1,152
Views
27
CrossRef citations to date
0
Altmetric
Reviews

Targeting EPO and EPO receptor pathways in anemia and dysregulated erythropoiesis

, & (Director) (Professor of Medicine) (Director) (Professor of Medicine)

Bibliography

  • Kawasaki N, Ohta M, Hyuga S, et al. Application of liquid chromatography/mass spectrometry and liquid chromatography with tandem mass spectrometry to the analysis of the site-specific carbohydrate heterogeneity in erythropoietin. Anal Biochem 2000;285(1):82-91
  • Froehlich JW, Barboza M, Chu C, et al. Nano-LC-MS/MS of glycopeptides produced by nonspecific proteolysis enables rapid and extensive site-specific glycosylation determination. Anal Chem 2011;83(14):5541-7
  • Bunn HF. Erythropoietin. Cold Spring Harb Perspect Med 2013;3(3):a011619
  • Suzuki N, Hirano I, Pan X, et al. Erythropoietin production in neuroepithelial and neural crest cells during primitive erythropoiesis. Nat Commun 2013;4:2902
  • Koury ST, Bondurant MC, Koury MJ. Localization of erythropoietin synthesizing cells in murine kidneys by in situ hybridization. Blood 1988;71(2):524-7
  • Minamishima YA, Kaelin WGJr. Reactivation of hepatic EPO synthesis in mice after PHD loss. Science 2010;329(5990):407
  • Becker V, Schilling M, Bachmann J, et al. Covering a broad dynamic range: information processing at the erythropoietin receptor. Science 2010;328(5984):1404-8
  • Kuhrt D, Wojchowski DM. Emerging EPO and EPO receptor regulators, and signal transducers. Blood 2015;125(23):3536-41
  • Arcasoy MO, Jiang X. Co-operative signalling mechanisms required for erythroid precursor expansion in response to erythropoietin and stem cell factor. Br J Haematol 2005;130(1):121-9
  • Schnoder TM, Arreba-Tutusaus P, Griehl I, et al. Epo-induced erythroid maturation is dependent on Plcgamma1 signaling. Cell death Differ 2015;22(6):974-85
  • Fang J, Menon M, Kapelle W, et al. EPO modulation of cell-cycle regulatory genes, and cell division, in primary bone marrow erythroblasts. Blood 2007;110(7):2361-70
  • Grover A, Mancini E, Moore S, et al. Erythropoietin guides multipotent hematopoietic progenitor cells toward an erythroid fate. J Exp Med 2014;211(2):181-8
  • Jelkmann W. The ESA scenario gets complex: from biosimilar epoetins to activin traps. Nephrol Dial Transplant 2015;30(4):553-9
  • Ramanath V, Gupta D, Jain J, et al. Anemia and chronic kidney disease: making sense of the recent trials. Rev Recent Clin Trials 2012;7(3):187-96
  • Oster HS, Neumann D, Hoffman M, Mittelman M. Erythropoietin: the swinging pendulum. Leuk Res 2012;36(8):939-44
  • Sullivan PS, Hanson DL, Richardson JT, Brooks JT. Trends in the treatment of anemia using recombinant human erythropoietin in patients with HIV infection. Open AIDS J 2011;5:113-18
  • Tran DH, Wong GT, Chee YE, Irwin MG. Effectiveness and safety of erythropoiesis-stimulating agent use in the perioperative period. Expert Opin Biol Ther 2014;14(1):51-61
  • Komrokji RS, Lancet JE, Swern AS, et al. Combined treatment with lenalidomide and epoetin alfa in lower-risk patients with myelodysplastic syndrome. Blood 2012;120(17):3419-24
  • Hellstrom-Lindberg E, van de Loosdrecht A. Erythropoiesis stimulating agents and other growth factors in low-risk MDS. Best Pract Res Clin Haematol 2013;26(4):401-10
  • Tanhehco YC, Berns JS. Red blood cell transfusion risks in patients with end-stage renal disease. Semin Dial 2012;25(5):539-44
  • Rizzo JD, Brouwers M, Hurley P, et al. American Society of Hematology/American Society of Clinical Oncology clinical practice guideline update on the use of epoetin and darbepoetin in adult patients with cancer. Blood 2010;116(20):4045-59
  • Singh AK, Szczech L, Tang KL, et al. Correction of anemia with epoetin alfa in chronic kidney disease. N Engl J Med 2006;355(20):2085-98
  • Horl WH. Anaemia management and mortality risk in chronic kidney disease. Nat Rev Nephrol 2013;9(5):291-301
  • Strippoli GF, Craig JC, Manno C, Schena FP. Hemoglobin targets for the anemia of chronic kidney disease: a meta-analysis of randomized, controlled trials. J Am Soc Nephrol 2004;15(12):3154-65
  • Krapf R, Hulter HN. Arterial hypertension induced by erythropoietin and erythropoiesis-stimulating agents (ESA). Clin J Am Soc Nephrol 2009;4(2):470-80
  • Vaziri ND, Zhou XJ. Potential mechanisms of adverse outcomes in trials of anemia correction with erythropoietin in chronic kidney disease. Nephrol Dial Transplant 2009;24(4):1082-8
  • Kanbay M, Akcay A, Delibasi T, et al. Comparison of effects of darbepoetin alfa and epoetin alfa on serum endothelin level and blood pressure. Adv Ther 2007;24(2):346-52
  • Barhoumi T, Briet M, Kasal DA, et al. Erythropoietin-induced hypertension and vascular injury in mice overexpressing human endothelin-1: exercise attenuated hypertension, oxidative stress, inflammation and immune response. J Hypertens 2014;32(4):784-94
  • Briet M, Barhoumi T, Mian MO, et al. Effects of recombinant human erythropoietin on resistance artery endothelial function in stage 4 chronic kidney disease. J Am Heart Assoc 2013;2(2):e000128
  • Heinisch BB, Vcelar B, Kapiotis S, et al. The effect of erythropoietin on platelet and endothelial activation markers: a prospective trial in healthy volunteers. Platelets 2012;23(5):352-8
  • Sinclair AM, Coxon A, McCaffery I, et al. Functional erythropoietin receptor is undetectable in endothelial, cardiac, neuronal, and renal cells. Blood 2010;115(21):4264-72
  • Glaspy J. Update on safety of ESAs in cancer-induced anemia. J Natl Compr Canc Netw 2012;10(5):659-66
  • Glaspy J, Crawford J, Vansteenkiste J, et al. Erythropoiesis-stimulating agents in oncology: a study-level meta-analysis of survival and other safety outcomes. Br J Cancer 2010;102(2):301-15
  • Pfeffer MA, Burdmann EA, Chen CY, et al. A trial of darbepoetin alfa in type 2 diabetes and chronic kidney disease. N Engl J Med 2009;361(21):2019-32
  • Bennett CL, Silver SM, Djulbegovic B, et al. Venous thromboembolism and mortality associated with recombinant erythropoietin and darbepoetin administration for the treatment of cancer-associated anemia. Jama 2008;299(8):914-24
  • Leyland-Jones B, Semiglazov V, Pawlicki M, et al. Maintaining normal hemoglobin levels with epoetin alfa in mainly nonanemic patients with metastatic breast cancer receiving first-line chemotherapy: a survival study. J Clin Oncol 2005;23(25):5960-72
  • Henke M, Laszig R, Rube C, et al. Erythropoietin to treat head and neck cancer patients with anaemia undergoing radiotherapy: randomised, double-blind, placebo-controlled trial. Lancet 2003;362(9392):1255-60
  • Untch M, von Minckwitz G, Konecny GE, et al. PREPARE trial: a randomized phase III trial comparing preoperative, dose-dense, dose-intensified chemotherapy with epirubicin, paclitaxel, and CMF versus a standard-dosed epirubicin-cyclophosphamide followed by paclitaxel with or without darbepoetin alfa in primary breast cancer–outcome on prognosis. Ann Oncol 2011;22(9):1999-2006
  • Debeljak N, Solar P, Sytkowski AJ. Erythropoietin and cancer: the unintended consequences of anemia correction. Front Immunol 2014;5:563
  • Elliott S, Busse L, McCaffery I, et al. Identification of a sensitive anti-erythropoietin receptor monoclonal antibody allows detection of low levels of EpoR in cells. J Immunol Methods 2010;352(1-2):126-39
  • Singh S, Verma R, Pradeep A, et al. Dynamic ligand modulation of EPO receptor pools, and dysregulation by polycythemia-associated EPOR alleles. PLoS One 2012;7(1):e29064
  • Elliott S, Busse L, Bass MB, et al. Anti-Epo receptor antibodies do not predict Epo receptor expression. Blood 2006;107(5):1892-5
  • Reinbothe S, Larsson AM, Vaapil M, et al. EPO-independent functional EPO receptor in breast cancer enhances estrogen receptor activity and promotes cell proliferation. Biochem Biophys Res Commun 2014;445(1):163-9
  • Swift S, Ellison AR, Kassner P, et al. Absence of functional EpoR expression in human tumor cell lines. Blood 2010;115(21):4254-63
  • Kataoka M, Moriya Y, Moriguchi Y, et al. Effect of erythropoietin on human tumor growth in xenograft models. Mol Med Rep 2010;3(1):95-101
  • Patterson SD, Rossi JM, Paweletz KL, et al. Functional EpoR pathway utilization is not detected in primary tumor cells isolated from human breast, non-small cell lung, colorectal, and ovarian tumor tissues. PLoS One 2015;10(3):e0122149
  • Kumar SM, Zhang G, Bastian BC, et al. Erythropoietin receptor contributes to melanoma cell survival in vivo. Oncogene 2012;31(13):1649-60
  • Wu P, Zhang N, Wang X, et al. The erythropoietin/erythropoietin receptor signaling pathway promotes growth and invasion abilities in human renal carcinoma cells. PLoS One 2012;7(9):e45122
  • Labrecque MP, Prefontaine GG, Beischlag TV. The aryl hydrocarbon receptor nuclear translocator (ARNT) family of proteins: transcriptional modifiers with multi-functional protein interfaces. Curr Mol Med 2013;13(7):1047-65
  • Madan A, Lin C, Hatch SLII, Curtin PT. Regulated basal, inducible, and tissue-specific human erythropoietin gene expression in transgenic mice requires multiple cis DNA sequences. Blood 1995;85(10):2735-41
  • Obara N, Suzuki N, Kim K, et al. Repression via the GATA box is essential for tissue-specific erythropoietin gene expression. Blood 2008;111(10):5223-32
  • Palazon A, Goldrath AW, Nizet V, Johnson RS. HIF transcription factors, inflammation, and immunity. Immunity 2014;41(4):518-28
  • Haase VH. Regulation of erythropoiesis by hypoxia-inducible factors. Blood Rev 2013;27(1):41-53
  • Anderson SA, Nizzi CP, Chang YI, et al. The IRP1-HIF-2alpha axis coordinates iron and oxygen sensing with erythropoiesis and iron absorption. Cell Metab 2013;17(2):282-90
  • Wilkinson N, Pantopoulos K. The IRP/IRE system in vivo: insights from mouse models. Front Pharmacol 2014;5:176
  • Mahapatra L, Mao C, Andruska N, et al. High-throughput fluorescence anisotropy screen for inhibitors of the oncogenic mRNA binding protein, IMP-1. J Biomol Screen 2014;19(3):427-36
  • Wu X, Lan L, Wilson DM, et al. Identification and Validation of Novel Small Molecule Disruptors of HuR-mRNA Interaction. ACS Chem Biol 2015;10(6):1476-84
  • Alonso N, Guillen R, Chambers JW, Leng F. A rapid and sensitive high-throughput screening method to identify compounds targeting protein-nucleic acids interactions. Nucleic Acids Res 2015;43(8):e52
  • Ferecatu I, Goncalves S, Golinelli-Cohen MP, et al. The diabetes drug target MitoNEET governs a novel trafficking pathway to rebuild an Fe-S cluster into cytosolic aconitase/iron regulatory protein 1. J Biol Chem 2014;289(41):28070-86
  • Russell RC, Sufan RI, Zhou B, et al. Loss of JAK2 regulation via a heterodimeric VHL-SOCS1 E3 ubiquitin ligase underlies Chuvash polycythemia. Nat Med 2011;17(7):845-53
  • Ang SO, Chen H, Hirota K, et al. Disruption of oxygen homeostasis underlies congenital Chuvash polycythemia. Nat Genet 2002;32(4):614-21
  • Bader HL, Hsu T. Systemic VHL gene functions and the VHL disease. FEBS Lett 2012;586(11):1562-9
  • Franke K, Gassmann M, Wielockx B. Erythrocytosis: the HIF pathway in control. Blood 2013;122(7):1122-8
  • Bishop T, Ratcliffe PJ. HIF Hydroxylase Pathways in Cardiovascular Physiology and Medicine. Circ Res 2015;117(1):65-79
  • Selvaraju V, Parinandi NL, Adluri RS, et al. Molecular mechanisms of action and therapeutic uses of pharmacological inhibitors of HIF-prolyl 4-hydroxylases for treatment of ischemic diseases. Antioxid Redox Signal 2014;20(16):2631-65
  • Goldmacher VS, Lambert JM, Young AY, et al. Expression of the common acute lymphoblastic leukemia antigen (CALLA) on the surface of individual cells of human lymphoblastoid lines. J Immunol 1986;136(1):320-5
  • Semenza GL. Molecular mechanisms mediating metastasis of hypoxic breast cancer cells. Trends Mol Med 2012;18(9):534-43
  • Scortegagna M, Ding K, Zhang Q, et al. HIF-2alpha regulates murine hematopoietic development in an erythropoietin-dependent manner. Blood 2005;105(8):3133-40
  • Percy MJ, Furlow PW, Lucas GS, et al. A gain-of-function mutation in the HIF2A gene in familial erythrocytosis. N Engl J Med 2008;358(2):162-8
  • Percy MJ, Beer PA, Campbell G, et al. Novel exon 12 mutations in the HIF2A gene associated with erythrocytosis. Blood 2008;111(11):5400-2
  • Kapitsinou PP, Liu Q, Unger TL, et al. Hepatic HIF-2 regulates erythropoietic responses to hypoxia in renal anemia. Blood 2010;116(16):3039-48
  • Lee FS, Percy MJ. The HIF pathway and erythrocytosis. Annu Rev Pathol 2011;6:165-92
  • Tojo Y, Sekine H, Hirano I, et al. Hypoxia Signaling Cascade for Erythropoietin Production in Hepatocytes. Mol Cell Biol 2015;35(15):2658-72
  • Rankin EB, Wu C, Khatri R, et al. The HIF signaling pathway in osteoblasts directly modulates erythropoiesis through the production of EPO. Cell 2012;149(1):63-74
  • Souma T, Nezu M, Nakano D, et al. Erythropoietin synthesis in renal myofibroblasts is restored by activation of hypoxia signaling. J Am Soc Nephrol 2015. [Epub ahead of print]
  • Laitala A, Aro E, Walkinshaw G, et al. Transmembrane prolyl 4-hydroxylase is a fourth prolyl 4-hydroxylase regulating EPO production and erythropoiesis. Blood 2012;120(16):3336-44
  • Flamme I, Oehme F, Ellinghaus P, et al. Mimicking hypoxia to treat anemia: HIF-stabilizer BAY 85-3934 (Molidustat) stimulates erythropoietin production without hypertensive effects. PLoS One 2014;9(11):e111838
  • Koditz J, Nesper J, Wottawa M, et al. Oxygen-dependent ATF-4 stability is mediated by the PHD3 oxygen sensor. Blood 2007;110(10):3610-17
  • Cummins EP, Berra E, Comerford KM, et al. Prolyl hydroxylase-1 negatively regulates IkappaB kinase-beta, giving insight into hypoxia-induced NFkappaB activity. Proc Natl Acad Sci U S A 2006;103(48):18154-9
  • Hiwatashi Y, Kanno K, Takasaki C, et al. PHD1 interacts with ATF4 and negatively regulates its transcriptional activity without prolyl hydroxylation. Exp Cell Res 2011;317(20):2789-99
  • Gilkes DM, Semenza GL. Role of hypoxia-inducible factors in breast cancer metastasis. Future Oncol 2013;9(11):1623-36
  • Xu M, Nagati JS, Xie J, et al. An acetate switch regulates stress erythropoiesis. Nat Med 2014;20(9):1018-26
  • Chen R, Xu M, Nagati JS, et al. The Acetate/ACSS2 switch regulates HIF-2 stress signaling in the tumor cell microenvironment. PLoS One 2015;10(2):e0116515
  • Livnah O, Stura EA, Middleton SA, et al. Crystallographic evidence for preformed dimers of erythropoietin receptor before ligand activation. Science 1999;283(5404):987-90
  • Pelletier S, Gingras S, Funakoshi-Tago M, et al. Two domains of the erythropoietin receptor are sufficient for Jak2 binding/activation and function. Mol Cell Biol 2006;26(22):8527-38
  • Zhang YL, Radhakrishnan ML, Lu X, et al. Symmetric signaling by an asymmetric 1 erythropoietin: 2 erythropoietin receptor complex. Mol Cell 2009;33(2):266-74
  • Song KE, Byeon J, Moon DB, et al. Structural identification of modified amino acids on the interface between EPO and its receptor from EPO BRP, human recombinant erythropoietin by LC/MS analysis. Mol Cells 2014;37(11):819-26
  • Elliott S, Lorenzini T, Chang D, et al. Mapping of the active site of recombinant human erythropoietin. Blood 1997;89(2):493-502
  • Liu Z, Stoll VS, Devries PJ, et al. A potent erythropoietin-mimicking human antibody interacts through a novel binding site. Blood 2007;110(7):2408-13
  • Lacy SE, DeVries PJ, Xie N, et al. The potency of erythropoietin-mimic antibodies correlates inversely with affinity. J Immunol 2008;181(2):1282-7
  • Casadevall N, Nataf J, Viron B, et al. Pure red-cell aplasia and antierythropoietin antibodies in patients treated with recombinant erythropoietin. N Engl J Med 2002;346(7):469-75
  • Aoki K, Ono Y, Tabata S, et al. Successful treatment of anti-erythropoietin antibody-mediated pure red cell aplasia with low-dose prednisolone. Int J Hematol 2013;97(2):272-4
  • Matsuki E, Miyakawa Y, Yamane A, Okamoto S. Humanized VB22B minibody for human Mpl stimulates human megakaryopoiesis but does not enhance platelet aggregation. Exp Hematol 2011;39(8):829-36
  • Wrighton NC, Farrell FX, Chang R, et al. Small peptides as potent mimetics of the protein hormone erythropoietin. Science 1996;273(5274):458-64
  • Barbone FP, Johnson DL, Farrell FX, et al. New epoetin molecules and novel therapeutic approaches. Nephrol Dial Transplant 1999;14(Suppl 2):80-4
  • Bugelski PJ, Capocasale RJ, Makropoulos D, et al. CNTO 530: molecular pharmacology in human UT-7EPO cells and pharmacokinetics and pharmacodynamics in mice. J Biotechnol 2008;134(1-2):171-80
  • Perez-Ruixo JJ, Krzyzanski W, Bouman-Thio E, et al. Pharmacokinetics and pharmacodynamics of the erythropoietin Mimetibody construct CNTO 528 in healthy subjects. Clin Pharmacokinet 2009;48(9):601-13
  • Sathyanarayana P, Houde E, Marshall D, et al. CNTO 530 functions as a potent EPO mimetic via unique sustained effects on bone marrow proerythroblast pools. Blood 2009;113(20):4955-62
  • Makropoulos DA, Achuthanandam R, Avery J, et al. CNTO 530 increases expression of HbA and HbF in murine models of beta-thalassemia and sickle cell anemia. Curr Pharm Biotechnol 2013;14(2):242-8
  • Fishbane S, Schiller B, Locatelli F, et al. Peginesatide in patients with anemia undergoing hemodialysis. N Engl J Med 2013;368(4):307-19
  • Macdougall IC, Rossert J, Casadevall N, et al. A peptide-based erythropoietin-receptor agonist for pure red-cell aplasia. N Engl J Med 2009;361(19):1848-55
  • Bennett CL, Jacob S, Hymes J, et al. Anaphylaxis and hypotension after administration of peginesatide. N Engl J Med 2014;370(21):2055-6
  • Zhang J, Randall MS, Loyd MR, et al. Role of erythropoietin receptor signaling in Friend virus-induced erythroblastosis and polycythemia. Blood 2006;107(1):73-8
  • Lu X, Gross AW, Lodish HF. Active conformation of the erythropoietin receptor: random and cysteine-scanning mutagenesis of the extracellular juxtamembrane and transmembrane domains. J Biol Chem 2006;281(11):7002-11
  • Cammett TJ, Jun SJ, Cohen EB, et al. Construction and genetic selection of small transmembrane proteins that activate the human erythropoietin receptor. Proc Natl Acad Sci U S A 2010;107(8):3447-52
  • Erickson-Miller CL, DeLorme E, Tian SS, et al. Discovery and characterization of a selective, nonpeptidyl thrombopoietin receptor agonist. Exp Hematol 2005;33(1):85-93
  • Nurden AT, Viallard JF, Nurden P. New-generation drugs that stimulate platelet production in chronic immune thrombocytopenic purpura. Lancet 2009;373(9674):1562-9
  • Kim YK, Lee SS, Jeong SH, et al. Efficacy and safety of eltrombopag in adult refractory immune thrombocytopenia. Blood Res 2015;50(1):19-25
  • Erickson-Miller CL, Delorme E, Tian SS, et al. Preclinical activity of eltrombopag (SB-497115), an oral, nonpeptide thrombopoietin receptor agonist. Stem Cells 2009;27(2):424-30
  • Kautz L, Jung G, Valore EV, et al. Identification of erythroferrone as an erythroid regulator of iron metabolism. Nat Genet 2014;46(7):678-84
  • Kautz L, Jung G, Nemeth E, Ganz T. Erythroferrone contributes to recovery from anemia of inflammation. Blood 2014;124(16):2569-74
  • Rochette L, Gudjoncik A, Guenancia C, et al. The iron-regulatory hormone hepcidin: a possible therapeutic target? Pharmacol Ther 2015;146:35-52
  • Fibach E, Rachmilewitz E. The role of oxidative stress in hemolytic anemia. Curr Mol Med 2008;8(7):609-19
  • Scheibye-Knudsen M, Fang EF, Croteau DL, et al. Protecting the mitochondrial powerhouse. Trends Cell Biol 2015;25(3):158-70
  • Prus E, Fibach E. Effect of iron chelators on labile iron and oxidative status of thalassaemic erythroid cells. Acta Haematol 2010;123(1):14-20
  • Dev A, Byrne SM, Verma R, et al. Erythropoietin-directed erythropoiesis depends on serpin inhibition of erythroblast lysosomal cathepsins. J Exp Med 2013;210(2):225-32
  • Sbongile M, Soliman ME. In silico identification of irreversible cathepsin B inhibitors as anti- cancer agents: virtual screening, covalent docking analysis and molecular dynamics simulations. Comb Chem High Throughput Screen 2015;18(4):399-410
  • Nabinger SC, Chan RJ. Shp2 function in hematopoietic stem cell biology and leukemogenesis. Curr Opin Hematol 2012;19(4):273-9
  • Chong ZZ, Maiese K. The Src homology 2 domain tyrosine phosphatases SHP-1 and SHP-2: diversified control of cell growth, inflammation, and injury. Histol Histopathol 2007;22(11):1251-67
  • Wojchowski DM, Su S, Johnson A, et al. Discovery of novel EPO/EPOR/JAK2 targets and signal transduction factors via proteomic-based interrogations of post-translational motif modifications. American Society of Hematology Annual Meeting Proceedings: Blood. 2014. p. 1335
  • Li S, Hsu DD, Wang H, Feng GS. Dual faces of SH2-containing protein-tyrosine phosphatase Shp2/PTPN11 in tumorigenesis. Front Med 2012;6(3):275-9
  • Usenko T, Chan G, Torlakovic E, et al. Leukemogenic Ptpn11 allele causes defective erythropoiesis in mice. PLoS One 2014;9(10):e109682
  • Sharma N, Kumar V, Everingham S, et al. SH2 domain-containing phosphatase 2 is a critical regulator of connective tissue mast cell survival and homeostasis in mice. Mol Cell Biol 2012;32(14):2653-63
  • Sharma N, Everingham S, Zeng LF, et al. Oncogenic KIT-induced aggressive systemic mastocytosis requires SHP2/PTPN11 phosphatase for disease progression in mice. Oncotarget 2014;5(15):6130-41
  • Poole AW, Jones ML. A SHPing tale: perspectives on the regulation of SHP-1 and SHP-2 tyrosine phosphatases by the C-terminal tail. Cell Signal 2005;17(11):1323-32
  • Van Zant G, Shultz L. Hematologic abnormalities of the immunodeficient mouse mutant, viable motheaten (mev). Exp Hematol 1989;17(2):81-7
  • Klingmuller U, Lorenz U, Cantley LC, et al. Specific recruitment of SH-PTP1 to the erythropoietin receptor causes inactivation of JAK2 and termination of proliferative signals. Cell 1995;80(5):729-38
  • Wang HM, Xu YF, Ning SL, et al. The catalytic region and PEST domain of PTPN18 distinctly regulate the HER2 phosphorylation and ubiquitination barcodes. Cell Res 2014;24(9):1067-90
  • Huang WQ, Lin Q, Zhuang X, et al. Structure, function, and pathogenesis of SHP2 in developmental disorders and tumorigenesis. Curr Cancer Drug Targets 2014;14(6):567-88
  • He RJ, Yu ZH, Zhang RY, Zhang ZY. Protein tyrosine phosphatases as potential therapeutic targets. Acta Pharmacol Sin 2014;35(10):1227-46
  • Chio CM, Lim CS, Bishop AC. Targeting a cryptic allosteric site for selective inhibition of the oncogenic protein tyrosine phosphatase Shp2. Biochem 2015;54(2):497-504
  • Geyer HL, Mesa RA. Therapy for myeloproliferative neoplasms: when, which agent, and how? Blood 2014;124(24):3529-37
  • Wang LJ, Jiang B, Wu N, et al. Small molecules as potent protein tyrosine phosphatase 1B (PTP1B) inhibitors documented in patents from 2009 to 2013. Mini Rev Med Chem 2015;15(2):104-22
  • Hou X, Li R, Li K, et al. Fast identification of novel lymphoid tyrosine phosphatase inhibitors using target-ligand interaction-based virtual screening. J Med Chem 2014;57(22):9309-22
  • Low JL, Chai CL, Yao SQ. Bidentate inhibitors of protein tyrosine phosphatases. Antioxid Redox Signal 2014;20(14):2225-50
  • Peslak SA, Wenger J, Bemis JC, et al. EPO-mediated expansion of late-stage erythroid progenitors in the bone marrow initiates recovery from sublethal radiation stress. Blood 2012;120(12):2501-11
  • Dussiot M, Maciel TT, Fricot A, et al. An activin receptor IIA ligand trap corrects ineffective erythropoiesis in beta-thalassemia. Nat Med 2014;20(4):398-407
  • Guihard S, Clay D, Cocault L, et al. The MAPK ERK1 is a negative regulator of the adult steady-state splenic erythropoiesis. Blood 2010;115(18):3686-94
  • Wu DC, Paulson RF. Hypoxia regulates BMP4 expression in the murine spleen during the recovery from acute anemia. PLoS One 2010;5(6):e11303
  • Harandi OF, Hedge S, Wu DC, et al. Murine erythroid short-term radioprotection requires a BMP4-dependent, self-renewing population of stress erythroid progenitors. J Clin Invest 2010;120(12):4507-19
  • Lau CI, Outram SV, Saldana JI, et al. Regulation of murine normal and stress-induced erythropoiesis by Desert Hedgehog. Blood 2012;119(20):4741-51
  • An X, Schulz VP, Li J, et al. Global transcriptome analyses of human and murine terminal erythroid differentiation. Blood 2014;123(22):3466-77
  • Paralkar VR, Mishra T, Luan J, et al. Lineage and species-specific long noncoding RNAs during erythro-megakaryocytic development. Blood 2014;123(12):1927-37
  • Khoriaty R, Vasievich MP, Jones M, et al. Absence of a red blood cell phenotype in mice with hematopoietic deficiency of SEC23B. Mol Cell Biol 2014;34(19):3721-34
  • Verma R, Su S, McCrann DJ, et al. RHEX, a novel regulator of human erythroid progenitor cell expansion and erythroblast development. J Exp Med 2014;211(9):1715-22
  • Trakarnsanga K, Wilson MC, Lau W, et al. Induction of adult levels of beta-globin in human erythroid cells that intrinsically express embryonic or fetal globin by transduction with KLF1 and BCL11A-XL. Haematologica 2014;99(11):1677-85
  • Huang X, Shah S, Wang J, et al. Extensive ex vivo expansion of functional human erythroid precursors established from umbilical cord blood cells by defined factors. Mol Ther 2014;22(2):451-63
  • Saliba J, Hamidi S, Lenglet G, et al. Heterozygous and homozygous JAK2(V617F) states modeled by induced pluripotent stem cells from myeloproliferative neoplasm patients. PLoS One 2013;8(9):e74257
  • Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med 2013;369(25):2379-90
  • Asada N, Takase M, Nakamura J, et al. Dysfunction of fibroblasts of extrarenal origin underlies renal fibrosis and renal anemia in mice. J Clin Invest 2011;121(10):3981-90
  • Xie L, Pi X, Townley-Tilson WH, et al. PHD2/3-dependent hydroxylation tunes cardiac response to beta-adrenergic stress via phospholamban. J Clin Invest 2015;125(7):2759-71
  • Miyake S, Muramatsu R, Hamaguchi M, Yamashita T. Prolyl hydroxylase regulates axonal rewiring and motor recovery after traumatic brain injury. Cell Death Dis 2015;6:e1638

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.