Targeted Erythropoietin Selectively Stimulates Red Blood Cell Expansion in Vivo

Total Page:16

File Type:pdf, Size:1020Kb

Targeted Erythropoietin Selectively Stimulates Red Blood Cell Expansion in Vivo Targeted erythropoietin selectively stimulates red blood cell expansion in vivo Devin R. Burrilla, Andyna Verneta, James J. Collinsa,b,c,d, Pamela A. Silvera,e,1, and Jeffrey C. Waya aWyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115; bSynthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139; cInstitute for Medical Engineering & Science, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139; dBroad Institute of MIT and Harvard, Cambridge, MA 02139; and eDepartment of Systems Biology, Harvard Medical School, Boston, MA 02115 Edited by Ronald A. DePinho, University of Texas MD Anderson Cancer Center, Houston, TX, and approved March 30, 2016 (received for review December 23, 2015) The design of cell-targeted protein therapeutics can be informed peptide linker that permits simultaneous binding of both elements by natural protein–protein interactions that use cooperative phys- to the same cell surface. The targeting element anchors the mu- ical contacts to achieve cell type specificity. Here we applied this tated activity element to the desired cell surface (Fig. 1A, Middle), approach in vivo to the anemia drug erythropoietin (EPO), to direct thereby creating a high local concentration and driving receptor its activity to EPO receptors (EPO-Rs) on red blood cell (RBC) pre- binding despite the mutation (Fig. 1A, Bottom). Off-target sig- cursors and prevent interaction with EPO-Rs on nonerythroid cells, naling should be minimal (Fig. 1B) and should decrease in pro- such as platelets. Our engineered EPO molecule was mutated to portion to the mutation strength. weaken its affinity for EPO-R, but its avidity for RBC precursors Here we tested the chimeric activator strategy in vivo using was rescued via tethering to an antibody fragment that specifi- erythropoietin (EPO) as the drug to be targeted. EPO is a cally binds the human RBC marker glycophorin A (huGYPA). We pleiotropic hormone that signals in diverse cell types (13). EPO systematically tested the impact of these engineering steps on promotes the differentiation of RBC precursors, and recombi- in vivo markers of efficacy, side effects, and pharmacokinetics. nant EPO is used to treat anemia due to chronic kidney disease huGYPA transgenic mice dosed with targeted EPO exhibited ele- vated RBC levels, with only minimal platelet effects. This in vivo and myelosuppressive cancer chemotherapy (13); however, EPO selectivity depended on the weakening EPO mutation, fusion to also signals on megakaryocytes, capillary endothelial cells, and the RBC-specific antibody, and expression of huGYPA. The termi- tumor cells, which may promote the thrombosis and tumor nal plasma half-life of targeted EPO was ∼28.3 h in transgenic mice progression that have been documented in clinical trials and vs. ∼15.5 h in nontransgenic mice, indicating that huGYPA on ma- have led to the inclusion of “black box” warnings on EPO-based ture RBCs acted as a significant drug sink but did not inhibit effi- products (13–16). The goal of the present work was to engineer a cacy. In a therapeutic context, our targeting approach may allow form of EPO that, by analogy to CNTF, depends on a non- higher restorative doses of EPO without platelet-mediated side signaling surface marker restricted to RBC precursors and can- effects, and also may improve drug pharmacokinetics. These results not activate cells that may cause EPO side effects. demonstrate how rational drug design can improve in vivo specific- Drug targeting schemes demonstrated in vitro often fail owing ity, with potential application to diverse protein therapeutics. to the demands of in vivo function. Targeted surface receptors might not be restricted to desired tissues; in particular, no known protein engineering | drug targeting | platelet | scFv surface proteins are expressed solely on RBC precursors. In vivo expression of targeted receptors may differ from that on im- n ongoing challenge in therapeutic protein design is speci- mortalized cells used in in vitro experiments. Finally, undesired Aficity of action. The receptor for a protein drug may exist on pharmacokinetics, distribution, and elimination can prevent tar- SCIENCES diverse cell types, resulting in undesired signaling. Numerous geted therapies from reaching desired tissues in adequate levels APPLIED BIOLOGICAL engineering strategies have been used to minimize side effects (17). We addressed these issues by systematically testing the design (1–7). One approach is to tether a protein drug to a cell-specific antibody or antibody fragment. This method can still produce Significance unwanted signaling, however. Even when fused to an antibody, a ligand can still bind to its receptor on cells that cause side effects (8). Erythropoietin is used to treat anemia but has prothrombotic Natural signaling systems often use multicomponent receptor side effects that limit its use. We have demonstrated in vivo the complexes to enable ligands to distinguish between the same ability to target erythropoietin to red blood cell precursors and receptor on two different cells. For example, ciliary neurotrophic away from platelet precursors, thereby potentially avoiding factor (CNTF) first binds a nonsignaling receptor, CNTFRα, off-target effects. We have systematically determined the that is expressed solely by neurons, followed by a second binding protein design features required for in vivo success of the event with the LIFRβ/gp130 signaling receptor complex (9). engineered protein. Our results reveal how rational engineer- CNTF has poor affinity for LIFRβ/gp130 and will activate it only ing of protein drugs can be used to reduce side effects, with on cells that also express CNTFRα. CNTFRα serves to anchor broad implications for designers of therapeutic signaling systems. CNTF on a subset of cells, placing it in a high local concentration β near LIFR and gp130. Thus, CNTF and other cytokines reveal Author contributions: D.R.B. and J.C.W. designed research; D.R.B. and A.V. performed re- that cell specificity can be created through binding to a high- search; D.R.B. and J.C.W. analyzed data; and D.R.B., J.J.C., P.A.S., and J.C.W. wrote the paper. affinity, nonsignaling surface protein that positions the signaling Conflict of interest statement: D.R.B., P.A.S., and J.C.W. are listed as inventors on patent molecule in physical proximity with low-affinity signaling receptors. applications relating to targeted erythropoietin fusion proteins. Earlier work defined a class of engineered proteins, termed This article is a PNAS Direct Submission. “ ” chimeric activators, that can direct signaling to one cell type Data deposition: The sequences reported in this paper have been deposited in the Gen- in vitro (Fig. 1) (10–12). These fusion proteins contain a “tar- Bank database (accession nos. KX026660–KX026663). geting element” (e.g., an antibody fragment) that binds a cell- 1To whom correspondence should be addressed. Email: [email protected]. A Top specific surface marker (Fig. 1 , ) and is tethered to a mutated This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. “activity element” (e.g., a hormone or cytokine) by a flexible 1073/pnas.1525388113/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1525388113 PNAS | May 10, 2016 | vol. 113 | no. 19 | 5245–5250 Downloaded by guest on September 28, 2021 AB We inferred that huGYPA and EPO-R are coexpressed on RBC precursors in vivo (Fig. 1D). Late RBC precursors (BFU-E descendants) express huGYPA approximately 5 d before ma- turing into erythrocytes (24), whereas EPO-R binding events on cultured late-stage precursor cells (CFU-E) can be detected as late as 24 h before the cells mature into reticulocytes or RBCs (25). These results suggest that EPO-R and huGYPA are coex- pressed during late erythropoiesis, although this idea has not been previously addressed directly in vivo. C We tested 10F7-EPOR150A and control variants in huGYPA transgenic mice (26) because 10F7 does not cross-react with murine GYPA (27), and human EPO activates murine EPO-Rs (13). This animal model reflects the normal expression pattern of huGYPA (26). Transgenic RBCs express less huGYPA than D human RBCs (Fig. S2A); 1.6 μM huGYPA is exposed to plasma in transgenic mice vs. 2.8 μM in humans (Fig. S2B). Treatment of huGYPA transgenic mice with 10F7-EPOR150A should stimulate erythropoiesis, and this production should depend on huGYPA expression and a functional 10F7 element. In Vitro Characterization of Chimeric Activator Variants. Our ex- perimental strategy was designed to identify the chimeric acti- vator features required for the desired in vivo behavior: RBC expansion in the absence of platelet production. We compared 10F7-EPOR150A with variants in which EPO was not mutated (10F7-EPO), 10F7 was mutated to mitigate huGYPA binding Fig. 1. Targeting EPO to RBC precursors with a chimeric activator. (A, Top) (10F7W99G-EPOR150A),andEPOwasmutatedtoeliminateEPO-R Chimeric activator schematic illustrating a targeting element (light blue) affinity (10F7-EPOK45D). Darbepoetin alfa (Aranesp) was used tethered by a linker (black circles) to a mutated activity element (red, black A = as a nontargeted form of EPO (28). Fig. 2 shows engineered X mutation). The chimeric activator initially binds (bold arrow) a target protein schematics and verification of their size and N-linked cell receptor (dark blue) via the targeting element, whereas the mutated activity element has a low affinity (dashed arrow) for its own receptor (pink). glycosylation. (A, Middle) Increased local concentration of the activity element overcomes We quantified the effect of the R150A mutation on the in- its receptor-binding deficit and facilitates interaction (bold arrow). (A, Bottom) teraction between EPO and EPO-R (Fig. 2B). EPO and 10F7- This interaction results in signaling via the activity receptor (bold arrow). (B)On EPO had similar binding kinetics (KD = 5.4 and 7.2 nM, re- off-target cells lacking a receptor for the targeting element, the activity ele- spectively) in accordance with published values (29), indicating ment has little effect (dashed arrow), owing to its mutation.
Recommended publications
  • Induction of Erythropoietin Increases the Cell Proliferation Rate in a Hypoxia‑Inducible Factor‑1‑Dependent and ‑Independent Manner in Renal Cell Carcinoma Cell Lines
    ONCOLOGY LETTERS 5: 1765-1770, 2013 Induction of erythropoietin increases the cell proliferation rate in a hypoxia‑inducible factor‑1‑dependent and ‑independent manner in renal cell carcinoma cell lines YUTAKA FUJISUE1, TAKATOSHI NAKAGAWA2, KIYOSHI TAKAHARA1, TERUO INAMOTO1, SATOSHI KIYAMA1, HARUHITO AZUMA1 and MICHIO ASAHI2 Departments of 1Urology and 2Pharmacology, Faculty of Medicine, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan Received November 28, 2012; Accepted February 25, 2013 DOI: 10.3892/ol.2013.1283 Abstract. Erythropoietin (Epo) is a potent inducer of erythro- Introduction poiesis that is mainly produced in the kidney. Epo is expressed not only in the normal kidney, but also in renal cell carcinomas Erythropoietin (Epo) is a 30-kDa glycoprotein that functions (RCCs). The aim of the present study was to gain insights as an important cytokine in erythrocytes. Epo is usually into the roles of Epo and its receptor (EpoR) in RCC cells. produced by stromal cells of the adult kidney cortex or fetal The study used two RCC cell lines, Caki-1 and SKRC44, in liver and then released into the blood, with its production which Epo and EpoR are known to be highly expressed. The initially induced by hypoxia or hypotension (1-5). In the bone proliferation rate and expression level of hypoxia-inducible marrow, Epo binds to the erythropoietin receptor (EpoR) factor-1α (HIF-1α) were measured prior to and following expressed in erythroid progenitor cells or undifferentiated Epo treatment and under normoxic and hypoxic conditions. erythroblasts, which induces signal transduction mechanisms To examine whether HIF-1α or Epo were involved in cellular that protect the undifferentiated erythrocytes from apoptosis proliferation during hypoxia, these proteins were knocked and promote their proliferation and differentiation.
    [Show full text]
  • Immunohistochemical Expression of Erythropoietin in Invasive Breast Carcinoma with Metastasis to Lymph Nodes
    Review and Research on Cancer Treatment Volume 4, Issue 1 (2018) ISSN 2544-2147 Immunohistochemical expression of erythropoietin in invasive breast carcinoma with metastasis to lymph nodes Maksimiuk M.* Students' Scientific Organization at the Medical University of Warsaw, Oczki 1a, 02-007 Warsaw Sobiborowicz A. Students' Scientific Organization at the Medical University of Warsaw, Oczki 1a, 02-007 Warsaw Sobieraj M. Students' Scientific Organization at the Medical University of Warsaw, Oczki 1a, 02-007 Warsaw Liszcz A. Students' Scientific Organization at the Medical University of Warsaw, Oczki 1a, 02-007 Warsaw Sobol M. Department of Biophysics and Human Physiology, Medical University of Warsaw, Chałubińskiego 5, 02-004 Warsaw Patera J. Department of Pathomorphology, Military Institute of Health Services, Warsaw, Poland Badowska-Kozakiewicz A.M. Department of Biophysics and Human Physiology, Medical University of Warsaw, Chałubińskiego 5, 02-004 Warsaw *corresponding author: Marta Maksimiuk, Department of Biophysics and Human Physiology, Medical University of Warsaw, Chałubińskiego 5, 02-004 Warsaw, [email protected], tel: 691 – 230 – 268 Abstract: Introduction: Tumor characteristics, such as size, lymph node status, histological type of the neoplasm and its grade, are well known prognostic factors in breast cancer. The ongoing search for new prognostic factors include Bcl2, Bax, Cox-2 or HIF1- alpha, which plays a key role in phenomenon of tumor hypoxia and might induce transcription of the EPO gene. Erythropoietin may influence lymph node metastasis or stimulate tumor progression, thus it seemed interesting to determine its expression in invasive breast cancers with lymph node metastases presenting different basic immunohistochemical profiles (ER, PR, HER2). Aim: To evaluate the relationship between histological grade, tumor size, lymph node status, expression of ER, PR, HER2 and immunohistochemical expression of erythropoietin in invasive breast cancer with metastasis to lymph nodes.
    [Show full text]
  • The Thrombopoietin Receptor : Revisiting the Master Regulator of Platelet Production
    This is a repository copy of The thrombopoietin receptor : revisiting the master regulator of platelet production. White Rose Research Online URL for this paper: https://eprints.whiterose.ac.uk/175234/ Version: Published Version Article: Hitchcock, Ian S orcid.org/0000-0001-7170-6703, Hafer, Maximillian, Sangkhae, Veena et al. (1 more author) (2021) The thrombopoietin receptor : revisiting the master regulator of platelet production. Platelets. pp. 1-9. ISSN 0953-7104 https://doi.org/10.1080/09537104.2021.1925102 Reuse This article is distributed under the terms of the Creative Commons Attribution (CC BY) licence. This licence allows you to distribute, remix, tweak, and build upon the work, even commercially, as long as you credit the authors for the original work. More information and the full terms of the licence here: https://creativecommons.org/licenses/ Takedown If you consider content in White Rose Research Online to be in breach of UK law, please notify us by emailing [email protected] including the URL of the record and the reason for the withdrawal request. [email protected] https://eprints.whiterose.ac.uk/ Platelets ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/iplt20 The thrombopoietin receptor: revisiting the master regulator of platelet production Ian S. Hitchcock, Maximillian Hafer, Veena Sangkhae & Julie A. Tucker To cite this article: Ian S. Hitchcock, Maximillian Hafer, Veena Sangkhae & Julie A. Tucker (2021): The thrombopoietin receptor: revisiting the master regulator of platelet production, Platelets, DOI: 10.1080/09537104.2021.1925102 To link to this article: https://doi.org/10.1080/09537104.2021.1925102 © 2021 The Author(s).
    [Show full text]
  • As a Key Regulator of Erythropoiesis, Bone Remodeling and Endothelial
    cells Review Erythropoietin (EPO) as a Key Regulator of Erythropoiesis, Bone Remodeling and Endothelial Transdifferentiation of Multipotent Mesenchymal Stem Cells (MSCs): Implications in Regenerative Medicine Asterios S. Tsiftsoglou Laboratory of Pharmacology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; [email protected] Abstract: Human erythropoietin (EPO) is an N-linked glycoprotein consisting of 166 aa that is pro- duced in the kidney during the adult life and acts both as a peptide hormone and hematopoietic growth factor (HGF), stimulating bone marrow erythropoiesis. EPO production is activated by hypoxia and is regulated via an oxygen-sensitive feedback loop. EPO acts via its homodimeric erythropoietin receptor (EPO-R) that increases cell survival and drives the terminal erythroid mat- uration of progenitors BFU-Es and CFU-Es to billions of mature RBCs. This pathway involves the activation of multiple erythroid transcription factors, such as GATA1, FOG1, TAL-1, EKLF and BCL11A, and leads to the overexpression of genes encoding enzymes involved in heme biosynthesis Citation: Tsiftsoglou, A.S. and the production of hemoglobin. The detection of a heterodimeric complex of EPO-R (consist- Erythropoietin (EPO) as a Key ing of one EPO-R chain and the CSF2RB β-chain, CD131) in several tissues (brain, heart, skeletal Regulator of Erythropoiesis, Bone muscle) explains the EPO pleotropic action as a protection factor for several cells, including the Remodeling and Endothelial multipotent MSCs as well as cells modulating the innate and adaptive immunity arms. EPO induces Transdifferentiation of Multipotent the osteogenic and endothelial transdifferentiation of the multipotent MSCs via the activation of Mesenchymal Stem Cells (MSCs): EPO-R signaling pathways, leading to bone remodeling, induction of angiogenesis and secretion Implications in Regenerative of a large number of trophic factors (secretome).
    [Show full text]
  • Correlation with Vasculogenic Mimicry and Poor Prognosis
    Int J Clin Exp Pathol 2015;8(4):4033-4043 www.ijcep.com /ISSN:1936-2625/IJCEP0006586 Original Article Erythropoietin and erythropoietin receptor in hepatocellular carcinoma: correlation with vasculogenic mimicry and poor prognosis Zhihong Yang1,2*, Baocun Sun1,2,3*, Xiulan Zhao1,2*, Bing Shao1, Jindan An1, Qiang Gu1,2, Yong Wang1, Xueyi Dong1, Yanhui Zhang1,3, Zhiqiang Qiu1,3 1Department of Pathology, Tianjin Medical University, Tianjin 300070, China; 2Department of Pathology, General Hospital of Tianjin Medical University, Tianjin 300052, China; 3Department of Pathology, Cancer Hospital of Tianjin Medical University, Tianjin 300060, China. *Equal contributors. Received February 1, 2015; Accepted March 30, 2015; Epub April 1, 2015; Published April 15, 2015 Abstract: To evaluate erythropoietin (Epo) and erythropoietin receptor (EpoR) expression, its relationship with vas- culogenic mimicry (VM) and its prognostic value in human hepatocellular carcinoma (HCC), we examined Epo/EpoR expression and VM formation using immunohistochemistry and CD31/PAS (periodic acid-Schiff) double staining on 92 HCC specimens. The correlation between Epo/EpoR expression and VM formation was analyzed using two-tailed Chi-square test and Spearman correlation analysis. Survival curves were generated using Kaplan-Meier method. Multivariate analysis was performed using Cox regression model to assess the prognostic values. Results showed positive correlation between Epo/EpoR expression and VM formation (P < 0.05). Patients with Epo or EpoR expres- sion exhibited poorer overall survival (OS) than Epo-negative or EpoR-negative patients (P < 0.05). Epo-positive/VM- positive and EpoR-positive/VM-positive patients had the worst OS (P < 0.05). In multivariate survival analysis, age, Epo and EpoR were independent prognostic factors related to OS.
    [Show full text]
  • Investigation of the Protective Effect of Erythropoietin on Spinal Cord Injury in Rats
    EXPERIMENTAL AND THERAPEUTIC MEDICINE 2: 837-841, 2011 Investigation of the protective effect of erythropoietin on spinal cord injury in rats ZHENGHUA HONG, HUAXING HONG, HAIXIAO CHEN, ZHANGFU WANG and DUN HONG Department of Orthopedics, Taizhou Hospital of Zhejiang Province, Taizhou 317000, P.R. China Received May 2, 2011; Accepted June 6, 2011 DOI: 10.3892/etm.2011.285 Abstract. Erythropoietin (EPO) is a promising therapeutic cell apoptosis occurs at the lesion site, affecting both neurons agent used in a variety of spinal cord injuries. Therefore, iden- and astrocytes (1,2) It has long been believed that the central tifying the specific molecular pathway mediating the neuronal nervous system (CNS) is incapable of regenerating itself after protective effect of EPO after spinal cord injury (SCI) is of injury and during the repair process after SCI. Thus, an ideal great value to the patients concerned. Platelet-derived growth treatment would be one administered systemically but without factor (PDGF)-B is an important factor in the recovery of significant side effects. Erythropoietin( EPO), a potent inhibitor neurological function. We explored changes in the expression of apoptosis and a promising therapeutic agent, has been used of PDGF-B in spinal cord injury rats after receiving EPO treat- in a variety of neurological insults, including traumatic brain ment. We used a weight-drop contusion SCI model, and EPO injury, brain stroke and spinal cord injury (3,4). Therefore, treatment group rats received single doses ofEPO (1,000 U/ identifying the specific molecular pathway mediating the EPO kg i.p.) immediately after the operation. Seven days after the neuronal protective effects of EPO after SCI is of great value operation, the results revealed a more rapid recovery as noted to the patients concerned.
    [Show full text]
  • Erythropoietin and Cancer: the Unintended Consequences of Anemia Correction
    REVIEW ARTICLE published: 11 November 2014 doi: 10.3389/fimmu.2014.00563 Erythropoietin and cancer: the unintended consequences of anemia correction Nataša Debeljak 1*, Peter Solár 2 and Arthur J. Sytkowski 3 1 Faculty of Medicine, Institute of Biochemistry, University of Ljubljana, Ljubljana, Slovenia 2 Department of Cell and Molecular Biology, Institute of Biology and Ecology, Faculty of Sciences, Pavol Jozef Šafárik University, Košice, Slovakia 3 Oncology Therapeutic Area, Quintiles Transnational, Arlington, MA, USA Edited by: Until 1990, erythropoietin (EPO) was considered to have a single biological purpose and Pietro Ghezzi, Brighton and Sussex action, the stimulation of red blood cell growth and differentiation. Slowly, scientific and Medical School, UK medical opinion evolved, beginning with the discovery of an effect on endothelial cell Reviewed by: Ralf Schindler, Charité, Germany growth in vitro and the identification of EPO receptors (EPORs) on neuronal cells. We Domenico Ribatti, University of Bari now know that EPO is a pleiotropic growth factor that exhibits an anti-apoptotic action on Medical School, Italy numerous cells and tissues, including malignant ones. In this article, we present a short Chris Thiemermann, Queen Mary discussion of EPO, receptors involved in EPO signal transduction, and their action on non- University of London, UK hematopoietic cells. This is followed by a more detailed presentation of both pre-clinical *Correspondence: Nataša Debeljak, Faculty of Medicine, and clinical data that demonstrate EPO’s action on cancer cells, as well as tumor angio- Institute of Biochemistry, University genesis and lymphangiogenesis. Clinical trials with reported adverse effects of chronic of Ljubljana, Vrazov trg 2, SI-1000 erythropoiesis-stimulating agents (ESAs) treatment as well as clinical studies exploring Ljubljana, Slovenia the prognostic significance of EPO and EPOR expression in cancer patients are reviewed.
    [Show full text]
  • Red Blood Cell Microrheological Effects of Some Antitumor Chemotherapy Drugs: in Vitro Study
    Journal of Cellular Biotechnology 1 (2015/2016) 151–158 151 DOI 10.3233/JCB-15015 IOS Press Red blood cell microrheological effects of some antitumor chemotherapy drugs: In vitro study A.V. Muravyova,∗, I.A. Tikhomirovaa, N.V. Kislovb and A.S. Petrochenkoa aState Pedagogical University, Yaroslavl, Russia bRegional Oncological Hospital, Yaroslavl, Russia Abstract. In consequence of limitation of capillary dilatation reserve the role of erythrocyte membrane elasticity and their microrheological properties is very important for the tissue perfusion. The drugs that enter blood flow could be as signaling molecules and their action can lead to alteration of red blood cell aggregation and whole cell deformability. Therefore the subject of this study was to estimate the effect of some antitumor chemotherapy drugs on red blood cell aggregation (RBCA) and deformability (RBCD). It was found that an exposure of red blood cells (RBCs) to cisplatin and epoetin alfa led to significant positive changes in the RBC microrheological properties. It was shown that cisplatin could activate tyrosine protein kinase (TPK). Preincubation of RBCs with the inhibitor of EGF-R and Src kinase lavendustin A was accompanied by a significant decrease of the cisplatin effect. It was found RBC aggregation rise after cell incubation with 5-fluorouracil. This effect was removed with Ca2+ chelation with EGTA and also with pentoxifylline. The drug for the targeted antitumor therapy sunitinib raised RBCA markedly. Whereas RBC deformability index was altered only slightly. The combination of two chemicals: sunitinib and cisplatin led to an elimination of proaggregative effect of sunitinib. Examination of the effects of some antitumor drugs on red blood cell microrheological properties has enabled us to suggest that the observed changes in red cell aggregation and deformability may be partly related to the effects of tyrosine protein kinase activation.
    [Show full text]
  • Inactivation of Erythropoietin Receptor Function by Point Mutations in a Region Having Homology with Other Cytokine Receptors OSAMU MIURA,'T JOHN L
    MOLECULAR AND CELLULAR BIOLOGY, Mar. 1993, p. 1788-1795 Vol. 13, No. 3 0270-7306/93/031788-08$02.00/0 Copyright © 1993, American Society for Microbiology Inactivation of Erythropoietin Receptor Function by Point Mutations in a Region Having Homology with Other Cytokine Receptors OSAMU MIURA,'t JOHN L. CLEVELAND,' AND JAMES N. IHLEl,2* Department ofBiochemistry, St. Jude Children's Research Hospital, Memphis, Tennessee 31051,1 and Department ofBiochemistry, University of Tennessee, Memphis, Tennessee 3816332 Received 13 July 1992/Returned for modification 12 August 1992/Accepted 21 December 1992 The cytoplasmic domain of the erythropoietin receptor (EpoR) contains a region, proximal to the transmembrane domain, that is essential for function and has homology with other members of the cytokine receptor family. To explore the functional significance of this region and to identify critical residues, we introduced several amino acid substitutions and examined their effects on erythropoietin-induced mitogenesis, tyrosine phosphorylation, and expression of immediate-early (c-fos, c-myc, and egr-1) and early (ornithine decarboxylase and T-cell receptor 'y) genes in interleukin-3-dependent cell lines. Amino acid substitution of W-282, which is strictly conserved at the middle portion of the homology region, completely abolished all the functions of the EpoR. Point mutation at L-306 or E-307, both of which are in a conserved LEVL motif, drastically impaired the function of the receptor in all assays. Other point mutations, introduced into less conserved amino acid residues, did not significantly impair the function of the receptor. These results demonstrate that conserved amino acid residues in this domain of the EpoR are required for mitogenesis, stimulation of tyrosine phosphorylation, and induction of immediate-early and early genes.
    [Show full text]
  • Erythropoietin Promotes Breast Tumorigenesis Through Tumor-Initiating Cell Self-Renewal Bing Zhou,1 Jeffrey S
    Downloaded on January 20, 2014. The Journal of Clinical Investigation. More information at www.jci.org/articles/view/69804 Research article Erythropoietin promotes breast tumorigenesis through tumor-initiating cell self-renewal Bing Zhou,1 Jeffrey S. Damrauer,1 Sean T. Bailey,1 Tanja Hadzic,1 Youngtae Jeong,2 Kelly Clark,1 Cheng Fan,1 Laura Murphy,1 Cleo Y. Lee,2 Melissa A. Troester,3 C. Ryan Miller,4 Jian Jin,1,5 David Darr,1 Charles M. Perou,1 Ross L. Levine,6 Maximilian Diehn,2 and William Y. Kim1 1Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA. 2Department of Radiation Oncology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA. 3Department of Epidemiology and 4Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA. 5Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA. 6Human Oncology and Pathogenesis Program, Leukemia Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA. Erythropoietin (EPO) is a hormone that induces red blood cell production. In its recombinant form, EPO is the one of most prescribed drugs to treat anemia, including that arising in cancer patients. In randomized trials, EPO administration to cancer patients has been associated with decreased survival. Here, we investi- gated the impact of EPO modulation on tumorigenesis. Using genetically engineered mouse models of breast cancer, we found that EPO promoted tumorigenesis by activating JAK/STAT signaling in breast tumor-initi- ating cells (TICs) and promoted TIC self renewal.
    [Show full text]
  • Erythropoietin and Growth Factors Exhibit Differential Angiogenic Potential in Mouse Heart
    in vivo 22 : 587-592 (2008) Erythropoietin and Growth Factors Exhibit Differential Angiogenic Potential in Mouse Heart OLGA PAGONOPOULOU 1, ANNA EFTHIMIADOU 1, MARIA LAMBROPOULOU 2, NIKOS PAPADOPOULOS 2 and NIKOS K. NIKOLETTOS 1 Departments of 1Physiology and 2Histology – Embryology, Democritus University of Thrace, Medical School, Alexandroupolis, 68100, Greece Abstract. Background: The possible angiogenic effect of functions of EPO, angiogenesis, the process through which recombinant human erythropoietin (rHuEpo) and several new blood vessels arise from preexisting ones, has been possibly angiogenic cytokines such as basic fibroblast growth indicated (5). The potential role of EPO in angiogenesis may factor (bFGF), acidic fibroblast growth factor (aFGF) and be considered as a subset of its possible function in improving vascular endothelial growth factor (VEGF) was investigated overall tissue oxygenation and its antiapoptotic role (5). in mouse heart. Materials and Methods: Mice were divided Recombinant human erythropoietin (rHuEPO) induces a into five groups (n=7/group): A, control; B, rHuEpo-treated; proangiogenic phenotype in human endothelial cells, including C, (aFGF-treated); D, (VEGF-treated); E, (bFGF-treated). both early ( i.e. increase in cell proliferation and matrix The antibody mouse anti-human CD31 was used to evaluate metalloproteinase-2 production) and late (differentiation into the vessels present in histological preparations. Results: The vascular tubes) angiogenic events. In vivo , in the chick embryo results show a significant increase of the vessel number per CAM assay, the angiogenic activity of rHuEPO was similar to optical field in the rHuEpo-treated, the bFGF-treated and that exerted by basic fibroblast growth factor (bFGF), a well- the VEGF-treated animals compared to controls whereas known angiogenic cytokine and endothelial cells of the CAM aFGF did not show a ny significant angiogenic activity.
    [Show full text]
  • Leukemia Inhibitory Factor Inhibits Erythropoietin-Induced Myelin Gene
    Gyetvai et al. Molecular Medicine (2018) 24:51 Molecular Medicine https://doi.org/10.1186/s10020-018-0052-3 RESEARCHARTICLE Open Access Leukemia inhibitory factor inhibits erythropoietin-induced myelin gene expression in oligodendrocytes Georgina Gyetvai, Cieron Roe, Lamia Heikal, Pietro Ghezzi* and Manuela Mengozzi Abstract Background: The pro-myelinating effects of leukemia inhibitory factor (LIF) and other cytokines of the gp130 family, including oncostatin M (OSM) and ciliary neurotrophic factor (CNTF), have long been known, but controversial results have also been reported. We recently overexpressed erythropoietin receptor (EPOR) in rat central glia-4 (CG4) oligodendrocyte progenitor cells (OPCs) to study the mechanisms mediating the pro-myelinating effects of erythropoietin (EPO). In this study, we investigated the effect of co-treatment with EPO and LIF. Methods: Gene expression in undifferentiated and differentiating CG4 cells in response to EPO and LIF was analysed by DNA microarrays and by RT-qPCR. Experiments were performed in biological replicates of N ≥ 4. Functional annotation and biological term enrichment was performed using DAVID (Database for Annotation, Visualization and Integrated Discovery). The gene-gene interaction network was visualised using STRING (Search Tool for the Retrieval of Interacting Genes). Results: In CG4 cells treated with 10 ng/ml of EPO and 10 ng/ml of LIF, EPO-induced myelin oligodendrocyte glycoprotein (MOG) expression, measured at day 3 of differentiation, was inhibited ≥4-fold (N =5,P < 0.001). Inhibition of EPO-induced MOG was also observed with OSM and CNTF. Analysis of the gene expression profile of CG4 differentiating cells treated for 20 h with EPO and LIF revealed LIF inhibition of EPO-induced genes involved in lipid transport and metabolism, previously identified as positive regulators of myelination in this system.
    [Show full text]