1
|
Xu Y, Wang B, Zhang M, Zhang J, Li Y, Jia P, Zhang H, Duan L, Li Y, Li Y, Qu X, Wang S, Liu D, Zhou W, Zhao H, Zhang H, Chen L, An X, Lu S, Zhang S. Carbon Dots as a Potential Therapeutic Agent for the Treatment of Cancer-Related Anemia. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2200905. [PMID: 35294781 DOI: 10.1002/adma.202200905] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/14/2022] [Indexed: 06/14/2023]
Abstract
Due to the adverse effects of erythropoietin (EPO) on cancer patient survival, it is necessary to develop new agents that can be used to efficiently manage and treat cancer-related anemia. In this study, novel distinctive carbon dots, J-CDs, derived from jujube are designed, synthesized, and characterized. Based on the obtained results, this material comprises sp2 and sp3 carbon atoms, as well as oxygen/nitrogen-based groups, and it specifically promotes the proliferation of erythroid cells by stimulating the self-renewal of erythroid progenitor cells in vitro and in vivo. Moreover, J-CDs have no discernible effects on tumor proliferation and metastasis, unlike EPO. Transcriptome profiling suggests that J-CDs upregulate the molecules involved in hypoxia response, and they also significantly increase the phosphorylation levels of STAT5, the major transducer of signals for erythroid progenitor cell proliferation. Overall, this study demonstrates that J-CDs effectively promote erythrocyte production without affecting tumor proliferation and metastasis; thus, they may be promising agents for the treatment of cancer-related anemia.
Collapse
Affiliation(s)
- Yuanlin Xu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 45001, China
| | - Boyang Wang
- College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China
| | - Mingming Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jingxin Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yudong Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Peijun Jia
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Huan Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Laboratory of Membrane Biology, New York Blood Center, New York, NY, 10065, USA
| | - Lulu Duan
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yan Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yating Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaoli Qu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Shihui Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Donghao Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenping Zhou
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 45001, China
| | - Huizhi Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hengchao Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Lixiang Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY, 10065, USA
| | - Siyu Lu
- College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China
| | - Shijie Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
2
|
The Role of PI3K/AKT and MAPK Signaling Pathways in Erythropoietin Signalization. Int J Mol Sci 2021; 22:ijms22147682. [PMID: 34299300 PMCID: PMC8307237 DOI: 10.3390/ijms22147682] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 12/11/2022] Open
Abstract
Erythropoietin (EPO) is a glycoprotein cytokine known for its pleiotropic effects on various types of cells and tissues. EPO and its receptor EPOR trigger signaling cascades JAK2/STAT5, MAPK, and PI3K/AKT that are interconnected and irreplaceable for cell survival. In this article, we describe the role of the MAPK and PI3K/AKT signaling pathways during red blood cell formation as well as in non-hematopoietic tissues and tumor cells. Although the central framework of these pathways is similar for most of cell types, there are some stage-specific, tissue, and cell-lineage differences. We summarize the current state of research in this field, highlight the novel members of EPO-induced PI3K and MAPK signaling, and in this respect also the differences between erythroid and non-erythroid cells.
Collapse
|
3
|
Fibronectin 1: A Potential Biomarker for Ovarian Cancer. DISEASE MARKERS 2021; 2021:5561651. [PMID: 34093898 PMCID: PMC8164534 DOI: 10.1155/2021/5561651] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 11/17/2022]
Abstract
Methods OVCAR3 and A2780 are the two common cell lines that are used for ovarian cancer studies. The different invasion and migration abilities were observed by scratch tests and transwell experiments in our preliminary study. Gene chip was used to screen the expression gene in these two different cell lines, and then, the differentially expressed genes (at least 2-fold difference, P value < 0.05) were analyzed using KEGG. Result Fibronectin 1 (FN1) was found to be the most strongly correlated with the invasion and migration abilities of the OVCAR3 cells. Real-time PCR and FN1 knockout cell line was conducted and confirmed this finding. Based on the Oncomine database analysis, comparing with normal people, ovarian cancer patients exhibited high levels of FN1 expression. Additionally, higher FN1 expression was found in patients with higher FIGO stages of cancer. Conclusion FN1 could be a new biomarker for ovarian cancer detection and progress indicator.
Collapse
|
4
|
Lee J, Vernet A, Redfield K, Lu S, Ghiran IC, Way JC, Silver PA. Rational Design of a Bifunctional AND-Gate Ligand To Modulate Cell-Cell Interactions. ACS Synth Biol 2020; 9:191-197. [PMID: 31834794 DOI: 10.1021/acssynbio.9b00273] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Protein "AND-gate" systems, in which a ligand acts only on cells with two different receptors, direct signaling activity to a particular cell type and avoid action on other cells. In a bifunctional AND-gate protein, the molecular geometry of the protein domains is crucial. Here we constructed a tissue-targeted erythropoietin (EPO) that stimulates red blood cell (RBC) production without triggering thrombosis. The EPO was directed to RBC precursors and mature RBCs by fusion to an anti-glycophorin A antibody V region. Many such constructs activated EPO receptors in vitro and stimulated RBC and not platelet production in mice but nonetheless enhanced thrombosis in mice and caused adhesion between RBCs and EPO-receptor-bearing cells. On the basis of a protein-structural model of the RBC surface, we rationally designed an anti-glycophorin-EPO fusion that does not induce cell adhesion in vitro or enhance thrombosis in vivo. Thus, mesoscale geometry can inform the design of synthetic-biological systems.
Collapse
Affiliation(s)
- Jungmin Lee
- Department of Chemistry and Chemical Biology , Harvard University , Cambridge , Massachusetts 02138 , United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston , Massachusetts 02115 , United States
| | - Andyna Vernet
- Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston , Massachusetts 02115 , United States
| | - Katherine Redfield
- Harvard-MIT Division of Health Sciences and Technology , Cambridge , Massachusetts 02139 , United States
| | - Shulin Lu
- Department of Medicine, Beth Israel Deaconess Medical Center , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Ionita C Ghiran
- Department of Medicine, Beth Israel Deaconess Medical Center , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Jeffrey C Way
- Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston , Massachusetts 02115 , United States
| | - Pamela A Silver
- Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston , Massachusetts 02115 , United States
- Department of Systems Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| |
Collapse
|
5
|
Fecková B, Kimáková P, Ilkovičová L, Szentpéteriová E, Macejová M, Košuth J, Zulli A, Debeljak N, Hudler P, Jašek K, Kašubová I, Kubatka P, Solár P. Methylation of the first exon in the erythropoietin receptor gene does not correlate with its mRNA and protein level in cancer cells. BMC Genet 2019; 20:1. [PMID: 30606107 PMCID: PMC6318971 DOI: 10.1186/s12863-018-0706-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/13/2018] [Indexed: 01/13/2023] Open
Abstract
Background Erythropoietin receptor (EPOR) is a functional membrane-bound cytokine receptor. Erythropoietin (EPO) represents an important hematopoietic factor for production, maturation and differentiation of erythroid progenitors. In non-hematopoietic tissue, EPO/EPOR signalization could also play cytoprotective and anti-apoptotic role. Several studies identified pro-stimulating EPO/EPOR effects in tumor cells; however, numerous studies opposed this fact due to the usage of unspecific EPOR antibodies and thus potential absence or very low levels of EPOR in tumor cells. It seems that this problem is more complex and therefore we have decided to focus on EPOR expression at several levels such as the role of methylation in the regulation of EPOR expression, identification of possible EPOR transcripts and the presence of EPOR protein in selected tumor cells. Methods Methylation status was analysed by bisulfite conversion reaction, PCR and sequencing. The expression of EPOR was monitored by quantitative RT-PCR and western blot analysis. Results In this study we investigated the methylation status of exon 1 of EPOR gene in selected human cancer cell lines. Our results indicated that CpGs methylation in exon 1 do not play a significant role in the regulation of EPOR transcription. However, methylation status of EPOR exon 1 was cell type dependent. We also observed the existence of two EPOR splice variants in human ovarian adenocarcinoma cell line - A2780 and confirmed the expression of EPOR protein in these cells using specific A82 anti-EPOR antibody. Conclusion We outlined the methylation status of all selected cancer cell lines in exon 1 of EPOR gene and these results could benefit future investigations. Moreover, A82 antibody confirmed our previous results demonstrating the presence of functional EPOR in human ovarian adenocarcinoma A2780 cells.
Collapse
Affiliation(s)
- Barbora Fecková
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, SK-04154, Košice, Slovak Republic
| | - Patrícia Kimáková
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, SK-04154, Košice, Slovak Republic
| | - Lenka Ilkovičová
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, SK-04154, Košice, Slovak Republic
| | - Erika Szentpéteriová
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, SK-04154, Košice, Slovak Republic
| | - Mária Macejová
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, SK-04154, Košice, Slovak Republic
| | - Ján Košuth
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, SK-04154, Košice, Slovak Republic
| | - Anthony Zulli
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Nataša Debeljak
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, SI1000, Ljubljana, Slovenia
| | - Petra Hudler
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, SI1000, Ljubljana, Slovenia
| | - Karin Jašek
- Biomedical Centre Martin, Division of Oncology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK03601, Martin, Slovak Republic
| | - Ivana Kašubová
- Biomedical Centre Martin, Division of Oncology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK03601, Martin, Slovak Republic
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK03601, Martin, Slovak Republic.,Department of Experimental Carcinogenesis, Biomedical Centre Martin, Division of Oncology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK03601, Martin, Slovak Republic
| | - Peter Solár
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, SK-04154, Košice, Slovak Republic. .,Institute of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, SK04011, Košice, Slovak Republic.
| |
Collapse
|
6
|
Annese T, Tamma R, Ruggieri S, Ribatti D. Erythropoietin in tumor angiogenesis. Exp Cell Res 2019; 374:266-273. [DOI: 10.1016/j.yexcr.2018.12.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/11/2018] [Accepted: 12/16/2018] [Indexed: 12/19/2022]
|
7
|
Frille A, Leithner K, Olschewski A, Olschewski H, Wohlkönig C, Hrzenjak A. No erythropoietin-induced growth is observed in non-small cell lung cancer cells. Int J Oncol 2017; 52:518-526. [PMID: 29345289 DOI: 10.3892/ijo.2017.4225] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/21/2017] [Indexed: 11/06/2022] Open
Abstract
Lung cancer patients have the highest incidence of anemia among patients with solid tumors. The use of recombinant human erythropoietin (Epo) has consistently been shown to reduce the need for blood transfusions and to increase hemoglobin levels in lung cancer patients with chemotherapy-induced anemia. However, clinical and preclinical studies have prompted concerns that Epo and the presence of its receptor, EpoR, in tumor cells may be responsible for adverse effects and, eventually, death. The question has been raised whether Epo promotes tumor growth and inhibits the death of cancer cells. In this study, we investigated the presence and functionality of EpoR, as well as the implications of Epo upon the proliferation and survival of lung cancer cells. Since the protein expression of both Epo and EpoR is induced by hypoxia, which is frequently present in lung cancer, the cells were treated with Epo under both normoxic and hypoxic conditions (1% O2). By using quantitative (real-time) PCR, western blot analysis, and immunocytochemical staining, three non-small cell lung cancer (NSCLC) cell lines (A427, A549 and NCI-H358) were analyzed for the expression of EpoR and its specific downstream signaling pathways [Janus kinase 2 (Jak2)-signal transducer and activator of transcription 5 (STAT5), phosphatidylinositol-3-kinase (PI3K)-Akt, mitogen-activated protein (MAP) kinase]. The effects of 100 U/ml Epo on cell proliferation and cisplatin-induced apoptosis were assessed. All NSCLC cell lines expressed EpoR mRNA and protein, while these levels differed considerably between the cell lines. We found the constitutive phosphorylation of EpoR and most of its downstream signaling pathways (STAT5, Akt and ERK1/2) independently of Epo administration. While Epo markedly enhanced the proliferation and reduced apoptosis of Epo-dependent UT-7/Epo leukemia cells, it did not affect tumor cell proliferation or the cisplatin-induced apoptosis of NSCLC cells. Thus, this in vitro study suggests that there are no tumor-promoting effects of Epo in the NSCLC cell lines studied, neither under normoxic nor under hypoxic conditions.
Collapse
Affiliation(s)
- Armin Frille
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Katharina Leithner
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria
| | - Horst Olschewski
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Christoph Wohlkönig
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Andelko Hrzenjak
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| |
Collapse
|
8
|
Ilkovičová L, Trošt N, Szentpéteriová E, Solár P, Komel R, Debeljak N. Overexpression of the erythropoietin receptor in RAMA 37 breast cancer cells alters cell growth and sensitivity to tamoxifen. Int J Oncol 2017; 51:737-746. [PMID: 28714517 DOI: 10.3892/ijo.2017.4061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 06/02/2017] [Indexed: 11/06/2022] Open
Abstract
Erythropoietin (EPO) is the main regulator of erythropoiesis, and its receptor (EPOR) is expressed in various tissues, including tumors. Expression of EPOR in breast cancer tissue has been shown to correlate with expression of the estrogen receptor (ER). However, EPOR promotes proliferation in an EPO-independent manner. In patients with breast cancer, EPOR is associated with impaired tamoxifen response in ER-positive tumors, but not in ER-negative tumors. Furthermore, a positive correlation between EPOR/ER status and increased local cancer recurrence has been demonstrated, and EPOR expression is associated with G-protein coupled ER (GPER). Herein, we assessed the effects of EPOR on cell physiology and tamoxifen response in the absence of EPO stimulation using two cell lines that differ only in their EPOR expression status: RAMA 37 cells (low EPOR expression) and RAMA 37-28 cells (high EPOR expression). Alterations in cell growth, morphology, response to tamoxifen cytotoxicity, and EPOR-activated signal transduction were observed. RAMA 37 cells showed higher proliferation capacity without tamoxifen treatment, while RAMA 37-28 cells were more resistant to tamoxifen and proliferated more rapidly in the presence of tamoxifen. EPOR overexpression induced cell-morphology changes upon tamoxifen treatment, which resulted in the production of cell protrusions and subsequent cell death. Short-term treatment with tamoxifen (6 h) prompted RAMA 37 cells to acquired longer protrusions than RAMA 37-28 cells, which indicated a pre-apoptotic stage. Furthermore, prolonged treatment with tamoxifen (72 h) caused a greater reduction in RAMA 37 cell numbers, which indicated a higher rate of cell death. RAMA 37-28 cells showed prolonged activation of AKT signaling. We propose sustained AKT phosphorylation in EPOR-overexpressing cells as a mechanism that can lead to EPOR-induced tamoxifen resistance.
Collapse
Affiliation(s)
- Lenka Ilkovičová
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University in Košice, 040 01 Košice, Slovakia
| | - Nina Trošt
- Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka 565-0871, Japan
| | - Erika Szentpéteriová
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University in Košice, 040 01 Košice, Slovakia
| | - Peter Solár
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University in Košice, 040 01 Košice, Slovakia
| | - Radovan Komel
- Medical Centre for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Nataša Debeljak
- Medical Centre for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
9
|
Vazquez-Mellado MJ, Monjaras-Embriz V, Rocha-Zavaleta L. Erythropoietin, Stem Cell Factor, and Cancer Cell Migration. VITAMINS AND HORMONES 2017. [DOI: 10.1016/bs.vh.2017.02.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
10
|
Wood MA, Goldman N, DePierri K, Somerville J, Riggs JE. Erythropoietin increases macrophage-mediated T cell suppression. Cell Immunol 2016; 306-307:17-24. [PMID: 27262376 PMCID: PMC4983461 DOI: 10.1016/j.cellimm.2016.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/06/2016] [Accepted: 05/25/2016] [Indexed: 12/22/2022]
Abstract
Erythropoietin (EPO), used to treat anemia in cancer patients, has been reported to accelerate tumor progression and increase mortality. Research of the mechanism for this effect has focused upon EPOR expression by tumor cells. We model the high macrophage to lymphocyte ratio found in tumor microenvironments (TMEs) by culturing peritoneal cavity (PerC) cells that naturally have a high macrophage to T cell ratio. Following TCR ligation, C57BL/6J PerC T cell proliferation is suppressed due to IFNγ-triggered inducible nitric oxide synthase (iNOS) expression. EPO was tested in the PerC culture model and found to increase T cell suppression. This effect could be abrogated by inhibiting iNOS by enzyme inhibition, genetic ablation, or blocking IFNγ signaling. Flow cytometry revealed the EPOR on CD11b(+)F4/80(+) macrophages. These results suggest that EPO could increase T cell suppression in the TME by acting directly on macrophages.
Collapse
Affiliation(s)
- Michelle A Wood
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA
| | - Naomi Goldman
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA
| | - Kelley DePierri
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA
| | - John Somerville
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA
| | - James E Riggs
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA.
| |
Collapse
|
11
|
Fecková B, Kimáková P, Ilkovičová L, Szentpéteriová E, Debeljak N, Solárová Z, Sačková V, Šemeláková M, Bhide M, Solár P. Far-western blotting as a solution to the non-specificity of the anti-erythropoietin receptor antibody. Oncol Lett 2016; 12:1575-1580. [PMID: 27446474 DOI: 10.3892/ol.2016.4782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 05/24/2016] [Indexed: 12/13/2022] Open
Abstract
The erythropoietin receptor (EpoR) is a member of the cytokine receptor family. The interaction between erythropoietin (Epo) and EpoR is important for the production and maturation of erythroid cells, resulting in the stimulation of hematopoiesis. The fact that EpoR was also detected in neoplastic cells has opened the question about the relevance of anemia treatment with recombinant Epo in cancer patients. Numerous studies have reported pro-stimulating and anti-apoptotic effects of Epo in cancer cells, thus demonstrating EpoR functionality in these cells. By contrast, a previous study claims the absence of EpoR in tumor cells. This apparent discrepancy is based, according to certain authors, on the use of non-specific anti-EpoR antibodies. With the aim of bypassing the direct detection of EpoR with an anti-EpoR antibody, the present authors propose a far-western blot methodology, which in addition, confirms the interaction of Epo with EpoR. Applying this technique, the presence of EpoR and its interaction with Epo in human ovarian adenocarcinoma A2780 and normal human umbilical vein endothelial cells was confirmed. Furthermore, modified immunoprecipitation of EpoR followed by matrix-assisted laser desorption/ionization-time-of-flight mass spectrometry analysis confirmed a 57 kDa protein as a human Epo-interacting protein in both cell lines.
Collapse
Affiliation(s)
- Barbora Fecková
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, SK-04154 Košice, Slovak Republic
| | - Patrícia Kimáková
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, SK-04154 Košice, Slovak Republic
| | - Lenka Ilkovičová
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, SK-04154 Košice, Slovak Republic
| | - Erika Szentpéteriová
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, SK-04154 Košice, Slovak Republic
| | - Nataša Debeljak
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Zuzana Solárová
- Institute of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, SK-04001 Košice, Slovak Republic
| | - Veronika Sačková
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, SK-04154 Košice, Slovak Republic
| | - Martina Šemeláková
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, SK-04154 Košice, Slovak Republic
| | - Mangesh Bhide
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine, SK-04181 Košice, Slovak Republic
| | - Peter Solár
- Department of Cell Biology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, SK-04154 Košice, Slovak Republic
| |
Collapse
|
12
|
Cai SY, Yang T, Chen Y, Wang JW, Li L, Xu MJ. Gene expression profiling of ovarian carcinomas and prognostic analysis of outcome. J Ovarian Res 2015; 8:50. [PMID: 26228058 PMCID: PMC4521463 DOI: 10.1186/s13048-015-0176-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/07/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ovarian cancer (OCA), the fifth leading deaths cancer to women, is famous for its low survival rate in epithelial ovarian cancer cases, which is very complicated and hard to be diagnosed from asymptomatic nature in the early stage. Thus, it is urgent to develop an effective genetic prognostic strategy. METHODS Current study using the Database for Annotation, Visualization and Integrated Discovery tool for the generation and analysis of quantitative gene expression profiles; all the annotated gene and biochemical pathway membership realized according to shared categorical data from Pathway and Kyoto Encyclopedia of Genes and Genomes; correlation networks based on current gene screening actualize by Weighted correlation network analysis to identify therapeutic targets gene and candidate bio-markers. RESULTS 3095 differentially expressed genes were collected from genome expression profiles of OCA patients (n = 53, 35 advanced, 8 early and 10 normal). By pathway enrichment, most genes showed contribution to cell cycle and chromosome maintenance.1073 differentially expression genes involved in the 4 dominant network modules are further generated for prognostic pattern establish, we divided a dataset with random OCA cases (n = 80) into 3 groups efficiently (p = 0.0323, 95% CIs in Kaplan-Meier). Finally, 6 prognosis related genes were selected out by COX regression analysis, TFCP2L1 related to cancer-stem cell, probably contributes to chemotherapy efficiency. CONCLUSIONS Our study presents an integrated original model of the differentially expression genes related to ovarian cancer progressing, providing the identification of genes relevant for its pathological physiology which can potentially be new clinical markers.
Collapse
Affiliation(s)
- Sheng-Yun Cai
- Department of Obstetrics & Gynecology, Changhai Hospital, Second Military Medical University, No.168, Changhai Road, Shanghai, 200433, China.
| | - Tian Yang
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, No.225, Changhai Road, Shanghai, 200438, China.
| | - Yu Chen
- Department of Obstetrics & Gynecology, Changhai Hospital, Second Military Medical University, No.168, Changhai Road, Shanghai, 200433, China.
| | - Jing-Wen Wang
- Department of Obstetrics & Gynecology, Changhai Hospital, Second Military Medical University, No.168, Changhai Road, Shanghai, 200433, China.
| | - Li Li
- Department of Obstetrics & Gynecology, Changhai Hospital, Second Military Medical University, No.168, Changhai Road, Shanghai, 200433, China.
| | - Ming-Juan Xu
- Department of Obstetrics & Gynecology, Changhai Hospital, Second Military Medical University, No.168, Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
13
|
Debeljak N, Solár P, Sytkowski AJ. Erythropoietin and cancer: the unintended consequences of anemia correction. Front Immunol 2014; 5:563. [PMID: 25426117 PMCID: PMC4227521 DOI: 10.3389/fimmu.2014.00563] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/22/2014] [Indexed: 01/12/2023] Open
Abstract
Until 1990, erythropoietin (EPO) was considered to have a single biological purpose and action, the stimulation of red blood cell growth and differentiation. Slowly, scientific and medical opinion evolved, beginning with the discovery of an effect on endothelial cell growth in vitro and the identification of EPO receptors (EPORs) on neuronal cells. We now know that EPO is a pleiotropic growth factor that exhibits an anti-apoptotic action on numerous cells and tissues, including malignant ones. In this article, we present a short discussion of EPO, receptors involved in EPO signal transduction, and their action on non-hematopoietic cells. This is followed by a more detailed presentation of both pre-clinical and clinical data that demonstrate EPO’s action on cancer cells, as well as tumor angiogenesis and lymphangiogenesis. Clinical trials with reported adverse effects of chronic erythropoiesis-stimulating agents (ESAs) treatment as well as clinical studies exploring the prognostic significance of EPO and EPOR expression in cancer patients are reviewed. Finally, we address the use of EPO and other ESAs in cancer patients.
Collapse
Affiliation(s)
- Nataša Debeljak
- Faculty of Medicine, Institute of Biochemistry, University of Ljubljana , Ljubljana , Slovenia
| | - Peter Solár
- Department of Cell and Molecular Biology, Institute of Biology and Ecology, Faculty of Sciences, Pavol Jozef Šafárik University , Košice , Slovakia
| | - Arthur J Sytkowski
- Oncology Therapeutic Area, Quintiles Transnational , Arlington, MA , USA
| |
Collapse
|
14
|
PARK SUNGLYEA, WON SEYEON, SONG JUNHUI, KIM WUNJAE, MOON SUNGKWON. EPO gene expression induces the proliferation, migration and invasion of bladder cancer cells through the p21WAF1-mediated ERK1/2/NF-κB/MMP-9 pathway. Oncol Rep 2014; 32:2207-14. [DOI: 10.3892/or.2014.3428] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 08/06/2014] [Indexed: 11/06/2022] Open
|
15
|
Li X, Yan Z, Kong D, Zou W, Wang J, Sun D, Jiang Y, Zheng C. Erythropoiesis-stimulating agents in the management of cancer patients with anemia: a meta-analysis. Chin J Cancer Res 2014; 26:268-76. [PMID: 25035653 DOI: 10.3978/j.issn.1000-9604.2014.05.03] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 05/26/2014] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Erythropoiesis-stimulating agents (ESAs) are widely used in the management of anemia in cancer patients. Despite their apparent effectiveness, recent studies have suggested that ESAs could result in serious adverse events and even higher mortality. The aim of the current study was to evaluate the benefits and risks of ESAs in the management of cancer patients with anemia using a meta-analysis. METHODS The initial literature search covered Medline, PubMed, Embase, and the Cochrane Center Register of Controlled Trials, and identified 1,569 articles. The final meta-analysis included eight randomized controlled trials (n=2,387) in cancer patients with <11 g/dL hemoglobin (Hb) at the baseline and target Hb (for stopping ESA treatment) at no more than 13 g/dL. The assessment measures included Hb response, blood transfusion rate and adverse events that included venous thromboemblism (VTE), hypertension, and on-study mortality. The results are expressed as pooled odds ratio (OR). Publication bias was assessed using funnel plot analysis. RESULTS ESAs significantly increased the Hb concentration [OR 7.85, 95% confidence interval (CI): 5.85 to 10.53, P<0.001] and reduced the red blood cell (RBC) transfusion rate (OR 0.52, 95% CI: 0.42 to 0.65, P<0.001). ESAs did not increase the accumulated adverse events (OR 0.95, P=0.82), or the on-study mortality (OR 1.09, P=0.47). CONCLUSIONS ESAs are not associated with increased frequency of severe adverse events in anemic cancer patients when the target Hb value is no more than 13 g/dL.
Collapse
Affiliation(s)
- Xiaomei Li
- 1 Cancer Center of the Second Hospital, 2 Institute of Biotherapy for Hematological Malignancies, 3 Cardiovascular Department of the Second Hospital, 4 Hematology Department of the Second Hospital, 5 Pharmacology Department of the Second Hospital, Shandong University, Jinan 250100, China
| | - Zhi Yan
- 1 Cancer Center of the Second Hospital, 2 Institute of Biotherapy for Hematological Malignancies, 3 Cardiovascular Department of the Second Hospital, 4 Hematology Department of the Second Hospital, 5 Pharmacology Department of the Second Hospital, Shandong University, Jinan 250100, China
| | - Dexiao Kong
- 1 Cancer Center of the Second Hospital, 2 Institute of Biotherapy for Hematological Malignancies, 3 Cardiovascular Department of the Second Hospital, 4 Hematology Department of the Second Hospital, 5 Pharmacology Department of the Second Hospital, Shandong University, Jinan 250100, China
| | - Wen Zou
- 1 Cancer Center of the Second Hospital, 2 Institute of Biotherapy for Hematological Malignancies, 3 Cardiovascular Department of the Second Hospital, 4 Hematology Department of the Second Hospital, 5 Pharmacology Department of the Second Hospital, Shandong University, Jinan 250100, China
| | - Jihua Wang
- 1 Cancer Center of the Second Hospital, 2 Institute of Biotherapy for Hematological Malignancies, 3 Cardiovascular Department of the Second Hospital, 4 Hematology Department of the Second Hospital, 5 Pharmacology Department of the Second Hospital, Shandong University, Jinan 250100, China
| | - Dianshui Sun
- 1 Cancer Center of the Second Hospital, 2 Institute of Biotherapy for Hematological Malignancies, 3 Cardiovascular Department of the Second Hospital, 4 Hematology Department of the Second Hospital, 5 Pharmacology Department of the Second Hospital, Shandong University, Jinan 250100, China
| | - Yuhua Jiang
- 1 Cancer Center of the Second Hospital, 2 Institute of Biotherapy for Hematological Malignancies, 3 Cardiovascular Department of the Second Hospital, 4 Hematology Department of the Second Hospital, 5 Pharmacology Department of the Second Hospital, Shandong University, Jinan 250100, China
| | - Chengyun Zheng
- 1 Cancer Center of the Second Hospital, 2 Institute of Biotherapy for Hematological Malignancies, 3 Cardiovascular Department of the Second Hospital, 4 Hematology Department of the Second Hospital, 5 Pharmacology Department of the Second Hospital, Shandong University, Jinan 250100, China
| |
Collapse
|
16
|
Yang Z, Wang H, Jiang Y, Hartnett ME. VEGFA activates erythropoietin receptor and enhances VEGFR2-mediated pathological angiogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1230-1239. [PMID: 24630601 DOI: 10.1016/j.ajpath.2013.12.023] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 11/27/2013] [Accepted: 12/05/2013] [Indexed: 10/25/2022]
Abstract
Clinical and animal studies implicate erythropoietin (EPO) and EPO receptor (EPOR) signaling in angiogenesis. In the eye, EPO is involved in both physiological and pathological angiogenesis in the retina. We hypothesized that EPOR signaling is important in pathological angiogenesis and tested this hypothesis using a rat model of oxygen-induced retinopathy that is representative of human retinopathy of prematurity. We first determined that EPOR expression and activation were increased and that activated EPOR was localized to retinal vascular endothelial cells (ECs) in retinas at postnatal day 18 (p18), when pathological angiogenesis in the form of intravitreal neovascularization occurred. In human retinal microvascular ECs, EPOR was up-regulated and activated by VEGF. Lentiviral-delivered shRNAs that knocked down Müller cell-expressed VEGF in the retinopathy of prematurity model also reduced phosphorylated EPOR (p-EPOR) and VEGFR2 (p-VEGFR2) in retinal ECs. In human retinal microvascular ECs, VEGFR2-activated EPOR caused an interaction between p-EPOR and p-VEGFR2; knockdown of EPOR by siRNA transfection reduced VEGF-induced EC proliferation in association with reduced p-VEGFR2 and p-STAT3; however, inhibition of VEGFR2 activation by siRNA transfection or semaxanib (SU5416) abolished VEGFA-induced proliferation of ECs and phosphorylation of VEGFR2, EPOR, and STAT3. Our results show that VEGFA-induced p-VEGFR2 activates EPOR and causes an interaction between p-EPOR and p-VEGFR2 to enhance VEGFA-induced EC proliferation by exacerbating STAT3 activation, leading to pathological angiogenesis.
Collapse
Affiliation(s)
- Zhihong Yang
- The John Moran Eye Center, The University of Utah, Salt Lake City, Utah
| | - Haibo Wang
- The John Moran Eye Center, The University of Utah, Salt Lake City, Utah
| | - Yanchao Jiang
- The John Moran Eye Center, The University of Utah, Salt Lake City, Utah
| | | |
Collapse
|
17
|
EPO-independent functional EPO receptor in breast cancer enhances estrogen receptor activity and promotes cell proliferation. Biochem Biophys Res Commun 2014; 445:163-9. [PMID: 24502950 DOI: 10.1016/j.bbrc.2014.01.165] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 01/27/2014] [Indexed: 12/28/2022]
Abstract
The main function of Erythropoietin (EPO) and its receptor (EPOR) is the stimulation of erythropoiesis. Recombinant human EPO (rhEPO) is therefore used to treat anemia in cancer patients. However, clinical trials have indicated that rhEPO treatment might promote tumor progression and has a negative effect on patient survival. In addition, EPOR expression has been detected in several cancer forms. Using a newly produced anti-EPOR antibody that reliably detects the full-length isoform of the EPOR we show that breast cancer tissue and cells express the EPOR protein. rhEPO stimulation of cultured EPOR expressing breast cancer cells did not result in increased proliferation, overt activation of EPOR (receptor phosphorylation) or a consistent activation of canonical EPOR signaling pathway mediators such as JAK2, STAT3, STAT5, or AKT. However, EPOR knockdown experiments suggested functional EPO receptors in estrogen receptor positive (ERα(+)) breast cancer cells, as reduced EPOR expression resulted in decreased proliferation. This effect on proliferation was not seen in ERα negative cells. EPOR knockdown decreased ERα activity further supports a mechanism by which EPOR affects proliferation via ERα-mediated mechanisms. We show that EPOR protein is expressed in breast cancer cells, where it appears to promote proliferation by an EPO-independent mechanism in ERα expressing breast cancer cells.
Collapse
|
18
|
Mamlouk S, Kalucka J, Singh RP, Franke K, Muschter A, Langer A, Jakob C, Gassmann M, Baretton GB, Wielockx B. Loss of prolyl hydroxylase-2 in myeloid cells and T-lymphocytes impairs tumor development. Int J Cancer 2013; 134:849-58. [DOI: 10.1002/ijc.28409] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 06/14/2013] [Accepted: 07/16/2013] [Indexed: 12/20/2022]
Affiliation(s)
- Soulafa Mamlouk
- Emmy Noether Research Group; University of Technology; Dresden Germany
- Institute of Pathology; University of Technology Dresden; Dresden Germany
| | - Joanna Kalucka
- Emmy Noether Research Group; University of Technology; Dresden Germany
- Institute of Pathology; University of Technology Dresden; Dresden Germany
| | - Rashim Pal Singh
- Emmy Noether Research Group; University of Technology; Dresden Germany
- Institute of Pathology; University of Technology Dresden; Dresden Germany
| | - Kristin Franke
- Emmy Noether Research Group; University of Technology; Dresden Germany
- Institute of Pathology; University of Technology Dresden; Dresden Germany
| | - Antje Muschter
- Emmy Noether Research Group; University of Technology; Dresden Germany
- Institute of Pathology; University of Technology Dresden; Dresden Germany
| | - Anika Langer
- Emmy Noether Research Group; University of Technology; Dresden Germany
- Institute of Pathology; University of Technology Dresden; Dresden Germany
| | - Christiane Jakob
- Institute of Pathology; University of Technology Dresden; Dresden Germany
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty and Zurich Center for Integrative Human Physiology (ZIHP); University of Zürich; Zürich Switzerland
- Universidad Peruana Cayetano Heredia (UPCH); Lima Peru
| | | | - Ben Wielockx
- Emmy Noether Research Group; University of Technology; Dresden Germany
- Institute of Pathology; University of Technology Dresden; Dresden Germany
- DFG Research Center and Cluster of Excellence for Regenerative Therapies Dresden; University of Technology Dresden; Dresden Germany
| |
Collapse
|
19
|
Kriška J, Solár P, Varinská L, Solárová Z, Kimáková P, Mojžiš J, Fedoročko P, Sytkowski AJ. Human erythropoietin increases the pro-angiogenic potential of A2780 ovarian adenocarcinoma cells under hypoxic conditions. Oncol Rep 2013; 30:1455-62. [PMID: 23807540 DOI: 10.3892/or.2013.2566] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 05/09/2013] [Indexed: 11/06/2022] Open
Abstract
Erythropoietin (Epo) is a key regulator of erythroid cell proliferation, differentiation and apoptosis. In the form of the recombinant protein, it is widely used to treat various types of anemias, including that associated with cancer and with the myelosuppressive effects of chemotherapy, particularly platinum-based regimens. Our previous studies confirmed the presence of Epo receptors (EpoRs) in ovarian adenocarcinoma cell lines and demonstrated that long-term Epo treatment of A2780 cells resulted in the development of a phenotype exhibiting both enhanced Epo signaling and increased paclitaxel resistance. In the present study, we carried out a series of experiments to analyze the pro-angiogenic potential of Epo-treated A2780 and SKOV-3 cells. Our studies revealed that conditioned media of Epo-treated A2780 cells had a stimulative effect on human umbilical vein endothelial cells (HUVECs). This effect was only seen when A2780 cells were incubated under hypoxic conditions. Furthermore, Epo increased the secretion of interleukin (IL)-4, IL-5, IL-6, IL-8, IL-10, IL-12, IL-13, GM-CSF and interferon-γ by A2780 cells that grew in hypoxic conditions. In this regard, conditioned media of hypoxic and Epo-treated A2780 cells induced a significant phosphorylation of STAT-5 in HUVECs. Our results may have important implications for ovarian cancer patients receiving Epo.
Collapse
Affiliation(s)
- Ján Kriška
- Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University, Košice, Slovak Republic
| | | | | | | | | | | | | | | |
Collapse
|
20
|
The erythropoietin receptor is a downstream effector of Klotho-induced cytoprotection. Kidney Int 2013; 84:468-81. [PMID: 23636173 PMCID: PMC3758776 DOI: 10.1038/ki.2013.149] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 02/11/2013] [Accepted: 02/14/2013] [Indexed: 12/22/2022]
Abstract
Although the role of the erythropoietin (EPO) receptor (EpoR) in erythropoiesis has been known for decades, its role in nonhematopoietic tissues is still not well defined. Klotho has been shown and EPo has been suggested to protect against acute ischemia-reperfusion injury in the kidney. Here we found in rat kidney and in a rat renal tubular epithelial cell line (NRK cells) EpoR transcript and antigen, and EpoR activity signified as EPo-induced phosphorylation of Jak2, ErK, Akt, and Stat5 indicating the presence of functional EpoR. Transgenic overexpression of Klotho or addition of exogenous recombinant Klotho increased kidney EpoR protein and transcript. In NRK cells, Klotho increased EpoR protein, enhanced EPo-triggered phosphorylation of Jak2 and Stat5, the nuclear translocation of phospho-Stat5, and protected NRK cells from hydrogen peroxide cytotoxicity. Knockdown of endogenous EpoR rendered NRK cells more vulnerable, and overexpression of EpoR more resistant to peroxide-induced cytotoxicity, indicating that EpoR mitigates oxidative damage. Knockdown of EpoR by siRNA abolished Epo-induced Jak2, and Stat5 phosphorylation, and blunted the protective effect of Klotho against peroxide-induced cytotoxicity. Thus in the kidney, EpoR and its activity are downstream effectors of Klotho enabling it to function as a cytoprotective protein against oxidative injury.
Collapse
|
21
|
Zhang C, Duan X, Xu L, Ye J, Zhao J, Liu Y. Erythropoietin receptor expression and its relationship with trastuzumab response and resistance in HER2-positive breast cancer cells. Breast Cancer Res Treat 2012; 136:739-48. [PMID: 23117856 DOI: 10.1007/s10549-012-2316-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2012] [Accepted: 10/25/2012] [Indexed: 11/24/2022]
Abstract
Resistance to trastuzumab is a major issue in the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer. Several potential resistance mechanisms have been investigated, but the results are controversial and no conclusion has been reached. Erythropoietin receptor (EPOR) may function in cell growth, and expressed in various cancer cells. Because the downstream signaling pathways for EPOR and HER2 partially overlapped, we hypothesized that EPOR may play a role in the inhibition effect of trastuzumab and resistance to trastuzumab. Here, we detected the expression of EPOR mRNA and protein in HER2-positive breast cancer cell lines and tissues. EPOR expressed in SKBR3, MDA-MB-453, and UACC-812 cell lines, but not in BT474. Of the 55 HER2-positive cancer tissues, EPOR was positive in 42 samples and highly expressed (H-score ≥ 25) in 24 by immunohistochemistry. The difference between EPOR expression and Ki67 index was significant (P = 0.033), and EPOR expression also positively correlated with higher pathological stage (Spearman correlation coefficient = 0.359; P = 0.007). Exogenous EPO antagonized trastuzumab-induced inhibition of cell proliferation in HER2/EPOR dual-positive breast cancer cells. We then exposed SKBR3 cells to trastuzumab for 4 months to obtain trastuzumab-resistant SKBR3 cell line, which demonstrated higher phosphorylated EPOR level, higher EPO expression and more extracellular secretion than non-resistant parental SKBR3 cells. Downregulation EPOR expression using short hairpin RNA resensitized trastuzumab-resistant cells to this drug, and SKBR3 cells with EPOR downregulation demonstrated attenuated trastuzumab resistance after the same resistance induction. EPOR downregulation plus trastuzumab produced a synergetic action in the inhibition of cell proliferation and invasion in SKBR3 and MDA-MB-453 cell lines. Therefore, EPOR expression may be involved in tumor progression and proliferation in HER2-positive breast cancer. EPO/EPOR contributes to the mechanism of trastuzumab resistance in SKBR3 cell lines, and EPOR downregulation can reverse the resistance to trastuzumab and increase the inhibition effect of this drug.
Collapse
Affiliation(s)
- Chi Zhang
- Peking University First Hospital Breast Disease Centre, Xishiku Street 8#, Xicheng District, Beijing, China
| | | | | | | | | | | |
Collapse
|
22
|
The erythropoietin/erythropoietin receptor signaling pathway promotes growth and invasion abilities in human renal carcinoma cells. PLoS One 2012; 7:e45122. [PMID: 23028796 PMCID: PMC3445554 DOI: 10.1371/journal.pone.0045122] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 08/13/2012] [Indexed: 01/31/2023] Open
Abstract
Co-expression of erythropoietin (Epo) and erythropoietin receptor (EpoR) has been found in various non-hematopoietic cancers including hereditary and sporadic renal cell carcinomas (RCC), but the Epo/EpoR autocrine and paracrine mechanisms in tumor progression have not yet been identified. In this study, we used RNA interference method to down-regulate EpoR to investigate the function of Epo/EpoR pathway in human RCC cells. Epo and EpoR co-expressed in primary renal cancer cells and 6 human RCC cell lines. EpoR signaling was constitutionally phosphorylated in primary renal cancer cells, 786-0 and Caki-1 cells, and recombinant human Epo (rhEpo) stimulation had no significant effects on further phosphorylation of EpoR pathway, proliferation, and invasiveness of the cells. Down-regulation of EpoR expression in 786-0 cells by lentivirus-introduced siRNA resulted in inhibition of growth and invasiveness in vitro and in vivo, and promotion of cell apoptosis. In addition, rhEpo stimulation slightly antagonized the anti-tumor effect of Sunitinib on 786-0 cells. Sunitinib could induce more apoptotic cells in 786-0 cells with knockdown EpoR expression. Our results suggested that Epo/EpoR pathway was involved in cell growth, invasion, survival, and sensitivity to the multi-kinases inhibitor Sunitinib in RCC cells.
Collapse
|
23
|
Abstract
Erythropoietin (Epo) is an essential hormone that binds and activates the Epo receptor (EpoR) resident on the surface of erythroid progenitor cells, thereby promoting erythropoiesis. Recombinant human erythropoietin has been used successfully for over 20 years to treat anemia in millions of patients. In addition to erythropoiesis, Epo has also been reported to have other effects, such as tissue protection and promotion of tumor cell growth or survival. This became of significant concern in 2003, when some clinical trials in cancer patients reported increased tumor progression and worse survival outcomes in patients treated with erythropoiesis-stimulating agents (ESAs). One of the potential mechanisms proffered to explain the observed safety issues was that functional EpoR was expressed in tumors and/or endothelial cells, and that ESAs directly stimulated tumor growth and/or antagonized tumor ablative therapies. Since then, numerous groups have performed further research evaluating this potential mechanism with conflicting data and conclusions. Here, we review the biology of endogenous Epo and EpoR expression and function in erythropoiesis, and evaluate the evidence pertaining to the expression of EpoR on normal nonhematopoietic and tumor cells.
Collapse
|
24
|
Kumar SM, Zhang G, Bastian BC, Arcasoy MO, Karande P, Pushparajan A, Acs G, Xu X. Erythropoietin receptor contributes to melanoma cell survival in vivo. Oncogene 2012; 31:1649-60. [PMID: 21860424 PMCID: PMC3441831 DOI: 10.1038/onc.2011.366] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 07/14/2011] [Indexed: 12/26/2022]
Abstract
Erythropoietin (Epo) is widely used clinically to treat anemia associated with various clinical conditions including cancer. Data from several clinical trials suggest significant adverse effect of Epo treatment on cancer patient survival. However, controversy exists whether Epo receptor (EpoR) is functional in cancer cells. In this study, we demonstrated that EpoR mRNA expression was detectable in 90.1% of 65 melanoma cell lines, and increased copy number of the Epo and EpoR loci occurred in 30 and 24.6% of 130 primary melanomas, respectively. EpoR knockdown in melanoma cells resulted in diminished ERK phosphorylation in response to Epo stimulation, decreased cell proliferation and increased response to the inhibitory effect of hypoxia and cisplatin in vitro. EpoR knockdown significantly decreased melanoma xenograft size and tumor invasion in vivo. On the contrary, constitutive activation of EpoR activated cell proliferation pathways in melanoma cells and resulted in increased cell proliferation and resistance to hypoxia and cisplatin treatment in vitro. EpoR activation resulted in significantly larger xenografts with increased tumor invasion of surrounding tissue in vivo. Daily administration of recombinant Epo fails to stimulate melanoma growth in vivo, but the treatment increased vascular size in the xenografts. Increased local recurrence after excision of the primary tumors was observed after Epo treatment. Epo induced angiogenesis in Matrigel plug assays, and neutralization of Epo secreted by melanoma cells results in decreased angiogenesis. These data support that EpoR is functional in melanoma and EpoR activation may promote melanoma progression, and suggest that Epo may stimulate angiogenesis and increase survival of melanoma cells under hypoxic condition in vivo.
Collapse
Affiliation(s)
- Suresh M. Kumar
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Gao Zhang
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, USA
| | - Boris C. Bastian
- Departments of Dermatology and Pathology, University of California, San Francisco, CA, USA
| | - Murat O. Arcasoy
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Pankaj Karande
- Departments of Chemical and Biological Engineering, Rensselaer Polytechnology Institute, Troy, NY, USA
| | - Anitha Pushparajan
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Geza Acs
- Departments of Anatomic Pathology and Women's Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
25
|
Elliott S, Busse L, Swift S, McCaffery I, Rossi J, Kassner P, Begley CG. Lack of expression and function of erythropoietin receptors in the kidney. Nephrol Dial Transplant 2011; 27:2733-45. [DOI: 10.1093/ndt/gfr698] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
26
|
Hand CC, Brines M. Promises and pitfalls in erythopoietin-mediated tissue protection: are nonerythropoietic derivatives a way forward? J Investig Med 2011; 59. [PMID: 20683348 PMCID: PMC3023830 DOI: 10.231/jim.0b013e3181ed30bf] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The essential biological role of erythropoietin (EPO) in maintaining erythrocyte mass has been well understood for many years. Although EPO is required for the maturation of red cells, it also has strong procoagulant effects on the vascular endothelium and platelets, which limit erythrocyte losses after hemorrhage. Like other members of the type 1 cytokine superfamily, EPO has multiple biological activities. For the past 10 years, multiple investigators have shown that EPO acts as a locally produced antagonist of proinflammatory cytokines that are generated by the innate immune response in response to infection, trauma, or metabolic stress. Specifically, EPO inhibits apoptosis of cells surrounding a locus of injury, reduces the influx of inflammatory cells, and recruits tissue-specific stem cells and endothelial progenitor cells. Available evidence suggests that these multiple, nonerythropoietic effects of EPO are mediated by a tissue protective receptor (TPR) that is distinct from the homodimeric receptor responsible for erythropoiesis. Notably, activation of the TPR requires a higher concentration of EPO than is needed for maximal erythropoiesis. Unfortunately, these higher concentrations of EPO also stimulate hematopoietic and procoagulant pathways, which can cause adverse effects and, therefore, potentially limit the clinical use of EPO for tissue protection. To circumvent these problems, the EPO molecule has been successfully modified in a variety of ways to interact only with the TPR. Early clinical experience has shown that these compounds appear to be safe, and proof of concept trials are ready to begin.
Collapse
|
27
|
Hand CC, Brines M. Promises and pitfalls in erythopoietin-mediated tissue protection: are nonerythropoietic derivatives a way forward? J Investig Med 2011; 59:1073-82. [PMID: 20683348 PMCID: PMC3023830 DOI: 10.2310/jim.0b013e3181ed30bf] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2025]
Abstract
The essential biological role of erythropoietin (EPO) in maintaining erythrocyte mass has been well understood for many years. Although EPO is required for the maturation of red cells, it also has strong procoagulant effects on the vascular endothelium and platelets, which limit erythrocyte losses after hemorrhage. Like other members of the type 1 cytokine superfamily, EPO has multiple biological activities. For the past 10 years, multiple investigators have shown that EPO acts as a locally produced antagonist of proinflammatory cytokines that are generated by the innate immune response in response to infection, trauma, or metabolic stress. Specifically, EPO inhibits apoptosis of cells surrounding a locus of injury, reduces the influx of inflammatory cells, and recruits tissue-specific stem cells and endothelial progenitor cells. Available evidence suggests that these multiple, nonerythropoietic effects of EPO are mediated by a tissue protective receptor (TPR) that is distinct from the homodimeric receptor responsible for erythropoiesis. Notably, activation of the TPR requires a higher concentration of EPO than is needed for maximal erythropoiesis. Unfortunately, these higher concentrations of EPO also stimulate hematopoietic and procoagulant pathways, which can cause adverse effects and, therefore, potentially limit the clinical use of EPO for tissue protection. To circumvent these problems, the EPO molecule has been successfully modified in a variety of ways to interact only with the TPR. Early clinical experience has shown that these compounds appear to be safe, and proof of concept trials are ready to begin.
Collapse
|
28
|
Pérès EA, Valable S, Guillamo JS, Marteau L, Bernaudin JF, Roussel S, Lechapt-Zalcman E, Bernaudin M, Petit E. Targeting the erythropoietin receptor on glioma cells reduces tumour growth. Exp Cell Res 2011; 317:2321-32. [PMID: 21749867 DOI: 10.1016/j.yexcr.2011.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 06/21/2011] [Accepted: 06/21/2011] [Indexed: 12/15/2022]
Abstract
Hypoxia has been shown to be one of the major events involved in EPO expression. Accordingly, EPO might be expressed by cerebral neoplastic cells, especially in glioblastoma, known to be highly hypoxic tumours. The expression of EPOR has been described in glioma cells. However, data from the literature remain descriptive and controversial. On the basis of an endogenous source of EPO in the brain, we have focused on a potential role of EPOR in brain tumour growth. In the present study, with complementary approaches to target EPO/EPOR signalling, we demonstrate the presence of a functional EPO/EPOR system on glioma cells leading to the activation of the ERK pathway. This EPO/EPOR system is involved in glioma cell proliferation in vitro. In vivo, we show that the down-regulation of EPOR expression on glioma cells reduces tumour growth and enhances animal survival. Our results support the hypothesis that EPOR signalling in tumour cells is involved in the control of glioma growth.
Collapse
Affiliation(s)
- Elodie A Pérès
- CERVOxy team Hypoxia and cerebrovascular pathophysiology, UMR 6232 CI-NAPS, Université de Caen Basse-Normandie, Université Paris-Descartes, Caen, France
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
|
30
|
Erythropoietin for oncology supportive care. Exp Cell Res 2011; 317:1246-54. [DOI: 10.1016/j.yexcr.2011.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 03/03/2011] [Accepted: 03/03/2011] [Indexed: 12/11/2022]
|
31
|
Abstract
RNA interference, a recently discovered new mechanism controlling gene expression via small RNAs, was shown to be involved in characterization and control of basic ovarian cell functions. The main classes of small RNAs, as well as their expression in ovaries have been described. Furthermore, the successful application of RNA interference for study and control of basic ovarian functions (proliferation, apoptosis, secretory activity, luteogenesis, oocyte maturation, and related ovarian cell malignant transformation) and production of recombinant proteins have been demonstrated. Application of RNA interference in reproductive biology and medicine can be successful in two main areas: (1) characterization and prediction of physiological and pathological state (association between particular small RNA and physiological or pathological processes), (2) application of small RNAs for regulation of reproductive processes and treatment of reproductive disorders or their particular indexes. Problems of improvement of small RNA delivery to target ovarian cells and potent RNA interference-related approaches for treatment of ovarian disorders (especially of ovarian cancer) have been discussed.
Collapse
Affiliation(s)
- Alexander V Sirotkin
- Institute of Animal Genetics and Reproduction, Animal Production Research Centre Nitra, Luzianky near Nitra, Slovakia.
| |
Collapse
|
32
|
Abstract
RNA interference (RNAi) is an evolutionary conserved mechanism for specific gene silencing. This mechanism has great potential for use in targeted cancer therapy. Understanding the RNAi mechanism has led to the development of several novel RNAi-based therapeutic approaches currently in the early phases of clinical trials. It remains difficult to effectively deliver the nucleic acids required in vivo to initiate RNAi, and intense effort is under way in developing effective and targeted systemic delivery systems for RNAi. Description of in vivo delivery systems is not the focus of this review. In this review, we cover the rationale for pursuing personalised cancer therapy with RNAi, briefly review the mechanism of each major RNAi therapeutic technique, summarise and sample recent results with animal models applying RNAi for cancer, and provide an update on current clinical trials with RNAi-based therapeutic agents for cancer therapy. RNAi-based cancer therapy is still in its infancy, and there are numerous obstacles and issues that need to be resolved before its application in personalised therapy focusing on patient-cancer-specific targets can become standard cancer treatment, either alone or in combination with other treatments.
Collapse
|
33
|
Szenajch J, Wcislo G, Jeong JY, Szczylik C, Feldman L. The role of erythropoietin and its receptor in growth, survival and therapeutic response of human tumor cells From clinic to bench - a critical review. Biochim Biophys Acta Rev Cancer 2010; 1806:82-95. [PMID: 20406667 DOI: 10.1016/j.bbcan.2010.04.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 04/05/2010] [Accepted: 04/11/2010] [Indexed: 12/27/2022]
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.
Collapse
Affiliation(s)
- Jolanta Szenajch
- Laboratory for Molecular Oncology, Military Institute of Medicine, Warsaw, Poland
| | | | | | | | | |
Collapse
|
34
|
Solár P, Koval J, Mikeš J, Kleban J, Solárová Z, Lazúr J, Hodorová I, Fedoročko P, Sytkowski AJ. Erythropoietin and Ovarian Cancer – Response. Mol Cancer Ther 2010. [DOI: 10.1158/1535-7163.mct-09-1217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Peter Solár
- P.J. Šafárik University, Košice, Slovak Republic
| | - Ján Koval
- P.J. Šafárik University, Košice, Slovak Republic
| | | | - Ján Kleban
- P.J. Šafárik University, Košice, Slovak Republic
| | | | - Ján Lazúr
- P.J. Šafárik University, Košice, Slovak Republic
| | | | | | - Arthur J. Sytkowski
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
35
|
Shi Z, Hodges VM, Dunlop EA, Percy MJ, Maxwell AP, El-Tanani M, Lappin TRJ. Erythropoietin-induced activation of the JAK2/STAT5, PI3K/Akt, and Ras/ERK pathways promotes malignant cell behavior in a modified breast cancer cell line. Mol Cancer Res 2010; 8:615-626. [PMID: 20353997 DOI: 10.1158/1541-7786.mcr-09-0264] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Erythropoietin (Epo), the major regulator of erythropoiesis, and its cognate receptor (EpoR) are also expressed in nonerythroid tissues, including tumors. Clinical studies have highlighted the potential adverse effects of erythropoiesis-stimulating agents when used to treat cancer-related anemia. We assessed the ability of EpoR to enhance tumor growth and invasiveness following Epo stimulation. A benign noninvasive rat mammary cell line, Rama 37, was used as a model system. Cell signaling and malignant cell behavior were compared between parental Rama 37 cells, which express few or no endogenous EpoRs, and a modified cell line stably transfected with human EpoR (Rama 37-28). The incubation of Rama 37-28 cells with pharmacologic levels of Epo led to the rapid and sustained increases in phosphorylation of signal transducers and activators of transcription 5, Akt, and extracellular signal-regulated kinase. The activation of these signaling pathways significantly increased invasion, migration, adhesion, and colony formation. The Epo-induced invasion capacity of Rama 37-28 cells was reduced by the small interfering RNA-mediated knockdown of EpoR mRNA levels and by inhibitors of the phosphoinositide 3-kinase/Akt and Ras/extracellular signal-regulated kinase signaling pathways with adhesion also reduced by Janus-activated kinase 2/signal transducers and activators of transcription 5 inhibition. These data show that Epo induces phenotypic changes in the behavior of breast cancer cell lines and establishes links between individual cell signaling pathways and the potential for cancer spread.
Collapse
Affiliation(s)
- Zhanzhong Shi
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Certain oncology trials showed worse clinical outcomes in the erythropoiesis-stimulating agent (ESA) arm. A potential explanation was that ESA-activated erythropoietin (Epo) receptors (EpoRs) promoted tumor cell growth. Although there were supportive data from preclinical studies, those findings often used invalidated reagents and methodologies and were in conflict with other studies. Here, we further investigate the expression and function of EpoR in tumor cell lines. EpoR mRNA levels in 209 human cell lines representing 16 tumor types were low compared with ESA-responsive positive controls. EpoR protein production was evaluated in a subset of 66 cell lines using a novel anti-EpoR antibody. EpoR(+) control cells had an estimated 10 000 to 100 000 EpoR dimers/cell. In contrast, 54 of 61 lines had EpoR protein levels lower than 100 dimers/cell. Cell lines with the highest EpoR protein levels (400-3200 dimers/cell) were studied further, and, although one line, NCI-H661, bound detectable levels of [(125)I]-recombinant human Epo (rHuEpo), none showed evidence of ESA-induced EpoR activation. There was no increased phosphorylation of STAT5, AKT, ERK, or S6RP with rHuEpo. In addition, EpoR knockdown with siRNAs did not affect viability in 2 cell lines previously reported to express functional EpoR (A2780 and SK-OV-3). These results conflict with the hypothesis that EpoR is functionally expressed in tumors.
Collapse
|