Skip to main content
Medical Science Monitor: International Medical Journal of Experimental and Clinical Research logoLink to Medical Science Monitor: International Medical Journal of Experimental and Clinical Research
. 2011 Nov 1;17(11):RA240–RA247. doi: 10.12659/MSM.882037

Erythropoietin update 2011

Mariusz Kowalczyk 1, Maciej Banach 2,, Dimitri P Mikhailidis 3, Jacek Rysz 1
PMCID: PMC3539497  PMID: 22037755

Summary

Traditionally, erythropoietin (EPO) is described as a hematopoietic cytokine, regulating proliferation and differentiation and survival of the erythroid progenitors. The recent finding of new sites of EPO production and the wide spread distribution of EPO receptors (EPO-R) on endothelial cells, cardiomyocytes, renal cells as well as the central and peripheral nervous system raised the possibility that EPO may exert pleiotropic actions on several targets. Indeed studies (mainly preclinical) have documented protective, non-hematopoietic, abilities of EPO in a variety of tissue. However, the data obtained from clinical studies are more skeptical about these properties. This article provides a comprehensive overview of EPO and its derivatives.

Keywords: erythropoietin, erythropoietin-receptor, erythropoiesis-stimulating agents, anemia

Background

Erythropoietin (EPO) is widely used for treatment of anemia in patients with chronic kidney disease (CKD) and in cancer patients receiving chemotherapy [1,2]. The biggest advantage of EPO administration is increased quality of life (QoL). In many studies EPO has been shown to posses properties beyond its traditional role as a hematopoietic cytokine (Table 1) [13]. EPO may directly and indirectly affect different cells, enhance antioxidant enzyme production, antagonize the cytotoxic action of glutamate, metabolize free radicals, normalize cerebral blood flow, affect neurotransmitter release, stimulate neoangiogenesis, modify inflammation and promote endothelial progenitor cell proliferation and differentiation [1,4,5]. On the other hand over the last decade several studies have reported increased mortality and cardiovascular events when erythropoiesis-stimulating agents (ESAs) were administered to CKD patients [4]. Moreover, a number of promising outcomes mediated by EPO, in preclinical studies (in vitro and in vivo), were not confirmed in clinical studies [5,6].

Table 1.

Non-hematopoietic mechanism of tissue protection by erythropoietin.

Type of tissue protection Possible mechanisms of action
Cardioprotection Reduces apoptosis, modifies inflammation, increases endothelial nitric oxide synthase, stimulates angiogenesis, promotes endothelial progenitor cells proliferation and differentiation, enhances antioxidant enzyme expression and reduces the rate of free radical production, improves cardiac function reflected by increased ventricular developed pressure (dP/dtmax) and relaxation (dP/dtmin), reduction of left ventricular mass index and increased ejection fraction.
Neuroprotection Antagonizes glutamate’s cytotoxic action, normalizes cerebral blood flow, stimulates neoangiogenesis, promotes endothelial progenitor cells proliferation and differentiation, affects neurotransmitter release, modifies inflammation and immune response, stimulates non-differentiated Schwann cells to proliferate, reduces apoptosis, enhances antioxidant enzyme expression and reduces the rate of free radical production.
Renoprotection Reduction of apoptosis and inflammatory response, promotion of vascular repair, increasing the proliferation of tubular cells, enhances antioxidant enzyme expression and reduces the rate of free radical production. Possible autocrine-paracrine action of erythropoietin within the kidney mediates cytoprotection.

This article provides an overview of EPO and its derivatives as a cardiovascular, neuroprotective or renoprotective agent. We also consider the relationship of EPO with malignancy and inflammation and discuss the common side effects associated with its administration.

EPO and EPO-receptors

EPO was purified in 1977, the gene was cloned in 1985 and the EPO receptor was cloned in 1989 [5]. Recombinant human EPO (rHuEPO) has become a successful clinical application of recombinant DNA technology [7]. There are a few forms of rHuEPO: EPOetin-α, EPOetin-β, EPOetin-δ, a long-acting analogue darbepoetin α (the second generation) and a continuous EPO receptor activator (CERA, the third generation). In humans, EPO is produced by peritubular fibroblast-like type 1 interstitial cells located in the renal cortex and outer medulla [7]. The kidney is a major producer of EPO in adults but not the only one; 10% of the circulating EPO originates from non-renal tissue [8]. EPO is expressed in the liver, brain, spleen, lung and testis. During fetal development, the main producer of EPO is the liver. A comprehensive study indicated that a liver to kidney shift in EPO production occurs in late gestation; the molecular mechanisms underlying this shift are still obscure. Moreover hepatocytes remain the primary cells responsible for extrarenal EPO synthesis [9,10].

EPO is a 34,000-da, 165 amino acid glycoprotein that is synthesized mainly under hypoxic condition [9]. In fact, EPO is the only hematopoietic growth factor whose production is regulated by hypoxia. The induction of EPO synthesis by low oxygen levels lead to discovery a widespread system of hypoxia-inducible factors (HIFs). HIF-1 and HIF-2 are transcriptional activators each composed of an α and β subunit [11]. The α subunit is a cytoplasmic protein containing an oxygen degradation domain (ODDD) and a transactivation domain (TAD) [11]. The β subunit is located in the nucleus. HIF-3 is a likely inhibitor of EPO gene transcription. The primary effect of EPO on the red blood cell (RBC) line, especially the colony forming units-erythroid (CFU-E), is the promotion of RBC survival by protecting these cells from apoptosis [11].

EPO receptors belong to the super-family of cytokine receptors and can be located on the plasma membrane of bone marrow erythroid progenitor cells, cardiomyocytes, cardiac fibroblasts, endothelial and vascular smooth muscle cells, gastric cells, retinal and prostate cells, human hair follicles [8] and auditory hair cells in the inner ear [1215]. EPO-R binding triggers at least 3 intracellular signaling cascades: (1) Janus tyrosine kinase 2 (JAK2) a cytoplasmic tyrosine kinase that phosphorylates tyrosine residues itself and provides docking sites for signal transducer and activator of transcription 5 (STAT5), (2) phosphatidylinositol-3 kinase (PI3K), and, (3) RAS/mitogen-activated protein kinase (MAPK) [16]. STAT5 and MAPK induce transcription of target genes involved, mainly with inhibition of apoptosis with cell proliferation. PI3K inhibits apoptosis by activating its downstream effector Akt, and the activation of Akt by EPO also induces activation by phosphorylation of endothelial nitric oxide synthase (eNOS) and prevents neointimal hyperplasia [16].

EPO and Cardiovascular Protection

The prevalence of anemia among patients with chronic heart failure (CHF) is estimated over 20% and has a multifactorial etiology [1719]. Anemia has been identified as a strong prognostic factor associated with poor outcomes (with worsening symptoms and increased mortality) among patients with CHF [2022]. In a systematic review and meta-analysis of randomized trials, the authors evaluated the efficacy and safety associated with the use of ESAs for correcting anemia in patients with CHF [23]. In patients with CHF and anemia ESAs compared with control, were associated with a significant reduction in CHF-related hospitalizations (OR=0.41; 95% Confidence Intervals [CI] 0.24–0.69) [23]. Moreover, ESA treatment was associated with improved quality of life and left ventricular ejection fraction, lower brain-natriuretic peptide (BNP) levels and improved exercise tolerance test performance. However, the effect of ESAs on mortality was inconclusive (OR=0.60; 95%CI 0.51–1.42), but available date could suggest that ESAs have a favorable effect on all-cause mortality [23]. The impact of ESAs on morbidity and mortality in patients with HF has been evaluated in the post-TREAT meta-analysis [24]. The use of ESAs to treat anemia in patients with HF was associated with neutral effect on both mortality (RR=1.03; 95%CI 0.89–1.12; p=0.68) and non-fatal HF events (RR=0.95; 95%CI: 0.82–1.10; p=0.46) [24].

Patients with CKD are more at risk of cardiovascular events, particularly young dialysis patient, whose mortality is up to 100 times greater than for the general population [2527]. In dialysis patient, cardiomyopathy predisposes to HF and death [27]. In contrast, a reduction in left ventricular mass index (LVMi) is associated with the increase in both all-cause and cardiovascular survival rate [28]. The changes in the LVMi among anemic CKD and end-stage renal disease patients treated with recombinant human erythropoietin were evaluated in a systematic review of papers published between 1990 and 2007 [28]. This meta-analysis revealed, that in patients with severe anemia, defined as mean baseline hemoglobin (Hb) <10 g/dl, a significant decrease in LVMi was observed. There was no such significant beneficial impact on LVMi in the moderate anemia group with target Hb above 12 g/dl. This study despite its limitations (e.g. potential for development of EPO-induced hypertension and lack of control group) supports EPO treatment of severe anemia in CKD patients [28].

In a Pilot Evaluation of the Long-term Effect of Combined Therapy With Intravenous Iron Sucrose and Erythropoietin in Elderly Patients With Advanced Chronic Heart Failure and Cardio-Renal Anemia Syndrome study, combined therapy with intravenous (IV) iron and rHuEPO showed an increase Hb level, reduction of N-terminal pro-B-type natriuretic peptide (NT-proBNP), improvement of functional capacity and cardiovascular hospitalization in elderly patients with advanced CHF and cardio-renal anemia syndrome with mild to moderate renal dysfunction [29].

ESAs in Malignancy

Anemia is a frequent complication in cancer patients and it is one of the main causes of cancer-related fatigue [30,31]. According to some authors up to 40% of cancer patients are anemic at diagnosis and this frequency is increased following chemotherapy [32]. Before the era of ESAs, oncologists relied on transfusions to correct anemia and to improve QoL [32]. Furthermore, tumor responsiveness to chemotherapy and radiotherapy seems to be weakened in anemic patients [32]. The introduction ESAs offered an alternative method to transfusion. Clinical trials with ESAs have reported an improved QoL and decreased treatment-related anemia (including numbers of blood transfusions) [33]. Other studies suggested an improvement in survival outcome of cancer patients that received rHuEPO for anemia [33].

However, rHuEPO as treatment for cancer-related anemia could also be harmful. In cystic renal disease increased endogenous production of EPO is associated with a higher incidence of cancer [34]. Von Hippel-Lindau (VHL) disease is another example when loss of ability to degrade HIF that regulates EPO synthesis is responsible for a high incidence of spontaneous renal cell cancer [3436]. Based on clinical trials rHuEPO was suspected to trigger tumor progression leading to decrease survival [37,38]. In the ENHANCE trial, patients with advanced head and neck cancer treated with radiotherapy had a higher risk for loco-regional progression when also receiving EPOetin β [38]. The BEST trial with metastatic breast cancer patients, who were receiving chemotherapy, was terminated prematurely because of a significant reduction of survival in women receiving ESAs [39]. A study of ESAs in non-small-cell lung cancer patients receiving palliative treatment was also terminated, because of unexpected worse survival in the ESAs arm [40]. Another problem was an increased risk of venous thromboembolism (VTE) in cancer patients following ESAs treatment. The association between cancer and increased risk of VTE is well established [41]. VTE could be an explanation of worse overall survival. However, a number of trials did not report increased VTE events. On the other hand, a number of clinical studies indicate better survival rate in cancer patients who receive anti-coagulants [4043]. Another explanation is the binding of tumour cell erythropoietin receptors by ESAs which could stimulate tumour cell growth, decrease cell apoptosis or increase resistance to therapy [4447]. The activation of EPO-R is similar to cell activation by growth receptors [44]. This activation depends on tyrosine kinase that phosphorylates tyrosine residues itself and provides docking sites for signal transducers [44]. Tyrosine kinase activity is important in many growth factor receptors and in oncogenes, and because of its ability to stimulate mitogenic potential it may play an important role in this mechanism. EPO-R or EPO-R mRNA are present on some cancer cells but do not necessarily indicate receptor functionality [44]. However, some researcher suggests that EPO-R expression like tumour size and lymphovascular invasion may act as a prognostic factor [48]. A very interesting hypothesis suggests that ESAs may directly stimulate tumour growth through activation of the coagulation cascade and subsequent stimulation of angiogenesis [46,48]. This hypothesis is supported by clinical evidence both of increased VTE rates and increased coagulability with ESAs exposure [46]. Pathological angiogenesis could be stimulated directly by EPO via recruitment endothelial progenitors cells or by the thrombosis pathways [46,49]. These processes are numerous and complicated and are an active area of research. In 2007 the American Society of Clinical Oncology/American Society of Hematology (ASCO/ASH) recommended against the use of EPO in anemic cancer patient not receiving chemotherapy with the exception of the low-risk myelodysplastic syndrome [50].

EPO and the Central and Peripheral Nervous System

Both EPO and its receptor are present in the central and peripheral nervous system [51]. Moreover, analysis of spinal fluid revealed a significant increase in EPO following its administration confirming that EPO crosses the blood-brain barrier [52]. In vitro, EPO protects nerve cells from hypoxia-induced glutamate toxicity, which is the main cause of hypoxia-induced nerve cell death [51]. Furthermore, in a multiple sclerosis animal model, the systematic administration of EPO alpha reduced the immune response and the inflammatory reaction including enhanced nerve recovery after spinal cord injury [53]. EPO, both in animal and human models, reduced the level of impairment after cerebral ischemia [53]. EPO and EPO-R play an essential role in neurogenesis, especially during embryonic neurogenesis [54]. In the peripheral nervous system, EPO is produced in the bodies and axon of normal ganglions in the rat dorsal root and an increased EPO level are seen in Schwann cells after peripheral nerve injury [55]. EPO stimulates non-differentiated Schwann cells to proliferate [55]. Experiments have shown that systemic administration of rHuEPO reduces apoptosis of dorsal root ganglion cells and contributes the recovery of mechanical allodynia following nerve injury [53]. EPO enhances antioxidant enzyme expression and reduces the rate of free radical production [5355]. EPO-R and dismutase peroxide (SOD) are activated via the same metabolic pathways [53]. Akt activation by EPO inhibits various metabolic pathways that are related to cell death, such as those related to glycogen synthetase kinase 3β (GSK3β), Bcl-2-associated cell death promoting protein (BAD) and caspase-9 [5356]. In a systematic review and meta-analysis of EPO in experimental stroke, 19 studies involving 346 animals for infarct size and 425 animals for neurobehavioral were evaluated [57]. EPO improved infarct size by 30.0% (95%CI: 21.3–38.8) and neurobehavioral outcome by 39.8% (95%CI: 33.7–45.9). The results are promising but when the impact of common sources of bias are considered, this efficacy falls, suggesting that the potential benefit may be overestimated [57]. The most recent clinical study reported that recombinant EPO failed to protect from damage induced by ischemic stroke [58].

EPO and Renoprotection

The discovery of EPO-R mRNA and EPO-R in kidney suggested that EPO may act as a protective agent in acute kidney injury (AKI). EPO-R is expressed by mesangial cells, epithelial cells of the proximal tubule and distal tubule and the collecting duct [59]. In cultured renal cells, EPO signaling occurs through the JAK/STAT5 pathway and results in increase DNA synthesis and proliferation [60]. It is hypothesized that autocrine-paracrine action within the kidney mediates cytoprotection [60]. AKI induces apoptosis and inflammatory response but EPO decreases this processes by anti-apoptosis mechanisms, promotion of vascular repair and increasing the proliferation of tubular cells [61,62]. There is evidence that in ischemic AKI, renal expression of EPO is significantly decreased whereas EPO-R level stay unchanged thus a cytoprotective effect maybe only possible by administration of exogenous EPO [60]. Studies performed on rodents revealed a protective effect of EPO/ESAs in the experimental setting of ischemic, septic AKI or induced by hemorrhagic shock, cisplatin or radio contrast media [60]. Moreover, the cytoprotective effect was achievable both 30 min and 6 h post ischemic kidney injury compared with the respective control group [63]. In most of these studies EPO/ESAs had no effect on Hb concentration within the time frame of the studies [6063]. Therefore, renoprotection may depend on mechanisms other than the hematopoietic properties of EPO-R [63].

However, in a recently published study – Early intervention with erythropoietin does not affect the outcome of acute kidney injury (the EARLYARF trial) [64], investigators performed a double-blind placebo-controlled trial to study whether early treatment (within 6 h of injury) with high-dose EPO (up to 50,000 U) could prevent the development of AKI in intensive care unit (ICU) patients. High dose of EPO did not alter the outcome of patients receiving EPO compared with placebo. There was no difference in the incidence of EPO-specific adverse events end early intervention with high-dose EPO was safe. However, this study had some limitations – a composite of 2 biomarkers (the proximal tubular brush border enzymes gamma-glutamyl transpeptidase and alkaline phosphatase) was insufficient for risk stratification in a patient population with a heterogeneous onset of AKI [64]. Therefore, further work is needed [65].

Few studies assessed the renoprotective effects of rHuEPO in CKD. The explanation could be that therapeutic efforts in CKD patients were made only to correct anemia and the putative hypoxic renal tissue damage as a result of anemia. Some results from recently published large trials in patients with CKD revealed no beneficial effect on progression CKD [4]. However, study by Gouva et al. in which rHuEPO therapy was started in CKD patients with only mild-to-moderate anemia, and the anemia was corrected only to subnormal levels over a period of 6 months [66]. The authors observed significantly reduced progression and significantly less need for renal replacement therapy in the group of rHuEPO-treated patients [66].

EPO and Inflammation

Anemia is very common in chronic inflammatory diseases [4]. Pro-inflammatory cytokines such as interferon (IFN)-γ, tumor necrosis factor (TNF)-α, interleukin (IL)-1 and −6β are responsible for suppression of EPO production both in vitro and in vivo[6771]. But EPO production in patients with cancer disease is not diminished enough to cause anemia, is not the only explanation of this mechanism [67]. Moreover IFN-γ, TNF-α, TRAIL and IL-1β are cytokines responsible for inhibition of the proliferation and differentiation of erythroid progenitor cells [71]. Therefore, a disturbance of erythropoiesis is most likely due to apoptosis induction, cell growth inhibition, EPO-R down regulation as a result of locally increased cytokines and as well due to impaired iron metabolism [72]. The role of reactive oxygen species (ROS) is even more complicated since they may either trigger or prevent hematopoietic proliferation and differentiation [72,73].

EPO and Safety

EPO has been abused in sport, particularly in disciplines requiring an adequate supply of oxygen to muscles. The first suspected cases of doping with EPO was in the 1980s; since then doping with EPO has been reported many times by the World Anti-Doping Agency [74]. Doping with EPO is associated with serious adverse side-effect beginning from hypertension, headaches and an increased number of thrombotic events and death [74]. Moreover, EPO withdrawal may be complicated in neocytolysis – hemolysis of young red blood cells in the presence of increased hematocrit [74]. The great safety concern was brought to the light when The Normal Hematocrit Study provided one of the first suggestions that the use of ESAs to raise Hb concentrations into the normal range could cause harm [75]. This concept was presented by large observational trials, all of them have shown an increase in mortality related to higher Hb values in kidney patients. The CHOIR has fuelled the debate on the safety of rHuEPO [76]. As a result, the Food and Drug Administration (FDA) has recommended the lowest possible dose to slowly raise the Hb concentration to the lowest level that will avoid the need for a blood transfusion [77]. However subsequent analysis of the trial results revealed that the cause of the worse outcomes was not the Hb level but the high ESA dose [77]. Solomon et al. observed that patients with a poor response to darbepoetin α who had subsequently higher doses of this drug to meet target Hb levels, as compared with those with a better response, had higher rates of the composite cardiovascular end point (adjusted HR 1.31; 95%Cl: 1.09–1.59) or death (adjusted HR 1.41; 95%CI: 1.12–1.78) [78].

EPO and Hypertension

The most common side effect of rHuEPO therapy seems to be hypertension which may occur even in healthy subjects [74,79,80]. rHuEPO increases peripheral vascular resistance and decreases cardiac output [4,74]. All of that is caused by increases in endothelins, angiotensin, impaired vascular endothelial relaxation, altered calcium levels in vascular smooth muscle cells and the release of serotonin by platelets [81]. EPO has direct vasoconstrictor effects in isolated renal resistance vessels [82]. Production of endogenous EPO is as well regulated by the renin-angiotensin-system [83]. When angiotensin II are given to normal human subjects, plasma EPO levels increased [83]. A similar situation is observed when inhibition of angiotensin enzyme decrease plasma EPO [84,85]. Angiotensin II stimulates growth factors similar to insulin by inducing tyrosine-kinase receptors in vascular smooth muscle cells, which in turn produces an increase in vascular intimal hyperplasia [86]. EPO and angiotensin II seem to be similar to other cytokines that activate tyrosine-kinase receptor and that may be responsible for harmful effects of both hypertension and vascular disease progress. The main EPO mechanism for the raising the hematocrit is an increase in RBC mass but a decrease plasma volume also occurs [87]. However, the link between hypertension and an increase in hematocrit has not be proven with certainty, and arterial hypertension may occur independently of EPO’s hematopoietic effect [8890].

EPO and Thrombosis

Some trials and meta-analyses reported an increased risk of VTE following ESAs treatment. Before the use of EPO to treat anemia the incidence of deep vein thrombosis (DVT) and pulmonary embolism (PE) was rare in patients with CKD [81,91]. There is evidence suggesting that rHuEPO enhances procoagulant pathways, which can cause adverse effects and, therefore, potentially limit the clinical use of EPO [92]. rHuEPO increases platelet aggregability and may decreased proteins C and S levels [92]. In hemodialysis patients rHuEPO raises the platelet count and mean platelet volume but did not change the numbers of surface platelet receptors [93]. The platelet aggregation can be reversed by using aspirin but taking aspirin cannot prevent vascular access thrombosis in hemodialysis patients [93]. Vascular access thrombosis is associated with intimal hyperplasia and smooth muscle cell proliferation [94]. Another hypothesis for increased thrombosis in heamodialysis patients could be thrombocytosis due to the presence of iron deficiency caused by ESA [95]. This hypothesis is supported by a study where the use of IV iron reduced the risk of thromboembolic events by 40% [96]. Another possible explanation of increased adverse events is that chronic administration of EPO may enhance angiogenesis in the atherosclerotic plaque by increased vascular endothelial growth factor production with subsequent plaque rupture and acute coronary syndrome or stroke. From the clinical perspective, another possible explanation for high incidence of thrombosis could be viscosity of the blood, which leads to a risk of vascular thrombosis [97]. By taking into account blood viscosity as a main determinant of the work of the heart, and elevated blood viscosity appears to be both a strong predictor of cardiovascular disease and an important pathophysiological factor in the development of atherothrombosis [98].

Conclusions

The role of ESAs and future indications are unclear. Tissue protection after ischemia and injury has been found in the brain, heart and kidney [99101]. Benefits include an increased Hb to the recommended level in anemic patients with CKD or HF or both or in the palliative chemotherapy setting remains. An increase QoL has been reported almost in every study with ESAs. However, QoL was not generally designed as a specific end-point. Furthermore, analysis of the current available data shows major inhomogeneities in the tools used for assessment of QoL and in data reporting which suggest that only partial correction of anemia with EPO may improve QoL [102]. On the other hand a better correction of anemia with higher Hb target is associated with increased risk for stroke, hypertension, vascular access thrombosis compared with a lower Hb target [103].

Studies have demonstrated a decrease survival outcome of cancer patients that received rHuEPO for anemia. This caused great concern regarding patients on hemodialysis who have previous cancer diagnoses. However, we know that the doses of ESAs have more than tripled in the USA since ESAs were introduced and that cancer specific mortality rates remained stable among US hemodialysis patients between 1995 and 2005 [25]. Despite all the promising outcomes in numerous preclinical studies (in vitro and in vivo), EPO use as a neuroprotective drug failed in clinical studies. However, this does not mean that the neuroprotective abilities of EPO are wrong but it means that we have to be more critical in evaluating the future use of EPO [104106]. To minimize possible side effect of ESAs therapy, a greater understanding of the physiology of this molecule and its receptors are required with the possible alternative method of administration. Perhaps the new generation of ESAs (asialo EPO and carbamylated EPO), without the erythropoietic activity of EPO, while preserving its tissue protective properties, will provide better outcome in ongoing clinical trials [107,108].

Footnotes

Conflict of interest

None.

Source of support: Self financing

References

  • 1.Malyszko J, Zbroch E, Malyszko J, et al. The cardio-renal-anaemia syndrome predicts survival in peritoneally dialyzed patients. Arch Med Sci. 2010;6:539–44. doi: 10.5114/aoms.2010.14465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kowalczyk M, Banach M, Rysz J. Ferumoxytol: a new era of iron deficiency anemia treatment for patients with chronic kidney disease. J Nephrol. 2011 doi: 10.5301/jn.5000025. [DOI] [PubMed] [Google Scholar]
  • 3.Elshamaa MF, Sabry S, Mokhtar I, et al. Aluminium and lead abnormalities in children on haemodialysis: relationship with some medications. Arch Med Sci. 2010;6:420–29. doi: 10.5114/aoms.2010.14266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Drueke TB, Locatelli F, Clyne N, et al. Normalization of hemoglobin level in patients with chronic kidney disease and anemia. N Engl J Med. 2006;355:2071–84. doi: 10.1056/NEJMoa062276. [DOI] [PubMed] [Google Scholar]
  • 5.Jelkmann W. Erythropoietin: structure, control of production, and function. Physiol Rev. 1992;72:449–89. doi: 10.1152/physrev.1992.72.2.449. [DOI] [PubMed] [Google Scholar]
  • 6.Tkocz M, Kupajski M, Duda D, Witosińska-Walica A. Spontaneous subcapsular kidney haemorrhage – the first symptom of renal cell carcinoma. Arch Med Sci. 2009;5:107–10. [Google Scholar]
  • 7.Bachmann S, Le Hir M, Eckardt KU. Co-localization of erythropoietin mRNA and ecto-5′-nucleotidase immunoreactivity in peritubular cells of rat renal cortex indicates that fibroblasts produce erythropoietin. J Histochem Cytochem. 1993;41:335–41. doi: 10.1177/41.3.8429197. [DOI] [PubMed] [Google Scholar]
  • 8.Fried W, Kilbridge T, Krantz S, et al. Studies on extrarenal erythropoietin. J Lab Clin Med. 1969;73:244–48. [PubMed] [Google Scholar]
  • 9.Rankin EB, Biju MP, Liu Q, et al. Hypoxia-inducible factor-2 (HIF-2) regulates hepatic erythropoietin in vivo. J Clin Invest. 2007;117:1068–77. doi: 10.1172/JCI30117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Nair DR, Mehta S, Mikhailidis DP. Assessing renal function – searching for the perfect marker continues! Arch Med Sci. 2011;7:565–67. doi: 10.5114/aoms.2011.24120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Haase VH. Hypoxic regulation of erythropoiesis and iron metabolism. Am J Physiol Renal Physiol. 2010;299:F1–F13. doi: 10.1152/ajprenal.00174.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Jones SS, D’Andrea AD, Haines LL, Wong GG. Human erythropoietin receptor: cloning, expression, and biologic characterization. Blood. 1990;76:31–35. [PubMed] [Google Scholar]
  • 13.Bodo E, Kromminga A, Funk W, et al. Human hair follicles are an extrarenal source and a nonhematopoietic target of erythropoietin. FASEB Journal. 2007;21:3346–54. doi: 10.1096/fj.07-8628com. [DOI] [PubMed] [Google Scholar]
  • 14.Lubas A, Żelichowski G, Próchnicka A, et al. Renal autoregulation in medical therapy of renovascular hypertension. Arch Med Sci. 2010;6:912–18. doi: 10.5114/aoms.2010.19301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Monge Naldi A, Gassmann M, Bodmer D. Erythropoietin but not VEGF has a protective effect on auditory hair cells in the inner ear. Cell Mol Life Sci. 2009;66:3595–99. doi: 10.1007/s00018-009-0144-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Urao N, Okigaki M, Yamada H, et al. Erythropoietin-mobilized endothelial progenitors enhance reendothelialization via Akt-endothelial nitric oxide synthase activation and prevent neointimal hyperplasia. Circ Res. 2006;98:1405–13. doi: 10.1161/01.RES.0000224117.59417.f3. [DOI] [PubMed] [Google Scholar]
  • 17.Tanner H, Moschovitis G, Kuster GM, et al. The prevalence of anemia in chronic heart failure. Int J Cardiol. 2002;86:115–21. doi: 10.1016/s0167-5273(02)00273-5. [DOI] [PubMed] [Google Scholar]
  • 18.Mujib M, Desai R, Levitan EB, et al. Prospective population studies of incident heart failure without data on baseline left ventricular ejection fraction. Arch Med Sci. 2010;6:686–88. doi: 10.5114/aoms.2010.17081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Tang YD, Katz SD. Anemia in chronic heart failure: prevalence, etiology, clinical correlates, and treatment options. Circulation. 2006;113:2454–61. doi: 10.1161/CIRCULATIONAHA.105.583666. [DOI] [PubMed] [Google Scholar]
  • 20.Ezekowitz JA, McAlister FA, Armstrong PW. Anemia is common in heart failure and is associated with poor outcomes: insights from a cohort of 12 065 patients with new-onset heart failure. Circulation. 2003;107:223–25. doi: 10.1161/01.cir.0000052622.51963.fc. [DOI] [PubMed] [Google Scholar]
  • 21.Anand I, McMurray JJ, Whitmore J, et al. Anemia and its relationship to clinical outcome in heart failure. Circulation. 2004;110:149–54. doi: 10.1161/01.CIR.0000134279.79571.73. [DOI] [PubMed] [Google Scholar]
  • 22.Ngo K, Kotecha D, Walters JA, et al. Erythropoiesis-stimulating agents for anaemia in chronic heart failure patients. Cochrane Database Syst Rev. 2010;(1):CD007613. doi: 10.1002/14651858.CD007613.pub2. [DOI] [PubMed] [Google Scholar]
  • 23.Lawler PR, Filion KB, Eisenberg MJ. Correcting Anemia in Heart Failure: The Efficacy and Safety of Erythropoiesis-Stimulating Agents. J Card Fail. 2010;16:649–58. doi: 10.1016/j.cardfail.2010.03.013. [DOI] [PubMed] [Google Scholar]
  • 24.Desai A, Lewis E, Solomon S, et al. Impact of erythropoiesis-stimulating agents on morbidity and mortality in patients with heart failure: an updated, post-TREAT meta-analysis. Eur J Heart Fail. 2010;12:936–42. doi: 10.1093/eurjhf/hfq094. [DOI] [PubMed] [Google Scholar]
  • 25.Foley RN, Roberts TL, Liu JN, et al. Mortality from Cancer among US Hemodialysis Patients, 1995–2005. Am J Nephrol. 2010;31:518–26. doi: 10.1159/000303754. [DOI] [PubMed] [Google Scholar]
  • 26.Sethi A, Arora RR. Ambulatory blood pressure as a predictor of cardiovascular risk. Arch Med Sci. 2009;5:3–9. [Google Scholar]
  • 27.Banach M, Rysz J. Current problems in hypertension and nephrology. Expert Opin Pharmacother. 2010;11:2575–78. doi: 10.1517/14656566.2010.522569. [DOI] [PubMed] [Google Scholar]
  • 28.Parfrey PS, Lauve M, Latremouille-Viau D, Lefebvre P. Erythropoietin Therapy and Left Ventricular Mass Index in CKD and ESRD Patients: A Meta-Analysis. Clin J Am Soc Nephrol. 2009;4:755–62. doi: 10.2215/CJN.02730608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Comin-Colet J, Ruiz S, Cladellas M, et al. A Pilot Evaluation of the Long-term Effect of Combined Therapy With Intravenous Iron Sucrose and Erythropoietin in Elderly Patients With Advanced Chronic Heart Failure and Cardio-Renal Anemia Syndrome: Influence on Neurohormonal Activation and Clinical Outcomes. J Card Fail. 2009;15:727–35. doi: 10.1016/j.cardfail.2009.05.010. [DOI] [PubMed] [Google Scholar]
  • 30.Stasi R, Abriani L, Beccaglia P, et al. Cancer-related fatigue: evolving concepts in evaluation and treatment. Cancer. 2003;98:1786–801. doi: 10.1002/cncr.11742. [DOI] [PubMed] [Google Scholar]
  • 31.Smolińska K, Paluszkiewicz P. Risk of colorectal cancer in relation to frequency and total amount of red meat consumption. Systematic review and meta-analysis. Arch Med Sci. 2010;6:605–10. doi: 10.5114/aoms.2010.14475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Knight K, Wade S, Balducci L. Prevalence and outcomes of anemia in cancer: a systematic review of the literature. Am J Med. 2004;116:11S–26S. doi: 10.1016/j.amjmed.2003.12.008. [DOI] [PubMed] [Google Scholar]
  • 33.Bohlius J, Wilson J, Seidenfeld J, et al. Erythropoietin or darbepoetin for patients with cancer. Cochrane Database Syst Rev. 2006;3:CD003407. doi: 10.1002/14651858.CD003407.pub4. [DOI] [PubMed] [Google Scholar]
  • 34.Diskin CJ, Stokes TJ, Dansby LM, et al. Beyond Anemia: The Clinical Impact of the Physiologic Effects of Erythropoietin. Seminars in Dialysis. 2008;21:447–54. doi: 10.1111/j.1525-139X.2008.00443.x. [DOI] [PubMed] [Google Scholar]
  • 35.Zemanek D, Veselka J, Adla T, et al. Uncommon cause of obstruction in the left ventricular outflow tract by a metastasis of adenocarcinoma. Arch Med Sci. 2010;6:981–83. doi: 10.5114/aoms.2010.19313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Chandra M, Miller ME, Garcia JF, et al. Serum immunoreactive erythropoietin levels in patients with polycystic kidney disease as compared with other hemodialysis patients. Nephron. 1985;39:26–29. doi: 10.1159/000183332. [DOI] [PubMed] [Google Scholar]
  • 37.Leyland-Jones B. Breast cancer trial with erythropoietin terminated unexpectedly. Lancet Oncol. 2003;4:459–60. doi: 10.1016/s1470-2045(03)01163-x. [DOI] [PubMed] [Google Scholar]
  • 38.Henke M, Laszig R, Rube C, et al. Erythropoietin to treat head and neck cancer patients with anaemia undergoing radiotherapy: randomised, double-blind, placebo-controlled trial. Lancet. 2003;362:1255–60. doi: 10.1016/S0140-6736(03)14567-9. [DOI] [PubMed] [Google Scholar]
  • 39.Leyland-Jones B, Semiglazov V, Pawlicki M, et al. Maintaining normal hemoglobin levels with epoetin alfa in mainly nonanemic patients with metastatic breast cancer receiving first-line chemotherapy: a survival study. J Clin Oncol. 2005;23:5960–72. doi: 10.1200/JCO.2005.06.150. [DOI] [PubMed] [Google Scholar]
  • 40.Wright JR, Ung YC, Julian JA, et al. Randomized, double-blind, placebo-controlled trial of erythropoietin in non-small-cell lung cancer with disease-related anemia. J Clin Oncol. 2007;25:1027–32. doi: 10.1200/JCO.2006.07.1514. [DOI] [PubMed] [Google Scholar]
  • 41.Kroger K, Weiland D, Ose C, et al. Risk factors for venous thromboembolic events in cancer patients. Ann Oncol. 2006;17:297–303. doi: 10.1093/annonc/mdj068. [DOI] [PubMed] [Google Scholar]
  • 42.Kakkar AK. Low-molecular-weight heparins: beyond thrombosis in the management of the cancer patient. Semin Thromb Hemost. 2003;29(Suppl 1):13–15. doi: 10.1055/s-2003-45412. [DOI] [PubMed] [Google Scholar]
  • 43.Kakkar AK, Levine MN, Kadziola Z, et al. Low molecular weight heparin, therapy with dalteparin, and survival in advanced cancer: the fragmin advanced malignancy outcome study (FAMOUS) J Clin Oncol. 2004;22:1944–48. doi: 10.1200/JCO.2004.10.002. [DOI] [PubMed] [Google Scholar]
  • 44.Solar P, Koval J, Mikes J, et al. Erythropoietin inhibits apoptosis induced by photodynamic therapy in ovarian cancer cells. Mol Cancer Ther. 2008;7:2263–71. doi: 10.1158/1535-7163.MCT-08-0483. [DOI] [PubMed] [Google Scholar]
  • 45.Yildiz Y, YaylIm-Eraltan I, Arikan S, et al. Is there any correlation between TNF-related apoptosis-inducing ligand (TRAIL) genetic variants and breast cancer? Arch Med Sci. 2010;6:932–36. doi: 10.5114/aoms.2010.19304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Barbera L, Thomas G. Erythropoiesis stimulating agents, thrombosis and cancer. Radiotherapy and Oncology. 2010;95:269–76. doi: 10.1016/j.radonc.2010.02.008. [DOI] [PubMed] [Google Scholar]
  • 47.Bielecka-Dąbrowa A, Hannam S, Rysz J, Banach M. Malignancy-associated dyslipidemia. Open Cardiovasc Med J. 2011;5:35–40. doi: 10.2174/1874192401105010035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Leo C, Horn LC, Rauscher C, et al. Expression of erythropoietin and erythropoietin receptor in cervical cancer and relationship to survival, hypoxia, and apoptosis. Clin Cancer Res. 2006;12:6894–900. doi: 10.1158/1078-0432.CCR-06-1285. [DOI] [PubMed] [Google Scholar]
  • 49.Heeschen C, Aicher A, Lehmann R, et al. Erythropoietin is a potent physiologic stimulus for endothelial progenitor cell mobilization. Blood. 2003;102:1340–46. doi: 10.1182/blood-2003-01-0223. [DOI] [PubMed] [Google Scholar]
  • 50.Rizzo JD, Somerfield MR, Hagerty KL, et al. American Society of Clinical Oncology; American Society of Hematology. Use of epoetin and darbepoetin in patients with cancer: 2007 American Society of Clinical Oncology/American Society of Hematology clinical practice guideline update. J Clin Oncol. 2008;26:132–49. doi: 10.1200/JCO.2007.14.3396. [DOI] [PubMed] [Google Scholar]
  • 51.Buemi M, Cavallaro E, Floccari F, et al. The pleiotropic effects of erythropoietin in the central nervous system. J Neuropathol Exp Neurol. 2003;62:228–36. doi: 10.1093/jnen/62.3.228. [DOI] [PubMed] [Google Scholar]
  • 52.Rasmussen P, Foged EM, Krogh-Madsen R, et al. Effects of erythropoietin administration on cerebral metabolism and exercise capacity in men. J Appl Physiol. 2010;109:476–83. doi: 10.1152/japplphysiol.00234.2010. [DOI] [PubMed] [Google Scholar]
  • 53.Lykissas MG, Korompilias AV, Vekris MD, et al. The role of erythropoietin in central and peripheral nerve injury. Clin Neurol Neurosurg. 2007;109:639–44. doi: 10.1016/j.clineuro.2007.05.013. [DOI] [PubMed] [Google Scholar]
  • 54.Tsai PT, Ohab JJ, Kertesz N, et al. A critical role of erythropoietin receptor in neurogenesis and post-stroke recovery. J Neurosci. 2006;26:1269–74. doi: 10.1523/JNEUROSCI.4480-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Campana WM, Myers RR. Erythropoietin and erythropoietin receptors in the peripheral nervous system: changes after nerve injury. FASEB J. 2001;15:1804–6. doi: 10.1096/fj.00-0857fje. [DOI] [PubMed] [Google Scholar]
  • 56.Lipton SA. Erythropoietin for neurologic protection and diabetic neuropathy. N Engl J Med. 2004;350:2516–17. doi: 10.1056/NEJMcibr041121. [DOI] [PubMed] [Google Scholar]
  • 57.Jerndal M, Forsberg K, Sena ES, et al. A systematic review and meta-analysis of erythropoietin in experimental stroke. J Cereb Blood Flow Metab. 2010;30:961–68. doi: 10.1038/jcbfm.2009.267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Digicaylioglu M. Erythropoietin in stroke: quo vadis. Expert Opin Biol Ther. 2010;10:937–49. doi: 10.1517/14712598.2010.481435. [DOI] [PubMed] [Google Scholar]
  • 59.Westenfelder C. Unexpected renal actions of erythropoietin. Exp Nephrol. 2005;10:294–98. doi: 10.1159/000065304. [DOI] [PubMed] [Google Scholar]
  • 60.Bernhardt WM, Eckardt KU. Physiological basis for the use of erythropoietin in critically ill patients at risk for acute kidney injury. Current Opinion in Critical Care. 2008;14:621–26. doi: 10.1097/MCC.0b013e328317ee82. [DOI] [PubMed] [Google Scholar]
  • 61.Bahlmann FH, Fliser D. Erythropoietin and renoprotection. Current Opinion in Nephrology and Hypertension. 2009;18:15–20. doi: 10.1097/MNH.0b013e32831a9dde. [DOI] [PubMed] [Google Scholar]
  • 62.Gluba A, Banach M, Hannam S, et al. The role of Toll-like receptors in renal diseases. Nat Rev Nephrol. 2010;6:224–35. doi: 10.1038/nrneph.2010.16. [DOI] [PubMed] [Google Scholar]
  • 63.Johnson DW, Pat B, Vesey DA, et al. Delayed administration of darbepoetin or erythropoietin protects against ischemic acute renal injury and failure. Kidney Int. 2006;69:1806–13. doi: 10.1038/sj.ki.5000356. [DOI] [PubMed] [Google Scholar]
  • 64.Endre ZH, Walker RJ, Pickering JW, et al. Early intervention with erythropoietin does not affect the outcome of acute kidney injury (the EARLYARF trial) Kidney Int. 2010;77:1020–30. doi: 10.1038/ki.2010.25. [DOI] [PubMed] [Google Scholar]
  • 65.Małyszko JS, Rams L, Drozdowska-Rams L, Małyszko J. Serum neutrophil gelatinase-associated lipocalin as a marker of kidney function in pregnancy – useful or doubtful? Arch Med Sci. 2010;6:744–47. doi: 10.5114/aoms.2010.17090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gouva C, Nikolopoulos P, Ioannidis JP, Siamopoulos KC. Treating anemia early in renal failure patients slows the decline of renal function: a randomized controlled trial. Kidney Int. 2004;66:753–60. doi: 10.1111/j.1523-1755.2004.00797.x. [DOI] [PubMed] [Google Scholar]
  • 67.Faquin WC, Schneider TJ, Goldberg MA. Effect of inflammatory cytokines on hypoxia-induced erythropoietin production. Blood. 1992;79:1987–94. [PubMed] [Google Scholar]
  • 68.Rysz J, Banach M, Cialkowska-Rysz A, et al. Blood serum levels of IL-2, IL-6, IL-8, TNF-alpha and IL-1beta in patients on maintenance hemodialysis. Cell Mol Immunol. 2006;3:151–54. [PubMed] [Google Scholar]
  • 69.Rysz J, Aronow WS, Stolarek RS, et al. Nephroprotective and clinical potential of statins in dialyzed patients. Expert Opin Ther Targets. 2009;13:541–50. doi: 10.1517/14728220902882130. [DOI] [PubMed] [Google Scholar]
  • 70.El-Ters M, Muthyala U, Philipneri MD, et al. Immune-complex deposits in “pauci-immune” glomerulonephritis: a case report and brief review of recent literature. Arch Med Sci. 2010;6:633–37. doi: 10.5114/aoms.2010.14479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Vannucchi AM, Grossi A, Rafanelli D, et al. Inhibition of erythropoietin production in vitro by human interferon gamma. Br J Haematol. 1994;87:18–23. doi: 10.1111/j.1365-2141.1994.tb04864.x. [DOI] [PubMed] [Google Scholar]
  • 72.Morceau F, Dicato M, Diederich M. Pro-Inflammatory Cytokine-Mediated Anemia: Regarding Molecular Mechanisms of Erythropoiesis. Mediators Inflamm. 2009;2009:405016. doi: 10.1155/2009/405016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.WoŸniak B, WoŸniak A, Drewa G, et al. The effect of treatment on lipid peroxidation in patients with subarachnoid haemorrhage. Arch Med Sci. 2009;5:394–400. [Google Scholar]
  • 74.Barroso O, Mazzoni I, Rabin O. Hormone abuse, in sports: the antidoping perspective. Asian J Androl. 2008;10:391–402. doi: 10.1111/j.1745-7262.2008.00402.x. [DOI] [PubMed] [Google Scholar]
  • 75.Besarab A, Bolton WK, Browne JK, et al. The effects of normal as compared with low hematocrit values in patients with cardiac disease who are receiving hemodialysis and epoetin. N Engl J Med. 1998;339:584–90. doi: 10.1056/NEJM199808273390903. [DOI] [PubMed] [Google Scholar]
  • 76.Singh AK, Szczech L, Tang KL, et al. CHOIR Investigators. Correction of anemia with epoetin alfa in chronic kidney disease. N Engl J Med. 2006;355:2085–98. doi: 10.1056/NEJMoa065485. [DOI] [PubMed] [Google Scholar]
  • 77.Villa G. Erythropoiesis-stimulating agents: from the origins to new challenge. G Ital Nefrol. 2010;27(Suppl 52):S38–46. [PubMed] [Google Scholar]
  • 78.Solomon SD, Uno H, Lewis EF, et al. Erythropoietic response and outcomes in kidney disease and type 2 diabetes. N Engl J Med. 2010;363:1146–55. doi: 10.1056/NEJMoa1005109. [DOI] [PubMed] [Google Scholar]
  • 79.Lubas A, Żelichowski G, Próchnicka A, et al. Renal vascular response to angiotensin II inhibition in intensive antihypertensive treatment of essential hypertension. Arch Med Sci. 2010;6:533–38. doi: 10.5114/aoms.2010.14464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Banach M, Kjeldsen SE, Narkiewicz K. Controversies in hypertension treatment. Curr Vasc Pharmacol. 2010;8:731–32. doi: 10.2174/157016110793563915. [DOI] [PubMed] [Google Scholar]
  • 81.Ortega LM, Contreras G. The clinical impact of the physiological effects of erythropoietin and erythropoietin-stimulating agents on the incidence of malignancy, and hypertension: Beyond anaemia. Nefrologia. 2009;29:288–94. doi: 10.3265/Nefrologia.2009.29.4.5282.en.full. [DOI] [PubMed] [Google Scholar]
  • 82.Heidenreich S, Rahn KH, Zidek W. Direct vasopressor effect of recombinant human erythropoietin on renal resistance vessels. Kidney Int. 1991;39:259–65. doi: 10.1038/ki.1991.31. [DOI] [PubMed] [Google Scholar]
  • 83.Dunn A, Lo V, Donnelly S. The role of the kidney in blood volume regulation: the kidney as a regulator of the hematocrit. Am J Med Sci. 2007;334:65–71. doi: 10.1097/MAJ.0b013e318095a4ae. [DOI] [PubMed] [Google Scholar]
  • 84.Pratt MC, Lewis-Barned NJ, Walker RJ, et al. Effect of angiotensin converting enzyme inhibitors on erythropoietin concentrations in healthy volunteers. Br J Clin Pharmacol. 1992;34:363–65. doi: 10.1111/j.1365-2125.1992.tb05644.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Bartnicki P, Majewska E, Wilk R, et al. Captopril and losartan modify mitogen-induced proliferative response and expression of some differentiation antigents on peripheral blood mononuclear cells in chronic uraemic patients. Arch Med Sci. 2009;5:401–7. [Google Scholar]
  • 86.Du J, Peng T, Scheidegger KJ, Delafontaine P. Angiotensin II activation of insulin-like growth factor 1 receptor transcription is mediated by a tyrosine kinase-dependent redox-sensitive mechanism. Arterioscler Thromb Vasc Biol. 1999;19:2119–26. doi: 10.1161/01.atv.19.9.2119. [DOI] [PubMed] [Google Scholar]
  • 87.Lundby C, Thomsen JJ, Boushel R, et al. Erythropoietin treatment elevates haemoglobin concentration by increasing red cell volume and depressing plasma volume. J Physiol. 2007;578:309–14. doi: 10.1113/jphysiol.2006.122689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Williams B, Edmunds ME, Thompson JP, et al. Does increasing haemoglobin concentration and haematocrit have a pressor effect in dialysis patients? Nephrol Dial Transplant. 1989;4:787–91. [PubMed] [Google Scholar]
  • 89.Krapf R, Hulter HN. Arterial Hypertension Induced by Erythropoietin and Erythropoiesis-Stimulating Agents (ESA) Clin J Am Soc Nephrol. 2009;4:470–80. doi: 10.2215/CJN.05040908. [DOI] [PubMed] [Google Scholar]
  • 90.Kędziora-Kornatowska K, Czuczejko J, Motyl J, et al. Effects of coenzyme Q10 supplementation on activities of selected antioxidative enzymes and lipid peroxidation in hypertensive patients treated with indapamide. A pilot study. Arch Med Sci. 2010;6:513–18. doi: 10.5114/aoms.2010.14461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Casserly LF, Reddy SM, Dember LM. Venous thromboembolism in end-stage renal disease. Am J Kidney Dis. 2000;36:405–11. doi: 10.1053/ajkd.2000.8983. [DOI] [PubMed] [Google Scholar]
  • 92.Jaar B, Denis A, Viron B, et al. Effects of long-term treatment with recombinant human erythropoietin on physiologic inhibitors of coagulation. Am J Nephrol. 1997;17:399–405. doi: 10.1159/000169130. [DOI] [PubMed] [Google Scholar]
  • 93.Krzeslowska J, Rysz J, Cierniewski CS, Luciak M. Expression of fibrinogen receptors and GPIIb molecules on uraemic platelets: effect of recombinant human erythropoietin therapy. Nephrol Dial Transplant. 1995;10:653–56. [PubMed] [Google Scholar]
  • 94.Diskin CJ, Stokes TJ, Thomas SG, et al. An analysis of the effect of routine medications on hemodialysis vascular access survival. Nephron. 1998;78:365–68. doi: 10.1159/000044961. [DOI] [PubMed] [Google Scholar]
  • 95.Keung YK, Owen J. Iron deficiency and thrombosis: literature review. Clin Appl Thromb Hemost. 2004;10:387–91. doi: 10.1177/107602960401000412. [DOI] [PubMed] [Google Scholar]
  • 96.Henry DH, Dahl NV, Auerbach M, et al. Intravenous ferric gluconate significantly improves response to epoetin alfa versus oral iron or no iron in anemic patients with cancer receiving chemotherapy. Oncologist. 2007;12:231–42. doi: 10.1634/theoncologist.12-2-231. [DOI] [PubMed] [Google Scholar]
  • 97.Pirnay F. Doping in sports. Rev Med Liege. 2001;56:265–68. [PubMed] [Google Scholar]
  • 98.Jeong SK, Cho YI, Duey M, Rosenson RS. Cardiovascular risks of anemia correction with erythrocyte stimulating agents: should blood viscosity be monitored for risk assessment? Cardiovasc Drugs Ther. 2010;24:151–60. doi: 10.1007/s10557-010-6239-7. [DOI] [PubMed] [Google Scholar]
  • 99.Yu M, Wen N, Wenzhong Z, et al. Effect of repeated ischaemic preconditioning on TLR4 and proinflammatory cytokines TNF-α and IL-1β in myocardial ischaemia-reperfusion injury in a rat model. Arch Med Sci. 2010;6:843–47. doi: 10.5114/aoms.2010.19289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Okoński P, Banach M, Rysz J, et al. L-arginine improves hemodynamic function and coronary flow in an experimental model of ischemia-reperfusion injury. Ann Transplant. 2006;11:28–34. [PubMed] [Google Scholar]
  • 101.Haleagrahara N, Yee TM, Chakravarthi S, Lee N. Protective effect of N-acetylcysteine on cyclosporine A-induced changes in lipid hydroperoxide levels and renal dysfunction in rats. Arch Med Sci. 2009;5:16–22. [Google Scholar]
  • 102.Covic A, Seica A, Gusbeth-Tatomir P, Goldsmith D. Hemoglobin normalization trials in chronic kidney disease: what should we learn about quality of life as an end point? J Nephrol. 2008;21:478–84. [PubMed] [Google Scholar]
  • 103.Palmer SC, Navaneethan SD, Craig JC, et al. Meta-analysis: erythropoiesis-stimulating agents in patients with chronic kidney disease. Ann Intern Med. 2010;153:23–33. doi: 10.7326/0003-4819-153-1-201007060-00252. [DOI] [PubMed] [Google Scholar]
  • 104.Liao D, Gong P, Li X, et al. Co-culture with Schwann cells is an effective way for adipose-derived stem cells neural transdifferentiation. Arch Med Sci. 2010;6:145–51. doi: 10.5114/aoms.2010.13885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Uyanikgil Y, Balcioglu HA. Neural stem cell therapy in neurological diseases. Arch Med Sci. 2009;5:296–302. [Google Scholar]
  • 106.Souvenir R, Fathali N, Ostrowski RP, et al. Tissue inhibitor of matrix metalloproteinase-1 mediates erythropoietin-induced neuroprotection in hypoxia ischemia. Neurobiol Dis. 2011;44(1):28–37. doi: 10.1016/j.nbd.2011.05.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Cicero AF, Ertek S. Preclinical and clinical evidence of nephro- and cardiovascular protective effects of glycosaminoglycans. Arch Med Sci. 2010;6:469–77. doi: 10.5114/aoms.2010.14456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Durmaz O, Demirkaya M, Sevinir B. Recombinant human erythropoietin β: the effect of weekly dosing on anemia, quality of life, and long-term outcomes in pediatric cancer patients. Pediatr Hematol Oncol. 2011;28:461–68. doi: 10.3109/08880018.2011.570857. [DOI] [PubMed] [Google Scholar]

Articles from Medical Science Monitor : International Medical Journal of Experimental and Clinical Research are provided here courtesy of International Scientific Information, Inc.

RESOURCES