Erythropoietin and hypoxia stimulate erythropoietin receptor and nitric oxide production by endothelial cells

Blood ◽  
2004 ◽  
Vol 104 (7) ◽  
pp. 2073-2080 ◽  
Author(s):  
Bojana B. Beleslin-Cokic ◽  
Vladan P. Cokic ◽  
Xiaobing Yu ◽  
Babette B. Weksler ◽  
Alan N. Schechter ◽  
...  

Abstract Erythropoietin (EPO), a hypoxia-inducible cytokine, is required for survival, proliferation, and differentiation of erythroid progenitor cells. EPO can also stimulate proliferation and angiogenesis of endothelial cells that express EPO receptors (EPORs). In this study we investigated the EPO response of vascular endothelial cells at reduced oxygen tension (5% and 2%), in particular the effect of EPO on nitric oxide (NO) release. Endothelial nitric oxide synthase (eNOS) produces NO, which maintains blood pressure homeostasis and blood flow. We find that EPOR is inducible by EPO in primary human endothelial cells of vein (HUVECs) and artery (HUAECs) and cells from a human bone marrow microvascular endothelial line (TrHBMEC) to a much greater extent at low oxygen tension than in room air. We found a corresponding increase in eNOS expression and NO production in response to EPO during hypoxia. Stimulation of NO production was dose dependent on EPO concentration and was maximal at 5 U/mL. NO activates soluble guanosine cyclase to produce cyclic guanosine monophosphate (cGMP), and we observed that EPO induced cGMP activity. These results suggest that low oxygen tension increases endothelial cell capacity to produce NO in response to EPO by induction of both EPOR and eNOS. This effect of EPO on eNOS may be a physiologically relevant mechanism to counterbalance the hypertensive effects of increased hemoglobin-related NO destruction resulting from hypoxia-induced increased red cell mass. (Blood. 2004;104:2073-2080)

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3724-3724
Author(s):  
Bojana B. Beleslin-Cokic ◽  
Vladan P. Cokic ◽  
Ljiljana Gojkovic-Bukarica ◽  
Constance Tom Noguchi

Abstract Erythopoietin (EPO) and erythropoietin receptor (EPOR) regulate survival, proliferation, differentiation and viability of erythroid progenitor cells. Beyond erythropoiesis, we have observed that human vascular endothelial cells respond to EPO stimulation by inducing EPOR and endothelial nitric oxide synthase (eNOS) expression, increasing NO production and cGMP, particularly under low oxygen tension. In this study, we investigate the response of vascular smooth muscle (VSM) to EPO stimulation and the contribution of blood vessel to relaxation/contraction. We found that VSM cells express EPOR and that treatment with EPO (5 U/ml) at reduced oxygen tension increased EPOR mRNA by 2 fold. This increased EPO responsiveness at low oxygen tension is accompanied by increased cell proliferation at 2% O2 more than 2 fold. Unlike endothelial cells, EPO did not induce eNOS or NO production in VSM cells. PI-3 kinase was involved in EPO stimulation with no change in MAP kinase. In an isolated blood vessel model system, we checked EPO responsiveness. EPO produced contractions (0.32 ± 0.3 g) of rat renal artery, and pretreatment with LY294002 (10 mM), an inhibitor of PI-3 kinase, statistically significantly inhibited this contraction (58.7 ± 7%). This response was also observed with the endothelium layer removed. In preparations of human internal mammary artery (HIMA) and human saphenous vein (HSV) from patients undergoing coronary artery bypass, EPO did not affect basal vascular tone of HIMA and HSV with the endothelium layer removed, but EPO (5 U/ml) potentated noradrenalin-induced contraction by up to 2 fold in HSV and HIMA. Also, pretreatment with EPO significantly increase angiotensin-evoked contractions of these blood vessels (50 ± 8%, 113 ± 17%, respectively P < 0.01), suggesting that EPO has synergistic effects on angiotensin or noradrenalin-induced [Ca2+] mobilization, particularly on intracellular Ca2+ release. These data suggest that EPO stimulation of vascular smooth muscle may act to modulate or balance the vasodilatory effects of increased NO production by EPO stimulated endothelium. Under oxygen stress vascular smooth muscle can respond to EPO by proliferation and PI-3 kinase activation as a protective effect and in conjunction with Ca2+ release likely contributes to the overall vascular response.


2006 ◽  
Vol 290 (6) ◽  
pp. L1111-L1116 ◽  
Author(s):  
Vivek Balasubramaniam ◽  
Anne M. Maxey ◽  
Brian W. Fouty ◽  
Steven H. Abman

Growth and development of the lung normally occur in the low oxygen environment of the fetus. The role of this low oxygen environment on fetal lung endothelial cell growth and function is unknown. We hypothesized that low oxygen tension during fetal life enhances pulmonary artery endothelial cell (PAEC) growth and function and that nitric oxide (NO) production modulates fetal PAEC responses to low oxygen tension. To test this hypothesis, we compared the effects of fetal (3%) and room air (RA) oxygen tension on fetal PAEC growth, proliferation, tube formation, and migration in the presence and absence of the NO synthase (NOS) inhibitor Nω-nitro-l-arginine (LNA), and an NO donor, S-nitroso- N-acetylpenicillamine (SNAP). Compared with fetal PAEC grown in RA, 3% O2 increased tube formation by over twofold ( P < 0.01). LNA treatment reduced tube formation in 3% O2 but had no affect on tube formation in RA. Treatment with SNAP increased tube formation during RA exposure to levels observed in 3% O2. Exposure to 3% O2 for 48 h attenuated cell number (by 56%), and treatment with LNA reduced PAEC growth by 44% in both RA and 3% O2. We conclude that low oxygen tension enhances fetal PAEC tube formation and that NO is essential for normal PAEC growth, migration, and tube formation. Furthermore, we conclude that in fetal cells exposed to the relative hyperoxia of RA, 21% O2, NO overcomes the inhibitory effects of the increased oxygen, allowing normal PAEC angiogenesis and branching. We speculate that NO production maintains intrauterine lung vascular growth and development during exposure to low O2 in the normal fetus. We further speculate that NO is essential for pulmonary angiogenesis in fetal animal exposed to increased oxygen tension of RA and that impaired endothelial NO production may contribute to the abnormalities of angiogenesis see in infants with bronchopulmonary dysplasia.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2125-2125 ◽  
Author(s):  
Marcel H.A.M. Fens ◽  
Sandra K Larkin ◽  
Claudia R. Morris ◽  
Bill Fitch ◽  
Jan Scicinski ◽  
...  

Abstract Abstract 2125 Introduction: Nitric oxide (NO) metabolism is dysregulated in sickle cell disease (SCD) and Thalassemia. Both higher consumption of arginine and arginase-released by hemolysis are implicated in decreased production of NO, negatively affecting vaso-dilation and resolve of vaso-occlusive crises (VOC). The Red Blood Cell (RBC) plays a role in NO transport, as well as in nitrite (NO2−) reduction to NO. We studied the relation between oxygen affinity of RBCs and their capacity to reduce NO2− to NO under different partial oxygen pressures. Methods: Blood samples were collected with IRB approval from normal controls, b-thalassamia (bThal) patients, and SCD patients including those who were treated with hydroxyurea (HU). RBC hemoglobin (Hb) was modified with compounds that increased the oxygen affinity (lowering P50) including anti-sickling agents such as 5-hydroxymethyl-2-furfural (5HMF). Samples at different hematocrits, with different fractions of modified RBCs or free Hb, were incubated in a Tonometer (Instrumentation Laboratory) at 37°C under a constant flow of an Air/Nitrogen gas mixture. Varying amounts of NO2− were added to the cells, and NO2− and NO3− were measured in the extracellular medium. The gas outflow from the Tonometer was collected in mylar balloons, and NO production was measured using a Nitric Oxide Analyzer (GE Analytical Instruments). Before and after incubation the samples were analyzed by spectroscopy, and the oxygen affinity was measured with the Hemox Analyzer (TCS Medical Products). Results: The concentration of NO2− decreased exponentially in the extracellular fluid with a rate that increased with hematocrit, and oxygen tension to reach an apparent equilibrium after 30–45 minutes. NO3−, but little NO, from any of the RBC mixtures was released under room air. NO was generated under nitrogen from all RBC mixtures, and the rate and amounts released in 30 minutes were directly related to NO2− concentration, hematocrit and fraction of free Hb present. High levels of NO2− resulted in irreversible modification of RBCs and a loss of oxygen carrying capacity as measured by spectroscopy and oxygen affinity analysis. When these cells were mixed with unmodified RBCs, NO production was related to the fraction of RBCs with an altered P50. Hb modification in normal and bThal cells increased NO production with decreasing P50. SCD-RBCs showed a higher NO generation as compared to normal RBCs, which increased with the presence of HbF. Modification of SCD-RBCs further increased NO production with a decreased P50. Together, any decrease (left-shift) in P50 resulted in an increase in NO production under low oxygen conditions. NO formation was dependent on the subpopulation of modified cells in the entire population. Conclusions: Any condition that decreased the P50, resulted in an increased reduction of NO2− to NO under low oxygen tension. These modifications include potential, and reported anti-sickling agents such as HU and 5HMF. In addition, mixing extensively modified RBCs or free Hb with untreated RBCs increased the capacity of the entire population to reduce NO2−. Interestingly, SCD-RBCs with low levels of HbF, which showed a slightly increased P50 as compared to normal RBCs, showed an increased ability to reduce NO2−. We hypothesize that the local presence of modified RBCs with an increased oxygen affinity or the presence of free Hb will increase the local formation of NO from NO2−. As such, anti-sickling agents may be beneficial to lower the incidence of VOC, not only by reducing the rate of Hb polymerization, but also by the increased capacity of the RBCs to generate NO for vaso-dilation. Disclosures: Fitch: RadioRx, Inc: Research Funding. Scicinski:RadioRx, Inc: Research Funding. Oronsky:RadioRx, Inc: Research Funding.


Molecules ◽  
2021 ◽  
Vol 26 (24) ◽  
pp. 7480
Author(s):  
Gabriele Serreli ◽  
Melanie Le Sayec ◽  
Camilla Diotallevi ◽  
Alice Teissier ◽  
Monica Deiana ◽  
...  

Nitric oxide (NO) is an important signaling molecule involved in many pathophysiological processes. NO mediates vasodilation and blood flow in the arteries, and its action contributes to maintaining vascular homeostasis by inhibiting vascular smooth muscle contraction and growth, platelet aggregation, and leukocyte adhesion to the endothelium. Dietary antioxidants and their metabolites have been found to be directly and/or indirectly involved in the modulation of the intracellular signals that lead to the production of NO. The purpose of this study was to investigate the contribution of conjugated metabolites of hydroxytyrosol (HT) and tyrosol (TYR) to the release of NO at the vascular level, and the related mechanism of action, in comparison to their parental forms. Experiments were performed in human aortic endothelial cells (HAEC) to evaluate the superoxide production, the release of NO and production of cyclic guanosine monophosphate (cGMP), the activation of serine/threonine-protein kinase 1 (Akt1), and the activation state of endothelial nitric oxide synthase (eNOS). It was observed that the tested phenolic compounds enhanced NO and cGMP concentration, inhibiting its depletion caused by superoxide overproduction. Moreover, some of them enhanced the activation of Akt (TYR, HT metabolites) and eNOS (HT, HVA, TYR-S, HT-3S). Overall, the obtained data showed that these compounds promote NO production and availability, suggesting that HT and TYR conjugated metabolites may contribute to the effects of parental extra virgin olive oil (EVOO) phenolics in the prevention of cardiovascular diseases.


Sign in / Sign up

Export Citation Format

Share Document