Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 206
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 168(6): 1053-1064.e15, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28283061

ABSTRACT

Cytokines are classically thought to stimulate downstream signaling pathways through monotonic activation of receptors. We describe a severe anemia resulting from a homozygous mutation (R150Q) in the cytokine erythropoietin (EPO). Surprisingly, the EPO R150Q mutant shows only a mild reduction in affinity for its receptor but has altered binding kinetics. The EPO mutant is less effective at stimulating erythroid cell proliferation and differentiation, even at maximally potent concentrations. While the EPO mutant can stimulate effectors such as STAT5 to a similar extent as the wild-type ligand, there is reduced JAK2-mediated phosphorylation of select downstream targets. This impairment in downstream signaling mechanistically arises from altered receptor dimerization dynamics due to extracellular binding changes. These results demonstrate how variation in a single cytokine can lead to biased downstream signaling and can thereby cause human disease. Moreover, we have defined a distinct treatable form of anemia through mutation identification and functional studies.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/pathology , Erythropoietin/genetics , Mutation, Missense , Signal Transduction , Anemia, Diamond-Blackfan/therapy , Child , Consanguinity , Enzyme Activation , Erythropoiesis , Erythropoietin/chemistry , Female , Humans , Janus Kinase 2/metabolism , Kinetics , Male , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism
2.
Cell ; 160(6): 1196-208, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25728669

ABSTRACT

Most cell-surface receptors for cytokines and growth factors signal as dimers, but it is unclear whether remodeling receptor dimer topology is a viable strategy to "tune" signaling output. We utilized diabodies (DA) as surrogate ligands in a prototypical dimeric receptor-ligand system, the cytokine Erythropoietin (EPO) and its receptor (EpoR), to dimerize EpoR ectodomains in non-native architectures. Diabody-induced signaling amplitudes varied from full to minimal agonism, and structures of these DA/EpoR complexes differed in EpoR dimer orientation and proximity. Diabodies also elicited biased or differential activation of signaling pathways and gene expression profiles compared to EPO. Non-signaling diabodies inhibited proliferation of erythroid precursors from patients with a myeloproliferative neoplasm due to a constitutively active JAK2V617F mutation. Thus, intracellular oncogenic mutations causing ligand-independent receptor activation can be counteracted by extracellular ligands that re-orient receptors into inactive dimer topologies. This approach has broad applications for tuning signaling output for many dimeric receptor systems.


Subject(s)
Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Cell Line , Crystallography, X-Ray , Dimerization , Erythropoietin/metabolism , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Point Mutation , Protein Engineering , Receptors, Erythropoietin/agonists , Receptors, Erythropoietin/antagonists & inhibitors , Sequence Alignment
3.
Nat Chem Biol ; 20(6): 751-760, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38480980

ABSTRACT

Transmembrane (TM) domains as simple as a single span can perform complex biological functions using entirely lipid-embedded chemical features. Computational design has the potential to generate custom tool molecules directly targeting membrane proteins at their functional TM regions. Thus far, designed TM domain-targeting agents have been limited to mimicking the binding modes and motifs of natural TM interaction partners. Here, we demonstrate the design of de novo TM proteins targeting the erythropoietin receptor (EpoR) TM domain in a custom binding topology competitive with receptor homodimerization. The TM proteins expressed in mammalian cells complex with EpoR and inhibit erythropoietin-induced cell proliferation. In vitro, the synthetic TM domain complex outcompetes EpoR homodimerization. Structural characterization reveals that the complex involves the intended amino acids and agrees with our designed molecular model of antiparallel TM helices at 1:1 stoichiometry. Thus, membrane protein TM regions can now be targeted in custom-designed topologies.


Subject(s)
Membrane Proteins , Protein Binding , Receptors, Erythropoietin , Humans , Membrane Proteins/metabolism , Membrane Proteins/chemistry , Receptors, Erythropoietin/metabolism , Receptors, Erythropoietin/chemistry , Models, Molecular , Cell Proliferation/drug effects , Receptors, Cytokine/metabolism , Receptors, Cytokine/chemistry , Amino Acid Sequence , Protein Multimerization , Animals , HEK293 Cells
4.
Am J Transplant ; 19(9): 2407-2414, 2019 09.
Article in English | MEDLINE | ID: mdl-30903735

ABSTRACT

Erythropoietin (EPO) is a glycoprotein produced mainly by the adult kidney in response to hypoxia and is the crucial regulator of red blood cell production. EPO receptors (EPORs), however, are not confined to erythroid cells, but are expressed by many organs including the heart, brain, retina, pancreas, and kidney, where they mediate EPO-induced, erythropoiesis-independent, tissue-protective effects. Some of these tissues also produce and locally release small amounts of EPO in response to organ injury as a mechanism of self-repair. Growing evidence shows that EPO possesses also important immune-modulating effects. Monocytes can produce EPO, and autocrine EPO/EPOR signaling in these cells is crucial in maintaining immunologic self-tolerance. New data in mice and humans also indicate that EPO has a direct inhibitory effect on effector/memory T cells, while it promotes formation of regulatory T cells. This review examines the nonerythropoietic effects of EPO, with a special emphasis on its modulating activity on innate immune cells and T cells and on how it affects transplant outcomes.


Subject(s)
Erythropoietin/physiology , Receptors, Erythropoietin/physiology , Animals , Apoptosis , Erythrocytes/metabolism , Erythropoietin/chemistry , Humans , Hypoxia , Immunity, Innate , Immunologic Memory , Mice , Monocytes/metabolism , Protein Binding , Receptors, Erythropoietin/chemistry , Regeneration , Signal Transduction , T-Lymphocytes , T-Lymphocytes, Regulatory/cytology , Transplants/immunology
5.
Biophys J ; 112(4): 595-604, 2017 Feb 28.
Article in English | MEDLINE | ID: mdl-28256220

ABSTRACT

Based on differences between the x-ray crystal structures of ligand-bound and unbound forms, the activation of the erythropoietin receptor (EPOR) was initially proposed to involve a cross-action scissorlike motion. However, the validity of the motions involved in the scissorlike model has been recently challenged. Here, atomistic molecular dynamics simulations are used to examine the structure of the extracellular domain of the EPOR dimer in the presence and absence of erythropoietin and a series of agonistic or antagonistic mimetic peptides free in solution. The simulations suggest that in the absence of crystal packing effects, the EPOR chains in the different dimers adopt very similar conformations with no clear distinction between the agonist and antagonist-bound complexes. This questions whether the available x-ray crystal structures of EPOR truly represent active or inactive conformations. The study demonstrates the difficulty in using such structures to infer a mechanism of action, especially in the case of membrane receptors where just part of the structure has been considered in addition to potential confounding effects that arise from the comparison of structures in a crystal as opposed to a membrane environment. The work highlights the danger of assigning functional significance to small differences between structures of proteins bound to different ligands in a crystal environment without consideration of the effects of the crystal lattice and thermal motion.


Subject(s)
Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/metabolism , Amino Acid Sequence , Apoproteins/chemistry , Apoproteins/metabolism , Crystallography, X-Ray , Ligands , Models, Molecular , Protein Binding , Protein Multimerization , Protein Structure, Quaternary
6.
Mol Cell ; 33(2): 266-74, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19187767

ABSTRACT

Via sites 1 and 2, erythropoietin binds asymmetrically to two identical receptor monomers, although it is unclear how asymmetry affects receptor activation and signaling. Here we report the design and validation of two mutant erythropoietin receptors that probe the role of individual members of the receptor dimer by selectively binding either site 1 or site 2 on erythropoietin. Ba/F3 cells expressing either mutant receptor do not respond to erythropoietin, but cells co-expressing both receptors respond to erythropoietin by proliferation and activation of the JAK2-Stat5 pathway. A truncated receptor with only one cytosolic tyrosine (Y343) is sufficient for signaling in response to erythropoietin, regardless of the monomer on which it is located. Similarly, only one receptor in the dimer needs a juxtamembrane hydrophobic L253 or W258 residue, essential for JAK2 activation. We conclude that despite asymmetry in the ligand-receptor interaction, both sides are competent for signaling, and appear to signal equally.


Subject(s)
Erythropoietin/chemistry , Erythropoietin/metabolism , Receptors, Erythropoietin/metabolism , Signal Transduction , Binding Sites , Cell Proliferation , Cells, Cultured , Computer Simulation , Humans , Janus Kinase 2/metabolism , Models, Molecular , Mutation , Protein Conformation , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , STAT5 Transcription Factor/metabolism , Tyrosine/genetics , Tyrosine/metabolism
7.
Protein Expr Purif ; 111: 1-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25795089

ABSTRACT

Recombinant human erythropoietin receptor (rhEPOR) has applicability as an affinity ligand for purification of recombinant human erythropoietin (rHuEPO) because of its specific binding to rHuEPO. For application of rhEPOR as a ligand for purification of rHuEPO, soluble rhEPOR was expressed in the periplasm of Escherichia coli and engineered by directed evolution through random mutagenesis and integration of mutations. From the screening of random mutagenesis, we identified an amino acid mutation (H114Y) contributing to rHuEPO binding and four amino acid mutations (R76S, A132D, A162D, and C181Y) contributing to expression of soluble rhEPOR. However, the rHuEPO that binds to engineered rhEPOR having H114Y mutation is difficult to dissociate from the engineered rhEPOR. Therefore, H114Y mutation was not suitable for the construction of the rhEPOR ligand. As a rhEPOR ligand, engineered rhEPOR containing four amino acid mutations (EPORm4L) was constructed by integration of mutations except for H114Y. The expression of EPORm4L (127mgl(-1) of culture medium) was markedly increased in comparison with wild-type rhEPOR (2mgl(-1) of culture medium). Small-scale affinity chromatography demonstrated that EPORm4L worked as an affinity ligand for purification of rHuEPO.


Subject(s)
Protein Engineering , Receptors, Erythropoietin , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Receptors, Erythropoietin/blood , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/isolation & purification , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
8.
Biophys J ; 107(10): 2325-36, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25418301

ABSTRACT

Erythropoietin receptor (EpoR) dimerization is an important step in erythrocyte formation. Its transmembrane domain (TMD) and juxtamembrane (JM) region are essential for signal transduction across the membrane. A construct compassing residues S212-P259 and containing the TMD and JM region of the human EpoR was purified and reconstituted in detergent micelles. The solution structure of the construct was determined in dodecylphosphocholine (DPC) micelles by solution NMR spectroscopy. Structural and dynamic studies demonstrated that the TMD and JM region are an ?-helix in DPC micelles, whereas residues S212-D224 at the N-terminus of the construct are not structured. The JM region is a helix that contains a hydrophobic patch formed by conserved hydrophobic residues (L253, I257, and W258). Nuclear Overhauser effect analysis, fluorescence spectroscopy, and paramagnetic relaxation enhancement experiments suggested that the JM region is exposed to the solvent. The structures of the TMD and JM region of the mouse EpoR were similar to those of the human EpoR.


Subject(s)
Cell Membrane , Micelles , Receptors, Erythropoietin/chemistry , Amino Acid Sequence , Animals , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Models, Molecular , Molecular Sequence Data , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Solvents/chemistry
9.
Br J Haematol ; 165(4): 519-28, 2014 May.
Article in English | MEDLINE | ID: mdl-24533580

ABSTRACT

Primary familial and congenital polycythaemia (PFCP) is a disease characterized by increased red blood cell mass, and can be associated with mutations in the intracellular region of the erythropoietin (EPO) receptor (EPOR). Here we explore the mechanisms by which EPOR mutations induce PFCP, using an experimental system based on chimeric receptors between epidermal growth factor receptor (EGFR) and EPOR. The design of the chimeras enabled EPOR signalling to be triggered by EGF binding. Using this system we analysed three novel EPOR mutations discovered in PFCP patients: a deletion mutation (Del1377-1411), a nonsense mutation (C1370A) and a missense mutation (G1445A). Three different chimeras, bearing these mutations in the cytosolic, EPOR region were generated; Hence, the differences in the chimera-related effects are specifically attributed to the mutations. The results show that the different mutations affect various aspects related to the signalling and metabolism of the chimeric receptors. These include slower degradation rate, higher levels of glycan-mature chimeric receptors, increased sensitivity to low levels of EGF (replacing EPO in this system) and extended signalling cascades. This study provides a novel experimental system to study polycythaemia-inducing mutations in the EPOR, and sheds new light on underlying mechanisms of EPOR over-activation in PFCP patients.


Subject(s)
Polycythemia/congenital , Receptors, Erythropoietin/genetics , Cell Line , Cell Membrane/metabolism , Codon, Nonsense , DNA Mutational Analysis , ErbB Receptors/genetics , Glycosylation , Humans , In Vitro Techniques , Janus Kinase 2/metabolism , MAP Kinase Signaling System , Mutation, Missense , Polycythemia/genetics , Polysaccharides/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Protein Structure, Tertiary , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/metabolism , Recombinant Fusion Proteins/metabolism , STAT5 Transcription Factor/physiology , Sequence Deletion , Signal Transduction/genetics , Structure-Activity Relationship , Transfection
10.
Anal Chem ; 86(24): 12014-21, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25423444

ABSTRACT

Human erythropoietin (hEPO) is an erythropoiesis stimulating hormone frequently employed in antianemia therapy. Its capability to increase the amount of red blood cells however makes hEPO and its derivatives also attractive to dishonest athletes aiming at an artificial and illicit enhancement of their endurance performance. A major objective of the international antidoping fight is the elimination of drug misuse and prevention of severe adverse effects caused by nontherapeutic administrations of highly potent drugs. The emergence of novel and innovative erythropoietin-mimetic agents (EMAs) has been continuously growing in the last years, and the option of using dedicated monoclonal antibodies (mAb) for analytical and sample preparation approaches is gradually reaching limits. In the present study the common ability and property of all EMAs, to bind on the human erythropoietin receptor (hEPOR), is therefore exploited. An alternative methodology to isolate and analyze EMAs, in particular endogenous EPO and the recombinant forms EPOzeta, darbepoetin alfa, and C.E.R.A., from human urine is described, employing conventional ultrafiltration for preconcentration of the target analytes followed by EMA-specific isolation via hEPOR-bound magnetic beads. Analytical data were generated by means of gel-based electrophoretic analysis and nanoliquid chromatography/high resolution/high accuracy tandem mass spectrometry. Limits of detection enabled by the established sample preparation protocols were approximately 20 pg/mL for EPOzeta, 30 pg/mL for darbepoetin alfa, and 80 pg/mL for C.E.R.A.


Subject(s)
Chromatography, Liquid , Doping in Sports , Erythropoietin/urine , Receptors, Erythropoietin/chemistry , Tandem Mass Spectrometry , Urinalysis/methods , Erythropoietin/chemistry , Humans , Magnetic Phenomena
11.
J Biol Chem ; 287(31): 26223-34, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22669948

ABSTRACT

Gene targeting experiments have shown that the cytokine erythropoietin (EPO), its cognate erythropoietin receptor (EPO-R), and associated Janus tyrosine kinase, JAK2, are all essential for erythropoiesis. Structural-functional and murine knock-in experiments have suggested that EPO-R Tyr-343 is important in EPO-mediated mitogenesis. Although Stat5 binds to EPO-R phosphotyrosine 343, the initial Stat5-deficient mice did not have profound erythroid abnormalities suggesting that additional Src homology 2 (SH2) domain-containing effectors may bind to EPO-R Tyr-343 and couple to downstream signaling pathways. We have utilized cloning of ligand target (COLT) screening to demonstrate that EPO-R Tyr(P)-343 and Tyr(P)-401 bind to the SH2 domain-containing adaptor protein SH2B1ß. Immunoprecipitation and in vitro mixing experiments reveal that EPO-R binds to SH2B1 in an SH2 domain-dependent manner and that the sequence that confers SH2B1 binding to the EPO-R is pYXXL. Previous studies have shown that SH2B1 binds directly to JAK2, but we show that in hematopoietic cells, SH2B1ß preferentially associates with the EPO-R. SH2B1 is capable of constitutive association with EPO-R, which is necessary for its optimal SH2-dependent recruitment to EPO-R-Tyr(P)-343/Tyr(P)-401. We also demonstrate that SH2B1 is responsive to EPO stimulation and becomes phosphorylated, most likely on serines/threonines, in an EPO dose- and time-dependent manner. In the absence of SH2B1, we observe enhanced activation of signaling pathways downstream of the EPO-R, indicating that SH2B1 is a negative regulator of EPO signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Receptors, Erythropoietin/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/isolation & purification , Animals , Cell Line , Erythroblasts/metabolism , Erythropoietin/physiology , Humans , Immunoprecipitation , Mice , Mice, Inbred C57BL , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Phosphorylation , Primary Cell Culture , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/isolation & purification , Signal Transduction
12.
Proteins ; 81(8): 1351-62, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23504705

ABSTRACT

Hot spot residues of proteins are fundamental interface residues that help proteins perform their functions. Detecting hot spots by experimental methods is costly and time-consuming. Sequential and structural information has been widely used in the computational prediction of hot spots. However, structural information is not always available. In this article, we investigated the problem of identifying hot spots using only physicochemical characteristics extracted from amino acid sequences. We first extracted 132 relatively independent physicochemical features from a set of the 544 properties in AAindex1, an amino acid index database. Each feature was utilized to train a classification model with a novel encoding schema for hot spot prediction by the IBk algorithm, an extension of the K-nearest neighbor algorithm. The combinations of the individual classifiers were explored and the classifiers that appeared frequently in the top performing combinations were selected. The hot spot predictor was built based on an ensemble of these classifiers and to work in a voting manner. Experimental results demonstrated that our method effectively exploited the feature space and allowed flexible weights of features for different queries. On the commonly used hot spot benchmark sets, our method significantly outperformed other machine learning algorithms and state-of-the-art hot spot predictors. The program is available at http://sfb.kaust.edu.sa/pages/software.aspx.


Subject(s)
Proteins/chemistry , Proteins/metabolism , Algorithms , Amino Acid Sequence , Amino Acids/chemistry , Amino Acids/metabolism , Animals , Artificial Intelligence , Databases, Protein , Drosophila/chemistry , Drosophila/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Humans , Juvenile Hormones/chemistry , Juvenile Hormones/metabolism , Models, Molecular , Protein Interaction Maps , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/metabolism
13.
Nat Genet ; 26(1): 64-6, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973250

ABSTRACT

A major obstacle to stem-cell gene therapy rests in the inability to deliver a gene into a therapeutically relevant fraction of stem cells. One way to circumvent this obstacle is to use selection. Vectors containing two linked genes serve as the basis for selection, with one gene encoding a selectable product and the other, a therapeutic protein. Applying selection in vivo has the potential to bring a minor population of genetically corrected cells into the therapeutic range. But strategies for achieving in vivo selection have traditionally relied on genes that confer resistance to cytotoxic drugs and are encumbered by toxicity. Here we describe a new system for in vivo selection that uses a 'cell-growth switch', allowing a minor population of genetically corrected cells into the therapeutic range. But strategies for achieving in vivo selection have traditionally relied on genes that confer resistance to cytotoxic drugs and are encumbered by toxicity. Here we describe a new system for in vivo selection that uses a 'cell-growth switch', allowing a minor population of genetically modified cells to be inducibly amplified, thereby averting the risks associated with cytotoxic drugs. This system provides a general platform for conditionally expanding genetically modified cell populations in vivo, and may have widespread applications in gene and cell therapy.


Subject(s)
Cell Separation , Genetic Therapy/methods , Genetic Vectors , Neoplasm Proteins , Receptors, Cytokine , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Blotting, Southern , Bone Marrow Transplantation , Cell Culture Techniques/methods , Dimerization , Dose-Response Relationship, Drug , Erythrocytes/cytology , Erythrocytes/metabolism , Flow Cytometry , Granulocytes/cytology , Granulocytes/metabolism , Green Fluorescent Proteins , Kinetics , Luminescent Proteins/metabolism , Mice , Oncogene Proteins/chemistry , Oncogene Proteins/metabolism , Phenotype , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-kit , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/metabolism , Receptors, Thrombopoietin , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Retroviridae/genetics , Time Factors , Transgenes
14.
J Mol Cell Cardiol ; 52(1): 256-63, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22062955

ABSTRACT

Initial clinical studies indicate a potential beneficial effect of erythropoietin (EPO) in patients with anemia and heart failure. Here, we investigate the direct contractile effects of erythropoietin on myocardial tissue. Treatment with EPO (50U/mL) using excitable murine and human left ventricular muscle preparations resulted in a 37% and 62% increase in twitch tension, respectively (P<0.05). Isolated murine cardiomyocytes exposed to EPO demonstrated a 41% increase in peak sarcomere shortening (P=0.012). Using compounds that specifically stimulate a non-erythropoietic EPO receptor yielded similar increases in contractile dynamics. Cardiomyocyte Ca(2+)dynamics showed an 18% increase in peak calcium in EPO treated cardiomyocytes over controls (P=0.03). Studies in muscle strips skinned after EPO treatment demonstrated a phosphorylation dependant increase in the viscous modulus as well as an increase in oscillatory work. The EPO mediated increase in peak sarcomere shortening was abrogated by PI3-K blockade via wortmannin and by non-isozyme specific PKC blockade by chelerythrine. Finally, EPO treatment resulted in an increase in PKCε in the particulate cellular fraction, indicating activation of this isoform. EPO exhibits direct positive inotropic and lusitropic effects in cardiomyocytes and ventricular muscle preparation. These effects are mediated through PI3-K and PKCε isoform signaling to directly affect both calcium release dynamics and myofilament function.


Subject(s)
Cardiotonic Agents/pharmacology , Erythropoietin/pharmacology , Heart/drug effects , Myocardial Contraction/drug effects , Myocardium/metabolism , Animals , Calcium/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Erythropoietin/metabolism , Humans , In Vitro Techniques , Mice , Myocytes, Cardiac/drug effects , Papillary Muscles/drug effects , Protein Kinase C/metabolism , Receptors, Erythropoietin/agonists , Receptors, Erythropoietin/chemistry , Signal Transduction
15.
Blood ; 116(24): 5357-67, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20826723

ABSTRACT

Erythropoietin (Epo) is required for erythroid progenitor differentiation. Although Epo crosslinking experiments have revealed the presence of Epo receptor (EpoR)-associated proteins that could never be identified, EpoR is considered to be a paradigm for homodimeric cytokine receptors. We purified EpoR-binding partners and identified the type 2 transferrin receptor (TfR2) as a component of the EpoR complex corresponding to proteins previously detected in cross-linking experiments. TfR2 is involved in iron metabolism by regulating hepcidin production in liver cells. We show that TfR2 and EpoR are synchronously coexpressed during the differentiation of erythroid progenitors. TfR2 associates with EpoR in the endoplasmic reticulum and is required for the efficient transport of this receptor to the cell surface. Erythroid progenitors from TfR2(-/-)mice show a decreased sensitivity to Epo and increased circulating Epo levels. In human erythroid progenitors, TfR2 knockdown delays the terminal differentiation. Erythroid cells produce growth differentiation factor-15, a cytokine that suppresses hepatic hepcidin production in certain erythroid diseases such as thalassemia. We show that the production of growth differentiation factor-15 by erythroid cells is dependent on both Epo and TfR2. Taken together, our results show that TfR2 exhibits a non hepatic function as a component of the EpoR complex and is required for efficient erythropoiesis.


Subject(s)
Erythropoiesis , Receptors, Erythropoietin/chemistry , Receptors, Transferrin/physiology , Animals , Cell Differentiation , Gene Expression Regulation , Growth Differentiation Factor 15/biosynthesis , Mice , Mice, Knockout , Multiprotein Complexes/chemistry , Protein Transport , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism
16.
Blood ; 115(4): 896-905, 2010 Jan 28.
Article in English | MEDLINE | ID: mdl-19965681

ABSTRACT

Vascular endothelial growth factor (VEGF) and erythropoietin (EPO) have profound effects on the endothelium and endothelial progenitor cells (EPCs), which originate from the bone marrow and differentiate into endothelial cells. Both EPO and VEGF have demonstrated an ability to increase the number and performance properties of EPCs. EPC behavior is highly dependent on nitric oxide (NO), and both VEGF and EPO can stimulate intracellular NO. EPO can bind to the homodimeric EPO receptor (EPO-R) and the heterodimeric receptor, EPO-R and the common beta receptor (betaC-R). Although VEGF has several receptors, VEGF-R2 appears most critical to EPC function. We demonstrate that EPO induction of NO is dependent on the betaC-R and VEGF-R2, that VEGF induction of NO is dependent on the expression of the betaC-R, and that the betaC-R and VEGF-R2 interact. This is the first definitive functional and structural evidence of an interaction between the 2 receptors and has implications for the side effects of EPO.


Subject(s)
Hematopoietic Stem Cells/metabolism , Nitric Oxide/metabolism , Receptor Cross-Talk/physiology , Receptors, Erythropoietin/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cells, Cultured , Dimerization , Erythropoietin/metabolism , Erythropoietin/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/physiology , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
17.
Nucleic Acids Res ; 38(Web Server issue): W516-22, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20511590

ABSTRACT

The proteins in a cell often assemble into complexes to carry out their functions and play an essential role of biological processes. The PCFamily server identifies template-based homologous protein complexes [called protein complex family (PCF)] and infers functional modules of the query proteins. This server first finds homologous structure complexes of the query using BLASTP to search the structural template database (11,263 complexes). PCFamily then searches the homologous complexes of the templates (query) from a complete genomic database (Integr8 with 6,352,363 protein sequences in 2274 species). According to these homologous complexes across multiple species, this sever infers binding models (e.g. hydrogen-bonds and conserved amino acids in the interfaces), functional modules, and the conserved interacting domains and Gene Ontology annotations of the PCF. Experimental results demonstrate that the PCFamily server can be useful for binding model visualizations and annotating the query proteins. We believe that the server is able to provide valuable insights for determining functional modules of biological networks across multiple species. The PCFamily sever is available at http://pcfamily.life.nctu.edu.tw.


Subject(s)
Multiprotein Complexes/chemistry , Software , Erythropoietin/chemistry , Internet , Multiprotein Complexes/genetics , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Erythropoietin/chemistry , S-Phase Kinase-Associated Proteins/chemistry , Sequence Analysis, Protein , Structural Homology, Protein , User-Computer Interface
18.
Biochim Biophys Acta ; 1806(1): 82-95, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20406667

ABSTRACT

Recombinant human erythropoietin (rhEPO) has been used clinically to alleviate cancer- and chemotherapy-related anemia. However, recent clinical trials have reported that rhEPO also may adversely impact disease progression and survival. The expression of functional EPO receptors (EPOR) has been demonstrated in many human cancer cells where, at least in vitro, rhEPO can stimulate cell growth and survival and may induce resistance to selected therapies. Responses to rhEPO measured by alterations in tumor cell growth or survival, activation of signaling pathways or modulation of sensitivity to anticancer agents are variable. Both methodological and inherent biological issues underlie the differential cell responses, including reported difficulties in EPOR protein detection, potential involvement of EPOR isoforms or of cytoplasmic EPOR, possible differential structure and/or binding affinities of hematopoietic versus non-hematopoietic cell EPOR, possible aberrant regulation of EPOR activity, and a functional EPO/EPOR autocrine/paracrine loop. The modulation by rhEPO of tumor cell response to anticancer agents is coincident with modulation of multiple signaling pathways, BCL-2 family proteins, caspases and NFkB. The molecular interplay of pro-survival and pro-death signals, triggered by EPO and/or by anticancer agents, is multifactorial and tightly coordinated. Expression microarray analysis may prove critical for deciphering this potentially novel network and its broad spectrum of genes and proteins.


Subject(s)
Erythropoietin/physiology , Neoplasms/etiology , Receptors, Erythropoietin/physiology , Animals , Cell Proliferation , Cell Survival , Drug Resistance, Neoplasm , Erythropoietin/chemistry , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Receptors, Erythropoietin/chemistry , Signal Transduction
19.
Anesthesiology ; 115(5): 1084-92, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21873879

ABSTRACT

BACKGROUND: Exogenous erythropoietin inhibits development of allodynia in experimental painful neuropathy because of its antiinflammatory and neuroprotective properties at spinal, supraspinal, and possibly peripheral sites. The authors assess the effect of a nonhematopoietic erythropoietin analog, ARA290, on tactile and cold allodynia in a model of neuropathic pain (spared nerve injury) in rats and mice lacking the ß-common receptor (ßcR mice), a component of the receptor complex mediating tissue protection. METHODS: Twenty-four hours after peripheral nerve injury, rats and mice were injected with ARA290 or vehicle (five 30-µg/kg intraperitoneal injections at 2-day intervals, followed by once/week, n = 8/group). In a separate group of eight rats, ARA290 treatment was restricted to five doses during the initial 2 weeks after surgery. RESULTS: In rats, irrespective of treatment paradigm, ARA290 produced effective, long-term (as long as 15 weeks) relief of tactile and cold allodynia (P < 0.001 vs. vehicle-treated animals). ARA290 was effective in wild-type mice, producing significant relief of allodynia. In contrast, in ßcR mice no effect of ARA290 was observed. CONCLUSIONS: ARA290 produces long-term relief of allodynia because of activation of the ß-common receptor. It is argued that relief of neuropathic pain attributable to ARA290 treatment is related to its antiinflammatory properties, possibly within the central nervous system. Because ARA290, in contrast to erythropoietin, is devoid of hematopoietic and cardiovascular side effects, ARA290 is a promising new drug in the prevention of peripheral nerve injury-induced neuropathic pain in humans.


Subject(s)
Erythropoietin/analogs & derivatives , Neuralgia/drug therapy , Peptides/therapeutic use , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/physiology , Animals , Erythropoietin/chemistry , Female , Hyperalgesia/drug therapy , Mice , Mice, Knockout , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Receptors, Erythropoietin/genetics
20.
Biochem J ; 427(2): 305-12, 2010 Mar 29.
Article in English | MEDLINE | ID: mdl-20136632

ABSTRACT

EPO (erythropoietin), the major hormone regulating erythropoiesis, functions via activation of its cell-surface receptor (EPO-R) present on erythroid progenitor cells. One of the most striking properties of EPO-R is its low expression on the cell surface, as opposed to its high intracellular levels. The low cell-surface expression of EPO-R may thus limit the efficacy of EPO that is routinely used to treat primary and secondary anaemia. In a recent study [Nahari, Barzilay, Hirschberg and Neumann (2008) Biochem. J. 410, 409-416] we have shown that insertion of an NPVY sequence into the intracellular domain of EPO-R increases its cell-surface expression. In the present study we demonstrate that this NPVY EPO-R insert has a selective effect on EPO-mediated downstream signalling in Ba/F3 cells expressing this receptor (NPVY-EPO-R). This is monitored by increased phosphorylation of the NPVY-EPO-R (on Tyr479), Akt, JAK2 (Janus kinase 2) and ERK1/2 (extracellular-signal-regulated kinase 1/2), but not STAT5 (signal transducer and activator of transcription 5), as compared with cells expressing wild-type EPO-R. This enhanced signalling is reflected in augmented proliferation at low EPO levels (0.05 units/ml) and protection against etoposide-induced apoptosis. Increased cell-surface levels of NPVY-EPO-R are most probably not sufficient to mediate these effects as the A234E-EPO-R mutant that is expressed at high cell-surface levels does not confer an augmented response to EPO. Taken together, we demonstrate that insertion of an NPVY sequence into the cytosolic domain of the EPO-R confers not only improved maturation, but also selectively affects EPO-mediated signalling resulting in an improved responsiveness to EPO reflected in cell proliferation and protection against apoptosis.


Subject(s)
Erythropoietin/metabolism , Receptors, Erythropoietin/chemistry , Signal Transduction , Amino Acid Sequence , Animals , Cell Line , Cell Proliferation , Cytosol , Humans , Mice , Mutation , Phosphorylation , Receptors, Erythropoietin/analysis , Receptors, Erythropoietin/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL