem30a deficient cells. Moreover, erythropoietin receptor (EPOR)-mediated activation of the STAT5 pathway was significantly reduced in Tmem30a deficient fetal liver cells. Consistently, knockdown of TMEM30A in human CD34+ cells also impaired erythropoiesis. Our findings demonstrate that Tmem30a plays a critical role in erythropoiesis by regulating the EPOR signaling pathway through the formation of membrane rafts in erythroid cells.
Collapse
Affiliation(s)
- Fan Yang
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Yumin Huang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xianda Chen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Lu Liu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Dandan Liao
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Huan Zhang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY, USA.,School of Life Science, Zhengzhou University, Zhengzhou, China
| | - Gang Huang
- Division of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Wenjing Liu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China and Chengdu, Sichuan, China
| | - Xianjun Zhu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China and Chengdu, Sichuan, China.,Chengdu Institute of Biology, Sichuan Translational Medicine Research Hospital, Chinese Academy of Sciences, Chengdu, China
| | - Wengong Wang
- Department of Biochemistry and Molecular Biology, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Cheryl A Lobo
- Laboratory of Blood-Borne Parasites, New York Blood Center, New York, NY, USA
| | | | - Xiuli An
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China .,Laboratory of Membrane Biology, New York Blood Center, New York, NY, USA.,School of Life Science, Zhengzhou University, Zhengzhou, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China .,Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
96
|
Dong X, Han Y, Abeysekera IR, Shao Z, Wang H. GDF11 is increased in patients with aplastic anemia. ACTA ACUST UNITED AC 2019; 24:331-336. [PMID: 30727851 DOI: 10.1080/16078454.2019.1574386] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECT To explore the critical role of growth differentiation factor 11 (GDF11) in the pathobiology of aplastic anemia (AA). METHODS We have examined the serum GDF11 levels for 79 AA patients and 30 healthy controls. A total of 79 AA patients, which included 29 new diagnosed (untreated) cases, 14 cases with no response, 21 partial remission (PR) cases and 15 complete remission (CR) cases after immunosuppressive therapy (IST). GDF11 serum levels were assessed by an enzyme-linked immunosorbent assay. GDF11 mRNA expression in peripheral blood mononuclear (PBMNC) was detected through real time polymerase chain reaction. The correlation between GDF11 expression and erythropoietic function was evaluated. RESULTS The serum GDF11 levels in untreated AA patients were higher than that of the control group. The serum GDF11 levels of PR patients or CR patients after IST was decreased, compared with untreated patients, but did not recover back to the normal levels. GDF11 levels had a negative correlation with hemoglobin (Hb) levels and reticulocyte counts in AA patients. GDF11 levels did not correlate with age, sex and severity of in AA patients. CONCLUSION Serum GDF11 levels were increased and negatively correlated with Hb levels and reticulocyte counts in AA patients. This suggests an impaired GDF11 response contributing to anemia in AA patients.
Collapse
Affiliation(s)
- Xifeng Dong
- a Department of Hematology, General Hospital , Tianjin Medical University , Tianjin , People's Republic of China
| | - Yu Han
- b Department of Hematology , Huaihe Hospital of Henan University , Kaifeng , People's Republic of China
| | - Iruni Roshanie Abeysekera
- c Department of Physiology and Pathophysiology, School of Basic Medical Sciences , Tianjin Medical University , Tianjin , People's Republic of China
| | - Zonghong Shao
- a Department of Hematology, General Hospital , Tianjin Medical University , Tianjin , People's Republic of China
| | - Huaquan Wang
- a Department of Hematology, General Hospital , Tianjin Medical University , Tianjin , People's Republic of China
| |
Collapse
|
97
|
Abstract
The regulation of erythropoiesis in the bone marrow microenvironment is a carefully orchestrated process that is dependent upon both systemic and local cues. Systemic erythropoietin (EPO) production by renal interstitial cells plays a critical role in maintaining erythropoietic homeostasis. In addition, there is increasing clinical and preclinical data linking changes in EPO and erythropoiesis to altered skeletal homeostasis, suggesting a functional relationship between the regulation of erythropoiesis and bone homeostasis. As key local components of the bone marrow microenvironment and erythropoietic niche, macrophage subsets play important roles in both processes. In this review, we summarize our current understanding of the cellular and molecular mechanisms that may facilitate the coordinated regulation of erythropoiesis and bone homeostasis.
Collapse
Affiliation(s)
- Joshua T Eggold
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA; Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Erinn B Rankin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA; Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA; Department of Obstetrics & Gynecologic Oncology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
98
|
Abstract
PURPOSE OF REVIEW Historically, the identity of O2-sensing renal erythropoietin (Epo)-producing (REP) cells was a matter of debate. This review summarizes how recent breakthroughs in transgenic mouse and in-situ hybridization techniques have facilitated sensitive and specific detection of REP cells and accelerated advancements in the understanding of the regulation of renal Epo production in health and disease. RECENT FINDINGS REP cells are a dynamically regulated unique subpopulation of tubulointerstitial cells with features of fibroblasts, pericytes and neurons. Under normal conditions, REP cells are located in the corticomedullary border region within a steep decrement in O2 availability. During the progression of chronic kidney disease (CKD), REP cells cease Epo production, dedifferentiate and contribute to the progression of renal fibrosis. However, CKD patients with renal anaemia still respond with elevated Epo production following treatment with hypoxia-mimicking agents. SUMMARY We hypothesize that REP cells are neuron-like setpoint providers and controllers, which integrate information about blood O2 concentration and local O2 consumption via tissue pO2, and combine these inputs with intrinsic negative feedback loops and perhaps tubular cross-talk, converging in Epo regulation.
Collapse
|
99
|
Aoto M, Iwashita A, Mita K, Ohkubo N, Tsujimoto Y, Mitsuda N. Transferrin receptor 1 is required for enucleation of mouse erythroblasts during terminal differentiation. FEBS Open Bio 2019; 9:291-303. [PMID: 30761254 PMCID: PMC6356176 DOI: 10.1002/2211-5463.12573] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/04/2018] [Accepted: 12/04/2018] [Indexed: 12/25/2022] Open
Abstract
Enucleation is the process whereby the nucleus is extruded from the erythroblast during late stage mammalian erythropoiesis. However, the specific signaling pathways involved in this process remain unclear. To better understand the mechanisms underlying erythroblast enucleation, we investigated erythroblast enucleation using both the spleens of adult mice with phenylhydrazine‐induced anemia and mouse fetal livers. Our results indicated that both iron‐bound transferrin (holo‐Tf) and the small‐molecule iron transporter hinokitiol with iron ions (hinokitiol plus iron) promote hemoglobin synthesis and the enucleation of mouse spleen‐derived erythroblasts. Although an antitransferrin receptor 1 (TfR1) monoclonal antibody inhibited both enucleation and hemoglobin synthesis promoted by holo‐Tf, it inhibited only enucleation, but not hemoglobin synthesis, promoted by hinokitiol plus iron. Furthermore, siRNA against mouse TfR1 were found to suppress the enucleation of mouse fetal liver‐derived erythroblasts, and the endocytosis inhibitor MitMAB inhibited enucleation, hemoglobin synthesis, and the internalization of TfR1 promoted by both types of stimuli. Collectively, our results suggest that TfR1, iron ions, and endocytosis play important roles in mouse erythroblast enucleation.
Collapse
Affiliation(s)
- Mamoru Aoto
- Department of Circulatory Physiology Graduate School of Medicine Ehime University Japan
| | - Akiho Iwashita
- Department of Circulatory Physiology Graduate School of Medicine Ehime University Japan
| | - Kanako Mita
- Department of Circulatory Physiology Graduate School of Medicine Ehime University Japan
| | - Nobutaka Ohkubo
- Department of Circulatory Physiology Graduate School of Medicine Ehime University Japan
| | - Yoshihide Tsujimoto
- Department of Molecular and Cellular Biology Research Center Osaka International Cancer Institute Japan
| | - Noriaki Mitsuda
- Department of Circulatory Physiology Graduate School of Medicine Ehime University Japan
| |
Collapse
|
100
|
Martin TD, Green MS, Whitehead MT, Scheett TP, Webster MJ, Hudson GM. Six weeks of oral Echinacea purpurea supplementation does not enhance the production of serum erythropoietin or erythropoietic status in recreationally active males with above-average aerobic fitness. Appl Physiol Nutr Metab 2019; 44:791-795. [PMID: 30608872 DOI: 10.1139/apnm-2018-0783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to investigate the effect of 6 weeks of oral Echinacea purpurea supplementation on serum erythropoietin (EPO) and erythropoietic status. Twenty-four males (mean ± SE; age = 25.2 ± 1.4 years, height = 178.1 ± 1.4 cm, body mass = 78.1 ± 1.6 kg, body fat = 12.7 ± 0.9%, maximal oxygen uptake = 52.9 ± 0.9 mL·kg-1·min-1) were randomly grouped using a matched-pair, double-blind design and self-administered 8000 mg·day-1 of either E. purpurea (n = 12) or placebo (n = 12) for 42 consecutive days. Blood samples were collected prior to supplementation (day 0) and every 2 weeks during the supplementation period (days 14, 28, and 42) and were analyzed for EPO, red blood cell count, hemoglobin concentration, hematocrit, mean corpuscular volume, and mean corpuscular hemoglobin concentration. Separate 2 × 4 (group × time) factorial ANOVA with repeated measures on time were used to determine statistical differences with significance set at p ≤ 0.05. There were no significant interaction, group, or time effects observed for EPO or erythropoietic status markers for any of the measurement points (p ≤ 0.05). The present study indicated that 6 weeks of oral E. purpurea supplementation in recreationally active males with above average aerobic fitness did not enhance EPO or erythropoietic status. These findings are in contrast with previous reports of E. purpurea supplementation in untrained participants with average fitness levels, but consistent with observations in trained endurance athletes.
Collapse
Affiliation(s)
- Tyler D Martin
- a Department of Kinesiology and Health Promotion, Troy University, Troy, AL 36082, USA
| | - Michael S Green
- a Department of Kinesiology and Health Promotion, Troy University, Troy, AL 36082, USA
| | - Malcolm T Whitehead
- b Department of Kinesiology and Health Science, Stephen F. Austin State University, Nacogdoches, TX 75962, USA
| | - Timothy P Scheett
- c Department of Health and Human Performance, College of Charleston, Charleston, SC 29424, USA
| | - Michael J Webster
- d College of Nursing and Health Sciences, Valdosta State University, Valdosta, GA 31698, USA
| | - Geoffrey M Hudson
- e Department of Health, Kinesiology, & Sport, The University of South Alabama, Mobile, AL 36688, USA
| |
Collapse
|