Skip to main content

Erythropoietin Receptor (EpoR) Agonism Is Used to Treat a Wide Range of Disease

Abstract

The erythropoietin receptor (EpoR) was discovered and described in red blood cells (RBCs), stimulating its proliferation and survival. The target in humans for EpoR agonists drugs appears clear—to treat anemia. However, there is evidence of the pleitropic actions of erythropoietin (Epo). For that reason, rhEpo therapy was suggested as a reliable approach for treating a broad range of pathologies, including heart and cardiovascular diseases, neurodegenerative disorders (Parkinson’s and Alzheimer’s disease), spinal cord injury, stroke, diabetic retinopathy and rare diseases (Friedreich ataxia). Unfortunately, the side effects of rhEpo are also evident. A new generation of nonhematopoietic EpoR agonists drugs (asialoEpo, Cepo and ARA 290) have been investigated and further developed. These EpoR agonists, without the erythropoietic activity of Epo, while preserving its tissue-protective properties, will provide better outcomes in ongoing clinical trials. Nonhematopoietic EpoR agonists represent safer and more effective surrogates for the treatment of several diseases such as brain and peripheral nerve injury, diabetic complications, renal ischemia, rare diseases, myocardial infarction, chronic heart disease and others.

In principle, the erythropoietin receptor (EpoR) was discovered and described in red blood cell (RBC) progenitors, stimulating its proliferation and survival. Erythropoietin (Epo) is mainly synthesized in fetal liver and adult kidneys (1–3). Therefore, it was hypothesized that Epo act exclusively on erythroid progenitor cells. Accordingly, the target in humans for EpoR agonists drugs (such as recombinant erythropoietin [rhEpo], in general, called erythropoiesis-simulating agents) appears clear (that is, to treat anemia). However, evidence of a kaleidoscope of pleitropic actions of Epo has been provided (4,5). The Epo/EpoR axis research involved an initial journey from laboratory basic research to clinical therapeutics. However, as a consequence of clinical observations, basic research on Epo/EpoR comes back to expand its clinical therapeutic applicability.

Although kidney and liver have long been considered the major sources of synthesis, Epo mRNA expression has also been detected in the brain (neurons and glial cells), lung, heart, bone marrow, spleen, hair follicles, reproductive tract and osteoblasts (6–17). Accordingly, EpoR was detected in other cells, such as neurons, astrocytes, microglia, immune cells, cancer cell lines, endothelial cells, bone marrow stromal cells and cells of heart, reproductive system, gastrointestinal tract, kidney, pancreas and skeletal muscle (18–27). Conversely, Sinclair et al. (28) reported data questioning the presence or function of EpoR on non-hematopoietic cells (endothelial, neuronal and cardiac cells), suggesting that further studies are needed to confirm the diversity of EpoR. Elliott et al. (29) also showed that EpoR is virtually undetectable in human renal cells and other tissues with no detectable EpoR on cell surfaces. These results have raised doubts about the preclinical basis for studies exploring pleiotropic actions of rhEpo (30).

For the above-mentioned data, a return to basic research studies has become necessary, and many studies in animal models have been initiated or have already been performed. The effect of rhEpo administration on angiogenesis, myogenesis, shift in muscle fiber types and oxidative enzyme activities in skeletal muscle (4,31), cardiac muscle mitochondrial biogenesis (32), cognitive effects (31), antiapoptotic and antiinflammatory actions (33–37) and plasma glucose concentrations (38) has been extensively studied. Neuro- and cardioprotection properties have been mainly described. Accordingly, rhEpo therapy was suggested as a reliable approach for treating a broad range of pathologies, including heart and cardiovascular diseases, neurodegenerative disorders (Parkinson’s and Alzheimer’s disease), spinal cord injury, stroke, diabetic retinopathy and rare diseases (Friedreich ataxia).

Unfortunately, the side effects of rhEpo are also evident. Epo is involved in regulating tumor angiogenesis (39) and probably in the survival and growth of tumor cells (25,40,41). rhEpo administration also induces serious side effects such as hypertension, polycythemia, myocardial infarction, stroke and seizures, platelet activation and increased thromboembolic risk, and immunogenicity (42–46), with the most common being hypertension (47,48). A new generation of non-hematopoietic EpoR agonists drugs have hence been investigated and further developed in animals models. These compounds, namely asialoerythropoietin (asialoEpo) and carbamylated Epo (Cepo), were developed for preserving tissue-protective properties but reducing the erythropoietic activity of native Epo (49,50). These drugs will provide better outcome in ongoing clinical trials. The advantage of using nonhematopoietic Epo analogs is to avoid the stimulation of hematopoiesis and thereby the prevention of an increased hematocrit with a subsequent procoagulant status or increased blood pressure. In this regard, a new study by van Rijt et al. has shed new light on this topic (51). A new non-hematopoietic EpoR agonist analog named ARA 290 has been developed, promising cytoprotective capacities to prevent renal ischemia/reperfusion injury (51). ARA 290 is a short peptide that has shown no safety concerns in preclinical and human studies. In addition, ARA 290 has proven efficacious in cardiac disorders (52,53), neuropathic pain (54) and sarcoidosis-induced chronic neuropathic pain (55). Thus, ARA 290 is a novel non-hematopoietic EpoR agonist with promising therapeutic options in treating a wide range of pathologies and without increased risks of cardiovascular events.

Overall, this new generation of EpoR agonists without the erythropoietic activity of Epo while preserving tissue-protective properties of Epo will provide better outcomes in ongoing clinical trials (49,50). Nonhematopoietic EpoR agonists represent safer and more effective surrogates for the treatment of several diseases, such as brain and peripheral nerve injury, diabetic complications, renal ischemia, rare diseases, myocardial infarction, chronic heart disease and others.

Disclosure

The authors declare that they have no competing interests as defined by Molecular Medicine, or other interests that might be perceived to influence the results and discussion reported in this paper.

References

  1. Digicaylioglu M, et al. (1995) Localization of specific erythropoietin binding sites in defined areas of the mouse brain. Proc. Natl. Acad. Sci. U. S. A. 92: 3717–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Jelkmann W. (1992) Erythropoietin: structure, control of production, and function. Physiol. Rev. 72: 449–89.

    Article  CAS  PubMed  Google Scholar 

  3. Haase VH. (2013) Regulation of erythropoiesis by hypoxia-inducible factors. Blood Rev. 27:41–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Rotter R, et al. (2008) Erythropoietin improves functional and histological recovery of traumatized skeletal muscle tissue. J. Orthop. Res. 26:1618–26.

    Article  CAS  PubMed  Google Scholar 

  5. Jelkmann W. (2007) Erythropoietin after a century of research: younger than ever. Eur. J. Haematol. 78:183–205.

    Article  CAS  PubMed  Google Scholar 

  6. Dame C, et al. (1998) Erythropoietin mRNA expression in human fetal and neonatal tissue. Blood. 92:3218–25.

    Article  CAS  PubMed  Google Scholar 

  7. Marti HH, et al. (1996) Erythropoietin gene expression in human, monkey and murine brain. Eur. J. Neurosci. 8:666–76.

    Article  CAS  PubMed  Google Scholar 

  8. Marti HH, et al. (1997) Detection of erythropoietin in human liquor: intrinsic erythropoietin production in the brain. Kidney Int. 51:416–8.

    Article  CAS  PubMed  Google Scholar 

  9. Bernaudin M, et al. (2000) Neurons and astrocytes express EPO mRNA: oxygen-sensing mechanisms that involve the redox-state of the brain. Glia. 30:271–8.

    Article  CAS  PubMed  Google Scholar 

  10. Magnanti M, et al. (2001) Erythropoietin expression in primary rat Sertoli and peritubular myoid cells. Blood. 98:2872–4.

    Article  CAS  PubMed  Google Scholar 

  11. Dame C, et al. (2000) Erythropoietin gene expression in different areas of the developing human central nervous system. Brain Res. Dev. Brain Res. 125:69–74.

    Article  CAS  PubMed  Google Scholar 

  12. Fandrey J, Bunn HF. (1993) In vivo and in vitro regulation of erythropoietin mRNA: measurement by competitive polymerase chain reaction. Blood. 81:617–23.

    Article  CAS  PubMed  Google Scholar 

  13. Bodo E, et al. (2007) Human hair follicles are an extrarenal source and a nonhematopoietic target of erythropoietin. FASEB J. 21:3346–54.

    Article  CAS  PubMed  Google Scholar 

  14. Yasuda Y, et al. (1998) Estrogen-dependent production of erythropoietin in uterus and its implication in uterine angiogenesis. J. Biol. Chem. 273:25381–7.

    Article  CAS  PubMed  Google Scholar 

  15. Kobayashi T, et al. (2002) Epididymis is a novel site of erythropoietin production in mouse reproductive organs. Biochem. Biophys. Res. Commun. 296:145–51.

    Article  CAS  PubMed  Google Scholar 

  16. Masuda S, et al. (2000) The oviduct produces erythropoietin in an estrogen- and oxygen-dependent manner. Am. J. Physiol. Endocrinol. Metab. 278:E1038–44.

    Article  CAS  PubMed  Google Scholar 

  17. Rankin EB, et al. (2012) The HIF signaling pathway in osteoblasts directly modulates erythropoiesis through the production of EPO. Cell. 149:63–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Acs G, et al. (2001) Erythropoietin and erythropoietin receptor expression in human cancer. Cancer Res. 61:3561–5.

    CAS  PubMed  Google Scholar 

  19. Lundby C, et al. (2008) Erythropoietin receptor in human skeletal muscle and the effects of acute and long-term injections with recombinant human erythropoietin on the skeletal muscle. J. Appl. Physiol. 104:1154–60.

    Article  CAS  PubMed  Google Scholar 

  20. Broxmeyer HE. (2011) Erythropoietin surprises: an immune saga. Immunity. 34:6–7.

    Article  CAS  PubMed  Google Scholar 

  21. Nairz M, et al. (2011) Erythropoietin contrastingly affects bacterial infection and experimental colitis by inhibiting nuclear factor-kappaB-inducible immune pathways. Immunity. 34:61–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Nairz M, et al. (2012) The pleiotropic effects of erythropoietin in infection and inflammation. Microbes Infect. 14:238–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Brines M, Cerami A. (2006) Discovering erythropoietin’s extra-hematopoietic functions: biology and clinical promise. Kidney Int. 70:246–50.

    Article  CAS  PubMed  Google Scholar 

  24. Choi D, et al. (2010) Erythropoietin protects against diabetes through direct effects on pancreatic beta cells. J. Exp. Med. 207:2831–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hand CC, Brines M. (2011) Promises and pitfalls in erythopoietin-mediated tissue protection: are nonerythropoietic derivatives a way forward? J. Investig. Med. 59:1073–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. McGee SJ, et al. (2012) Effects of erythropoietin on the bone microenvironment. Growth Factors. 30:22–8.

    Article  CAS  PubMed  Google Scholar 

  27. Sytkowski AJ. (2011) The neurobiology of erythropoietin. Cell. Mol. Neurobiol. 31:931–7.

    Article  PubMed  Google Scholar 

  28. Sinclair AM, et al. (2010) Functional erythropoietin receptor is undetectable in endothelial, cardiac, neuronal, and renal cells. Blood. 115:4264–72.

    Article  CAS  PubMed  Google Scholar 

  29. Elliott S, et al. (2011) Lack of expression and function of erythropoietin receptors in the kidney. Nephrol. Dial. Transplant. 27:2733–45.

    Article  PubMed  Google Scholar 

  30. Jelkmann W, Elliott S. (2012) Erythropoietin and the vascular wall: the controversy continues. Nutr. Metab. Cardiovasc. Dis. 2012, Jun 6. [Epub ahead of print].

  31. Lundby C, Olsen NV. (2011) Effects of recombinant human erythropoietin in normal humans. J. Physiol. 589:1265–71.

    Article  CAS  PubMed  Google Scholar 

  32. Carraway MS, et al. (2010) Erythropoietin activates mitochondrial biogenesis and couples red cell mass to mitochondrial mass in the heart. Circ. Res. 106:1722–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Prunier F, et al. (2012) Single high-dose erythropoietin administration immediately after reperfusion in patients with ST-segment elevation myocardial infarction: results of the erythropoietin in myocardial infarction trial. Am. Heart J. 163:200–7. e1.

    Article  CAS  PubMed  Google Scholar 

  34. Moon C, et al. (2003) Erythropoietin reduces myocardial infarction and left ventricular functional decline after coronary artery ligation in rats. Proc. Natl. Acad. Sci. U. S. A. 100:11612–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Parsa CJ, et al. (2003) A novel protective effect of erythropoietin in the infarcted heart. J. Clin. Invest. 112:999–1007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Tamareille S, et al. (2009) Myocardial reperfusion injury management: erythropoietin compared with postconditioning. Am. J. Physiol. Heart Circ. Physiol. 297:H2035–43.

    Article  CAS  PubMed  Google Scholar 

  37. Prunier F, et al. (2007) Delayed erythropoietin therapy reduces post-MI cardiac remodeling only at a dose that mobilizes endothelial progenitor cells. Am. J. Physiol. Heart Circ. Physiol. 292:H522–9.

    Article  CAS  PubMed  Google Scholar 

  38. Cayla J, et al. (1999) Effects of recombinant erythropoietin (r-HuEPO) on plasma glucose concentration in endurance-trained rats. Acta Physiol. Scand. 166:247–9.

    Article  CAS  PubMed  Google Scholar 

  39. Ribatti D. (2010) Erythropoietin and tumor angiogenesis. Stem Cells Dev. 19:1–4.

    Article  CAS  PubMed  Google Scholar 

  40. Szenajch J, et al. (2010) 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. 1806:82–95.

    CAS  PubMed  Google Scholar 

  41. Oster HS, et al. (2012) Erythropoietin: the swinging pendulum. Leuk. Res. 36:939–44.

    Article  CAS  PubMed  Google Scholar 

  42. Shin SK, et al. (2012) Immunogenicity of recombinant human erythropoietin in Korea: a two-year cross-sectional study. Biologicals. 40:254–61.

    Article  CAS  PubMed  Google Scholar 

  43. Macdougall IC, et al. (2012) Antibody-mediated pure red cell aplasia in chronic kidney disease patients receiving erythropoiesis-stimulating agents: new insights. Kidney Int. 81:727–32.

    Article  CAS  PubMed  Google Scholar 

  44. Ponikowski P, et al. (2007) Effect of darbepoetin alfa on exercise tolerance in anemic patients with symptomatic chronic heart failure: a randomized, double-blind, placebo-controlled trial. J. Am. Coll. Cardiol. 49:753–62.

    Article  CAS  PubMed  Google Scholar 

  45. van Veldhuisen DJ, et al. (2007) Randomized, double-blind, placebo-controlled study to evaluate the effect of two dosing regimens of darbepoetin alfa in patients with heart failure and anaemia. Eur. Heart J. 28:2208–16.

    Article  PubMed  Google Scholar 

  46. Marchioli R, et al. (2013) Cardiovascular events and intensity of treatment in polycythemia vera. N. Engl. J. Med. 368:22–33.

    Article  CAS  PubMed  Google Scholar 

  47. Lubas A, et al. (2010) Renal vascular response to angiotensin II inhibition in intensive antihypertensive treatment of essential hypertension. Arch. Med. Sci. 6:533–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Banach M, et al. (2010) Controversies in hypertension treatment. Curr. Vasc. Pharmacol. 8:731–2.

    Article  CAS  PubMed  Google Scholar 

  49. Cicero AF, Ertek S. (2010) Preclinical and clinical evidence of nephro- and cardiovascular protective effects of glycosaminoglycans. Arch. Med. Sci. 6:469–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Durmaz O, et al. (2011) Recombinant human erythropoietin beta: the effect of weekly dosing on anemia, quality of life, and long-term outcomes in pediatric cancer patients. Pediatr. Hematol. Oncol. 28:461–8.

    Article  CAS  PubMed  Google Scholar 

  51. van Rijt WG, et al. (2013) ARA290, a non-erythropoietic EPO derivative, attenuates renal ischemia/reperfusion injury. J. Transl. Med. 11:9.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Joshi D, et al. (2012) Potential of novel EPO derivatives in limb ischemia. Cardiol. Res. Pract. 2012:213785.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Ahmet I, et al. (2011) A small nonerythropoietic helix B surface peptide based upon erythropoietin structure is cardioprotective against ischemic myocardial damage. Mol. Med. 17:194–200.

    Article  CAS  PubMed  Google Scholar 

  54. Swartjes M, et al. (2011) ARA290, a peptide derived from the tertiary structure of erythropoietin, produces long-term relief of neuropathic pain: an experimental study in rats and beta-common receptor knockout mice. Anesthesiology. 115:1084–92.

    Article  CAS  PubMed  Google Scholar 

  55. Heij L, et al. (2013) Safety and efficacy of ARA 290 in sarcoidosis patients with symptoms of small fiber neuropathy: a randomized, doubleblind pilot study. Mol. Med. 18:1430–6.

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Fabian Sanchis-Gomar.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, and provide a link to the Creative Commons license. You do not have permission under this license to share adapted material derived from this article or parts of it.

The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this license, visit (https://doi.org/creativecommons.org/licenses/by-nc-nd/4.0/)

Reprints and permissions

About this article

Cite this article

Sanchis-Gomar, F., Perez-Quilis, C. & Lippi, G. Erythropoietin Receptor (EpoR) Agonism Is Used to Treat a Wide Range of Disease. Mol Med 19, 62–64 (2013). https://doi.org/10.2119/molmed.2013.00025

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2119/molmed.2013.00025

Keywords