RBC-NOS-dependent S-nitrosylation of cytoskeletal proteins improves RBC deformability

Marijke Grau, Sebastian Pauly, Jamal Ali, Katja Walpurgis, Mario Thevis, Wilhelm Bloch, Frank Suhr, Marijke Grau, Sebastian Pauly, Jamal Ali, Katja Walpurgis, Mario Thevis, Wilhelm Bloch, Frank Suhr

Abstract

Background: Red blood cells (RBC) possess a nitric oxide synthase (RBC-NOS) whose activation depends on the PI3-kinase/Akt kinase pathway. RBC-NOS-produced NO exhibits important biological functions like maintaining RBC deformability. Until now, the cellular target structure for NO, to exert its influence on RBC deformability, remains unknown. In the present study we analyzed the modification of RBC-NOS activity by pharmacological treatments, the resulting influence on RBC deformability and provide first evidence for possible target proteins of RBC-NOS-produced NO in the RBC cytoskeletal scaffold.

Methods/findings: Blood from fifteen male subjects was incubated with the NOS substrate L-arginine to directly stimulate enzyme activity. Direct inhibition of enzyme activity was induced by L-N5-(1-Iminoethyl)-ornithin (L-NIO). Indirect stimulation and inhibition of RBC-NOS were achieved by applying insulin and wortmannin, respectively, substances known to affect PI3-kinase/Akt kinase pathway. The NO donor sodium nitroprusside (SNP) and the NO scavenger 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (cPTIO) were additionally applied as NO positive and negative controls, respectively. Immunohistochemical staining was used to determine phosphorylation and thus activation of RBC-NOS. As a marker for NO synthesis nitrite was measured in plasma and RBCs using chemiluminescence detection. S-nitrosylation of erythrocyte proteins was determined by biotin switch assay and modified proteins were identified using LC-MS. RBC deformability was determined by ektacytometry. The data reveal that activated RBC-NOS leads to increased NO production, S-nitrosylation of RBC proteins and RBC deformability, whereas RBC-NOS inhibition resulted in contrary effects.

Conclusion/significance: This study first-time provides strong evidence that RBC-NOS-produced NO modifies RBC deformability through direct S-nitrosylation of cytoskeleton proteins, most likely α- and β-spectrins. Our data, therefore, gain novel insights into biological functions of RBC-NOS by connecting impaired RBC deformability abilities to specific posttranslational modifications of RBC proteins. By identifying likely NO-target proteins in RBC, our results will stimulate new therapeutic approaches for patients with microvascular disorders.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Changes of RBC-NOSSer 1177 levels…
Figure 1. Changes of RBC-NOSSer1177 levels after RBC incubation with RBC-NOS stimulants, inhibitors and NO controls.
(A) Bars show statistical analysis of gray values [au] after a 60 min incubation of RBCs with insulin and wortmannin, respectively. Application of RBC-NOS stimulant insulin led to a significant increase in enzyme phosphorylation compared to control (P<0.01). Wortmannin (P<0.001) application decreased phosphorylation, respectively. (B) Pictures show representative RBC-NOSSer1177 staining after 60 min incubation, respectively. Magnification for all images was 400-fold. Data in (A) are presented as mean±S.E.M., n = 6.
Figure 2. Changes in nitrite concentration after…
Figure 2. Changes in nitrite concentration after modification of RBC-NOS activity, and plasma ADMA content after L-arginine application.
Nitrite was measured as representative of NO synthesis after 60 min incubation of RBCs with RBC-NOS stimulants, inhibitors and NO controls. (A) RBC nitrite level significantly increased after L-arginine (P<0.01), insulin (P<0.01) and SNP (P<0.01) application, while L-NIO (P<0.001), wortmannin (P<0.01) and cPTIO (P<0.05) significantly decreased RBC nitrite content compared to control samples. (B) Plasma ADMA concentration was additionally measured after L-arginine incubation to present an explanation for the L-arginine paradox. Bars show that ADMA concentration in the plasma fraction significantly increased upon substrate addition (P<0.001). (C) Plasma nitrite concentration was not altered by the applied substances, except SNP. Data in (A–C) are presented as mean±S.E.M., n = 15.
Figure 3. S-nitrosylation of RBC proteins after…
Figure 3. S-nitrosylation of RBC proteins after modification of RBC-NOS-dependent NO production.
(A) Representative western blot bands after 60 min incubation of RBCs with RBC-NOS stimulants, inhibitors and NO controls are demonstrated. The image shows two distinctive bands with varying intensity depending on RBC-NOS modulation. LC-MS/MS analysis identified 240 kDa band as possible α- spectrin and 220 kDa band as possible β-spectrin. (B) Calculation of relative intensity in relation to the control sample revealed that S-nitrosylation of both, α- and β-spectrin, significantly increased upon L-arginine, insulin and SNP incubation and significantly decreased upon L-NIO, wortmannin and cPTIO incubation, respectively. Data in (B) are presented as mean±S.E.M., n = 6.
Figure 4. Maximum deformability (EI max) after…
Figure 4. Maximum deformability (EI max) after RBC incubation with RBC-NOS stimulants, inhibitors and NO controls and correlation of EI max with S-nitrosylation of α- and β-spectrin.
(A) Bars show that EI max significantly increased after 60 min incubation with L-arginine (P

References

    1. Ignarro LJ (1989) Heme-dependent activation of soluble guanylate cyclase by nitric oxide: regulation of enzyme activity by porphyrins and metalloporphyrins. Semin Hematol 26: 63–76.
    1. Radomski MW, Palmer RM, Moncada S (1990) Characterization of the L-arginine:nitric oxide pathway in human platelets. Br J Pharmacol 101: 325–328.
    1. Bryan NS, Fernandez BO, Bauer SM, Garcia-Saura MF, Milsom AB, et al. (2005) Nitrite is a signaling molecule and regulator of gene expression in mammalian tissues. Nat Chem Biol 1: 290–297.
    1. Gladwin MT (2005) Nitrite as an intrinsic signaling molecule. Nat Chem Biol 1: 245–246.
    1. Cosby K, Partovi KS, Crawford JH, Patel RP, Reiter CD, et al. (2003) Nitrite reduction to nitric oxide by deoxyhemoglobin vasodilates the human circulation. Nat Med 9: 1498–1505.
    1. Crawford JH, Isbell TS, Huang Z, Shiva S, Chacko BK, et al. (2006) Hypoxia, red blood cells, and nitrite regulate NO-dependent hypoxic vasodilation. Blood 107: 566–574.
    1. Lundberg JO, Weitzberg E (2005) NO generation from nitrite and its role in vascular control. Arterioscler Thromb Vasc Biol 25: 915–922.
    1. Nagababu E, Ramasamy S, Abernethy DR, Rifkind JM (2003) Active nitric oxide produced in the red cell under hypoxic conditions by deoxyhemoglobin-mediated nitrite reduction. J Biol Chem 278: 46349–46356.
    1. Totzeck M, Hendgen-Cotta UB, Luedike P, Berenbrink M, Klare JP, et al. (2012) Nitrite regulates hypoxic vasodilation via myoglobin-dependent nitric oxide generation. Circulation 126: 325–334.
    1. Ulker P, Gunduz F, Meiselman HJ, Baskurt OK (2012) Nitric oxide generated by red blood cells following exposure to shear stress dilates isolated small mesenteric arteries under hypoxic conditions. Clin Hemorheol Microcirc
    1. Foster MW, McMahon TJ, Stamler JS (2003) S-nitrosylation in health and disease. Trends Mol Med 9: 160–168.
    1. Bruckdorfer R (2005) The basics about nitric oxide. Mol Aspects Med 26: 3–31.
    1. Martinez-Ruiz A, Lamas S (2004) S-nitrosylation: a potential new paradigm in signal transduction. Cardiovasc Res 62: 43–52.
    1. Lane P, Hao G, Gross SS (2001) S-nitrosylation is emerging as a specific and fundamental posttranslational protein modification: head-to-head comparison with O-phosphorylation. Sci STKE 2001: re1.
    1. Broillet MC (1999) S-nitrosylation of proteins. Cell Mol Life Sci 55: 1036–1042.
    1. Hess DT, Matsumoto A, Kim SO, Marshall HE, Stamler JS (2005) Protein S-nitrosylation: purview and parameters. Nat Rev Mol Cell Biol 6: 150–166.
    1. Chung KK, Thomas B, Li X, Pletnikova O, Troncoso JC, et al. (2004) S-nitrosylation of parkin regulates ubiquitination and compromises parkin's protective function. Science 304: 1328–1331.
    1. Heitzer T, Schlinzig T, Krohn K, Meinertz T, Munzel T (2001) Endothelial dysfunction, oxidative stress, and risk of cardiovascular events in patients with coronary artery disease. Circulation 104: 2673–2678.
    1. Kleinbongard P, Dejam A, Lauer T, Jax T, Kerber S, et al. (2006) Plasma nitrite concentrations reflect the degree of endothelial dysfunction in humans. Free Radic Biol Med 40: 295–302.
    1. Agarwal J, Gupta SC, Singh PA, Bisht D, Keswani NK (1992) A study of humoral factors in carcinoma cervix. Indian J Pathol Microbiol 35: 5–10.
    1. Jubelin BC, Gierman JL (1996) Erythrocytes may synthesize their own nitric oxide. Am J Hypertens 9: 1214–1219.
    1. Kleinbongard P, Schulz R, Rassaf T, Lauer T, Dejam A, et al. (2006) Red blood cells express a functional endothelial nitric oxide synthase. Blood 107: 2943–2951.
    1. Cortese-Krott MM, Rodriguez-Mateos A, Sansone R, Kuhnle GG, Thasian-Sivarajah S, et al... (2012) Human red blood cells at work: identification and visualization of erythrocytic eNOS activity in health and disease. Blood
    1. Suhr F, Brenig J, Muller R, Behrens H, Bloch W, et al. (2012) Moderate Exercise Promotes Human RBC-NOS Activity, NO Production and Deformability through Akt Kinase Pathway. PLoS One 7: e45982.
    1. Mihov D, Vogel J, Gassmann M, Bogdanova A (2009) Erythropoietin activates nitric oxide synthase in murine erythrocytes. Am J Physiol Cell Physiol 297: C378–C388.
    1. Suhr F, Porten S, Hertrich T, Brixius K, Schmidt A, et al. (2009) Intensive exercise induces changes of endothelial nitric oxide synthase pattern in human erythrocytes. Nitric Oxide 20: 95–103.
    1. Bor-Kucukatay M, Wenby RB, Meiselman HJ, Baskurt OK (2003) Effects of nitric oxide on red blood cell deformability. Am J Physiol Heart Circ Physiol 284: H1577–H1584.
    1. Ulker P, Yaras N, Yalcin O, Celik-Ozenci C, Johnson PC, et al. (2011) Shear stress activation of nitric oxide synthase and increased nitric oxide levels in human red blood cells. Nitric Oxide 24: 184–191.
    1. Rees DD, Palmer RM, Schulz R, Hodson HF, Moncada S (1990) Characterization of three inhibitors of endothelial nitric oxide synthase in vitro and in vivo. Br J Pharmacol 101: 746–752.
    1. McCall TB, Feelisch M, Palmer RM, Moncada S (1991) Identification of N-iminoethyl-L-ornithine as an irreversible inhibitor of nitric oxide synthase in phagocytic cells. Br J Pharmacol 102: 234–238.
    1. Imbrogno S, De IL, Mazza R, Tota B (2001) Nitric oxide modulates cardiac performance in the heart of Anguilla anguilla. J Exp Biol 204: 1719–1727.
    1. Michell BJ, Griffiths JE, Mitchelhill KI, Rodriguez-Crespo I, Tiganis T, et al. (1999) The Akt kinase signals directly to endothelial nitric oxide synthase. Curr Biol 9: 845–848.
    1. Ziche M, Morbidelli L, Masini E, Amerini S, Granger HJ, et al. (1994) Nitric oxide mediates angiogenesis in vivo and endothelial cell growth and migration in vitro promoted by substance P. J Clin Invest. 94: 2036–2044.
    1. Ziche M, Morbidelli L, Masini E, Granger H, Geppetti P, et al. (1993) Nitric oxide promotes DNA synthesis and cyclic GMP formation in endothelial cells from postcapillary venules. Biochem Biophys Res Commun 192: 1198–1203.
    1. Vallette G, Tenaud I, Branka JE, Jarry A, Sainte-Marie I, et al. (1998) Control of growth and differentiation of normal human epithelial cells through the manipulation of reactive nitrogen species. Biochem J 331 (Pt 3): 713–717.
    1. Lakshmi VM, Zenser TV (2007) 2-(4-Carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide potentiates nitrosation of a heterocyclic amine carcinogen by nitric oxide. Life Sci 80: 644–649.
    1. Forstermann U, Closs EI, Pollock JS, Nakane M, Schwarz P, et al. (1994) Nitric oxide synthase isozymes. Characterization, purification, molecular cloning, and functions. Hypertension 23: 1121–1131.
    1. Bode-Boger SM, Boger RH, Galland A, Tsikas D, Frolich JC (1998) L-arginine-induced vasodilation in healthy humans: pharmacokinetic-pharmacodynamic relationship. Br J Clin Pharmacol 46: 489–497.
    1. Hishikawa K, Nakaki T, Suzuki H, Kato R, Saruta T (1993) Role of L-arginine-nitric oxide pathway in hypertension. J Hypertens 11: 639–645.
    1. Vallance P, Leone A, Calver A, Collier J, Moncada S (1992) Accumulation of an endogenous inhibitor of nitric oxide synthesis in chronic renal failure. Lancet 339: 572–575.
    1. Boger RH, Bode-Boger SM, Szuba A, Tsao PS, Chan JR, et al. (1998) Asymmetric dimethylarginine (ADMA): a novel risk factor for endothelial dysfunction: its role in hypercholesterolemia. Circulation 98: 1842–1847.
    1. Ludolph B, Bloch W, Kelm M, Schulz R, Kleinbongard P (2007) Short-term effect of the HMG-CoA reductase inhibitor rosuvastatin on erythrocyte nitric oxide synthase activity. Vasc Health Risk Manag 3: 1069–1073.
    1. Kleinbongard P, Dejam A, Lauer T, Rassaf T, Schindler A, et al. (2003) Plasma nitrite reflects constitutive nitric oxide synthase activity in mammals. Free Radic Biol Med 35: 790–796.
    1. Lauer T, Preik M, Rassaf T, Strauer BE, Deussen A, et al. (2001) Plasma nitrite rather than nitrate reflects regional endothelial nitric oxide synthase activity but lacks intrinsic vasodilator action. Proc Natl Acad Sci U S A 98: 12814–12819.
    1. Kelm M, Preik-Steinhoff H, Preik M, Strauer BE (1999) Serum nitrite sensitively reflects endothelial NO formation in human forearm vasculature: evidence for biochemical assessment of the endothelial L-arginine-NO pathway. Cardiovasc Res 41: 765–772.
    1. Pelletier MM, Kleinbongard P, Ringwood L, Hito R, Hunter CJ, et al. (2006) The measurement of blood and plasma nitrite by chemiluminescence: pitfalls and solutions. Free Radic Biol Med 41: 541–548.
    1. Hendgen-Cotta U, Grau M, Rassaf T, Gharini P, Kelm M, et al. (2008) Reductive gas-phase chemiluminescence and flow injection analysis for measurement of the nitric oxide pool in biological matrices. Methods Enzymol 441: 295–315.
    1. Grau M, Hendgen-Cotta UB, Brouzos P, Drexhage C, Rassaf T, et al. (2007) Recent methodological advances in the analysis of nitrite in the human circulation: nitrite as a biochemical parameter of the L-arginine/NO pathway. J Chromatogr B Analyt Technol Biomed Life Sci 851: 106–123.
    1. Stamler JS, Lamas S, Fang FC (2001) Nitrosylation. the prototypic redox-based signaling mechanism. Cell 106: 675–683.
    1. Jaffrey SR, Erdjument-Bromage H, Ferris CD, Tempst P, Snyder SH (2001) Protein S-nitrosylation: a physiological signal for neuronal nitric oxide. Nat Cell Biol 3: 193–197.
    1. Walpurgis K, Kohler M, Thomas A, Wenzel F, Geyer H, et al. (2012) Storage-induced changes of the cytosolic red blood cell proteome analyzed by 2D DIGE and high-resolution/high-accuracy MS. Proteomics 12: 3263–3272.
    1. Hardeman MR, Dobbe JG, Ince C (2001) The Laser-assisted Optical Rotational Cell Analyzer (LORCA) as red blood cell aggregometer. Clin Hemorheol Microcirc 25: 1–11.
    1. Baskurt OK, Boynard M, Cokelet GC, Connes P, Cooke BM, et al. (2009) New guidelines for hemorheological laboratory techniques. Clin Hemorheol Microcirc 42: 75–97.
    1. Tsikas D, Boger RH, Sandmann J, Bode-Boger SM, Frolich JC (2000) Endogenous nitric oxide synthase inhibitors are responsible for the L-arginine paradox. FEBS Lett 478: 1–3.
    1. Foster MW, Hess DT, Stamler JS (2009) Protein S-nitrosylation in health and disease: a current perspective. Trends Mol Med 15: 391–404.
    1. Bateman RM, Sharpe MD, Ellis CG (2003) Bench-to-bedside review: microvascular dysfunction in sepsis--hemodynamics, oxygen transport, and nitric oxide. Crit Care 7: 359–373.
    1. Pries AR, Secomb TW (2003) Rheology of the microcirculation. Clin Hemorheol Microcirc 29: 143–148.
    1. Sessa WC (2004) eNOS at a glance. J Cell Sci 117: 2427–2429.
    1. Kang ES, Ford K, Grokulsky G, Wang YB, Chiang TM, et al. (2000) Normal circulating adult human red blood cells contain inactive NOS proteins. J Lab Clin Med 135: 444–451.
    1. Bohmer A, Beckmann B, Sandmann J, Tsikas D (2012) Doubts concerning functional endothelial nitric oxide synthase in human erythrocytes. Blood 119: 1322–1323.
    1. Chakrabarti A, Kelkar DA, Chattopadhyay A (2006) Spectrin organization and dynamics: new insights. Biosci Rep 26: 369–386.
    1. Forsyth AM, Braunmuller S, Wan J, Franke T, Stone HA (2012) The effects of membrane cholesterol and simvastatin on red blood cell deformability and ATP release. Microvasc Res 83: 347–351.
    1. Franco T, Low PS (2010) Erythrocyte adducin: a structural regulator of the red blood cell membrane. Transfus Clin Biol 17: 87–94.
    1. Banerjee R, Nageshwari K, Puniyani RR (1998) The diagnostic relevance of red cell rigidity. Clin Hemorheol Microcirc 19: 21–24.
    1. Tziakas DN, Kaski JC, Chalikias GK, Romero C, Fredericks S, et al. (2007) Total cholesterol content of erythrocyte membranes is increased in patients with acute coronary syndrome: a new marker of clinical instability? J Am Coll Cardiol 49: 2081–2089.
    1. Gladwin MT, Crawford JH, Patel RP (2004) The biochemistry of nitric oxide, nitrite, and hemoglobin: role in blood flow regulation. Free Radic Biol Med 36: 707–717.

Source: PubMed

3
Abonner