Comprehensive Physiology Wiley Online Library

Nitric Oxide Transport in Blood: A Third Gas in the Respiratory Cycle

Full Article on Wiley Online Library



Abstract

The trapping, processing, and delivery of nitric oxide (NO) bioactivity by red blood cells (RBCs) have emerged as a conserved mechanism through which regional blood flow is linked to biochemical cues of perfusion sufficiency. We present here an expanded paradigm for the human respiratory cycle based on the coordinated transport of three gases: NO, O2, and CO2. By linking O2 and NO flux, RBCs couple vessel caliber (and thus blood flow) to O2 availability in the lung and to O2 need in the periphery. The elements required for regulated O2‐based signal transduction via controlled NO processing within RBCs are presented herein, including S‐nitrosothiol (SNO) synthesis by hemoglobin and O2‐regulated delivery of NO bioactivity (capture, activation, and delivery of NO groups at sites remote from NO synthesis by NO synthase). The role of NO transport in the respiratory cycle at molecular, microcirculatory, and system levels is reviewed. We elucidate the mechanism through which regulated NO transport in blood supports O2 homeostasis, not only through adaptive regulation of regional systemic blood flow but also by optimizing ventilation‐perfusion matching in the lung. Furthermore, we discuss the role of NO transport in the central control of breathing and in baroreceptor control of blood pressure, which subserve O2 supply to tissue. Additionally, malfunctions of this transport and signaling system that are implicated in a wide array of human pathophysiologies are described. Understanding the (dys)function of NO processing in blood is a prerequisite for the development of novel therapies that target the vasoactive capacities of RBCs. © 2011 American Physiological Society. Compr Physiol 1:541‐568, 2011.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1.

Local vascular reflexes support maintenance of O2 delivery to tissue in the setting of progressive hypoxia. In a classic paper 244, Guyton demonstrated regional autoregulation of systemic blood flow in normal dogs (following spinal anesthesia) by observing variation in blood flow during constant pressure blood perfusion of the femoral artery, while reducing the hemoglobin oxygen saturation (Hb So2%) from 100% to 0% in the perfusing blood. (A) Stepwise reduction in Hb So2% caused a progressive increase in blood flow through the leg. (B) These data demonstrate that autoregulation of blood flow occurs at a local level and this regulation serves to improve oxygen supply when blood oxygen content falls. In addition, effects on blood flow were replicated by injecting partially deoxygenated versus oxygenated red blood cells into the artery, demonstrating that effects could be elicited during arteriovenous transit (<1 s). Reproduced with permission from 244.

Figure 2. Figure 2.

Red blood cells (RBCs) transduce regional O2 gradients in tissue to control nitric oxide (NO) bioactivity in plasma by trapping or delivering NO groups as a function of hemoglobin (Hb) O2 saturation. (A) In this fashion, circulating NO groups are processed by Hb into the highly vasoactive (thiol‐based) NO congener, S‐nitrosothiol (SNO). By exporting SNOs as a function of Hb deoxygenation, RBCs precisely dispense vasodilator bioactivity in direct proportion to regional blood flow lack. (B) O2 delivery homeostasis requires biochemical coupling of vessel tone to environmental cues that matches perfusion sufficiency to metabolic demand. Because oxy‐ and deoxy‐Hb process NO differently (see text), allosteric transitions in Hb conformation afford context‐responsive (O2‐coupled) control of NO bioavailability, thereby linking the sensor and effector arms of this system. Specifically, Hb conformation governs the equilibria among deoxy‐HbFeNO (A; NO sink), SNO‐oxy‐Hb (B; NO store), and acceptor thiols including the membrane protein SNO‐AE‐1 (C; bioactive NO source). Direct SNO export from RBCs or S‐transnitrosylation from RBCs to plasma thiols (D) or to endothelial cells directly (not shown) yields vasoactive SNOs, which influence resistance vessel caliber and close this signaling loop. Thus, RBCs either trap (A) or export (D) NO groups to optimize blood flow. (C) NO processing in RBCs (A and B) couples vessel tone to tissue pO2; this system subserves hypoxic vasodilation in the arterial periphery and thereby calibrates blood flow to regional tissue hypoxia. See Figures 10 and 11 and related discussion for more details.

Figure 3. Figure 3.

Hemoglobin oxygen saturation (Hb So2%) exerts coordinated governance of red blood cell (RBC) S‐nitrosothiol (SNO) content, RBC vasoactivity, and human peripheral blood flow. (A) Human blood gas measurements of SNO and O2 64. RBCs with (black) or without (red) added extracellular glutathione (GSH) were deoxygenated under inert gas. The natural logarithm of SNO content in RBCs (SNO RBC) is linearly related to Hb So2. GSH accelerated SNO RBC decay, consistent with O2‐linked export of NO groups to extraerythrocytic thiols. (B) RBCs induce graded relaxation of systemic arteries (aortic ring bioassay) that is inversely related to Hb So2 across the physiological range, recapitulating hypoxic vasodilation (HVD) 196. Hb So2 spanned from red (oxy, >90% Hb So2) to blue (deoxy, <40% Hb So2). (C) Leg vascular conductance increases as blood O2 content falls (HVD) 99. Hb So2 and thus arterial blood O2 content were manipulated by CO exposure ± varying Fio2 (fraction of inspired O2; hypoxia versus hyperoxia). Neither vascular conductance nor blood flow correlated with blood pO2 per se. Note the similarity between observations in panels A, B, and C and Guyton's original observations 244 (Fig. 1). Panel (C) Reproduced with permission from 99.

Figure 4. Figure 4.

Hypoxic vasodilation (1% O2) by native (untreated) human red blood cells (RBCs) of mouse aortic segments requires neither eNOS nor intact endothelium 62. (A) Hypoxiacoupled vasodilation by RBCs is preserved despite endothelial denudation, inhibition of NOS activity with L‐NAME, or eNOS deletion (eNOS‐/‐). RBC‐induced vasorelaxation was greatly attenuated by the guanylate cyclase inhibitor ODQ. (n = 9 to 34; * P < 0.05). (B, C) Representative tracings from the vascular ring preparation show that removal of endothelium eliminates acetylcholine (ACh)‐induced vasorelaxation but has no effect on RBC‐induced vasorelaxation [N.B. vessel rings are precontracted with phenylephrine (PE)].

Figure 5. Figure 5.

Hypoxic vasodilation (HVD) by native human red blood cells (RBCs) of rabbit aortic segments is S‐nitrosothiol (SNO) based 62. (A) RBC‐induced vasorelaxation was potentiated by cysteine and by N‐acetylcysteine but was unaffected by nitrite (NO2) or the prostaglandin synthesis inhibitors indomethacin and aspirin (all agents added 2 min before RBCs) (n = 5 to 11; * P < 0.05). (BD), Representative tracings illustrate the augmentation of relaxation by cysteine and the attenuation of relaxation by S‐nitroso‐hemoglobin depletion (∼80% depletion). (E) Addition of NO2 to the bath (even at supraphysiologic concentrations of 1 μM) before (NO2 → RBC), simultaneously with the addition of RBCs (RBC + NO2), or near the termination of RBC‐induced vasorelaxation (RBC → NO2), had no discernible effect on the magnitude or duration of RBC‐induced HVD.

Figure 6. Figure 6.

Conformation‐specific binding between hemoglobin (Hb) and the red blood cell (RBC) membrane protein AE‐1 affords O2 responsive control of NO trapping 111. (A) Under oxygenated conditions (normoxic RBC, at left), the RBC membrane constitutes a significant barrier to NO entry via tight association between the submembrane cytoskeleton and the cytoplasmic domain of the band 3 (AE‐1) membrane protein (ankyrin successfully competes with oxy‐Hb for binding to AE‐1). Upon RBC deoxygenation (hypoxic RBC), oxy‐Hb R‐state is converted to deoxy‐Hb T‐state, which now successfully competes with ankyrin for the AE‐1 cytoplasmic domain (affinity for AE‐1 ranks as follows: deoxy‐Hb > ankyrin > oxy‐Hb). Proximal apposition of heme to the membrane and diminished cytoskeleton‐membrane interaction allows increased NO entry and affords intraerythrocytic Hb greater access to extraerythrocytic NO. Moreover, if deoxy‐Hb encounters high concentrations of NO, “super‐T” Hb‐α‐Fe(II)NO may form [in which NO bound to the α‐subunit disrupts normal heme‐globin linkage, locking Hb in the deoxy‐ or T‐conformation 211,312; see text for details]. (B) Increasing the proportion of intraerythrocytic T‐state Hb [by forming either deoxy‐Hb or α‐Fe(II)NO, both of which are T‐state tetramers and bind avidly to AE‐1] increases NO consumption by intact RBCs, as measured by a competition assay. In this plot, increased consumption in treated vs. control RBCs appears as an increased KRBC/KRBC control ratio (y‐axis) (+ P < 0.05, n = 4 to 8). (C) Increased NO uptake by NO‐pretreated hypoxic RBCs correlates with the formation of α‐Fe(II)NO (i.e., “super‐T” Hb) (+ P < 0.05, n = 4 for each sample). Reproduced with permission from 111.

Figure 7. Figure 7.

O2 exerts allosteric control over hemoglobin (Hb) S‐nitrosylation 149. (A) O2 loading accelerates S‐nitrosylation of Hb. Rates of Hb S‐nitrosylation by S‐nitrosothiol (SNO)‐cysteine are faster in the oxy conformation [HbFe(II)O2] than in the deoxy state [HbFe(II)]. (B) Deoxygenation accelerates denitrosylation of S‐nitroso‐hemoglobin (SNO‐Hb). Rates of SNO decomposition (and transnitrosylative transfer) are much faster in the deoxy conformation [SNO‐HbFe(II)] than in the oxy state [SNO‐HbFe(II)O2]. The decomposition of SNO‐HbFe(II) is further accelerated by the presence of excess glutathione (GSH) (Note: parallel to data in Figure 3). Within the dead time of the assay system (∼15 s), a major fraction of SNO‐HbFe(II) was converted to S‐nitroso‐glutathione (dashed line).

Figure 8. Figure 8.

Crystal structures and models of S‐nitrosothiol hemoglobin (SNO‐Hb). A crystal structure of R‐state SNO‐Hb has been published by Arnone and colleagues 38. SNO‐Hb did not crystallize in T‐state, consistent with thermodynamic predictions of instability [Note: subsequent work by Zhao and Houk suggests that SNO‐Cysβ93 in Hb crystals is a radical species 315]. Structural models of Hb and SNO‐Hb demonstrate that positioning of Cysβ93 and SNO‐βCys93 is dependent upon Hb conformation, consistent with crystallographic findings 262. (A) Deoxy‐Hb: Cysβ93 is exposed on the protein surface, above the external His146‐Asp94 salt bridge; Tyrβ145 packs alongside its side chain. The γ‐sulfur of Cysβ93 (yellow) is accessible to solvent in a cavity formed by the COOH‐terminus of β‐helix F, the COOH‐terminus of the β‐subunit, and helix C of the α2‐subunit. The α‐ and β‐carbons of Cysβ93 are gray. (B) Oxy‐Hb: Cysβ93 rotates in and away from solvent (and the broken salt bridge); Tyrβ145 is positioned over the top of its side chain. The γ‐sulfur of Cysβ93 is buried below its β‐carbon (gray) and is further shielded by structural changes. (C) SNO‐deoxy‐Hb: In the T‐structure, SNO is highly exposed to solvent (favoring NO group donation). The SNO is positioned either above or below the His146‐Asp94 salt bridge; if positioned up, the sulfur (yellow) and nitrogen (blue) are exposed, and if positioned down, the sulfur and oxygen (red) are exposed. (D) SNO‐oxy‐Hb: In the R‐structure, SNO is protected from solvent (disfavoring NO group donation). The NO group (blue and red) is accommodated behind the β‐heme, under Tyrβ145. The β‐carbon (gray) buries the sulfur (yellow). The SNO is also shielded by the backbone and side chain conformations.

Figure 9. Figure 9.

O2‐dependent variation in S‐nitrosothiol hemoglobin (SNO‐Hb) (▪) and Hb[FeNO] (□) demonstrate the association between Hb conformation and intramolecular heme → thiol migration of NO groups in red blood cells 196. (A‐D) Moles NO per mole Hb tetramer in arterial and mixed venous blood from humans breathing either 21% O2 at 1 ATA (absolute atmospheres) (A and C), 21% O2 at 0.56 ATA (equivalent to ∼12% O2 at 1 ATA) (B), or 100% O2 at 3 ATA (D). Total Hb‐bound NO equals the sum of the two bars for each condition. These data demonstrate O2‐dependent shuttling of Hb‐bound NO groups between heme and βCys93. (E and F) SNO content of blood Hb, presented as the fraction of NO‐Hb (% SNO), correlates with Hb O2 saturation (E) but not with pO2 (F). * P < 0.05, paired t‐test, indicates amounts of SNO‐Hb significantly different from zero; ** P < 0.05, paired t‐test, indicates a significant arterial‐venous difference in SNO‐Hb. Data represent the mean (s.e.m.) from 8‐11 individuals. See also Figure 3 showing similar results in human blood across a wide range of Hb O2 saturations.

Figure 10. Figure 10.

pO2‐regulated export of NO groups from red blood cells (RBCs) can occur via NO group transfer from S‐nitrosothiol hemoglobin (SNO‐Hb) to an extraerythrocytic thiol reactant 223. Circulatory transit was simulated for human whole blood in a thin‐film tonometer (under 5% CO2, balance N2, pH 7.4) after spiking the sample with the nonnative, low‐mass thiol, N‐acetylcysteine (NAC, 100 μM in plasma). The concentration of S‐nitroso‐N‐acetylcysteine (SNOAC) that formed in plasma was measured by mass spectrometry and confirmed by mass labeling with 15N. The conversion of extraerythrocytic NAC to SNOAC correlated with RBC O2 and SNO content. Serum SNOAC formed as a function of Hb So2. (A) Liquid chromatogram demonstrating coelution of RBC‐generated SNOAC with the 15N‐labeled SNOAC standard. (B) Mass spectrum demonstrating paired signals from RBC‐generated SNOAC and the 15N‐labeled SNOAC standard (m/z 194). (C) Extraerythrocytic SNOAC concentration follows oxy‐Hb desaturation (by co‐oximetry). “♦” ‐ extraerythrocytic SNOAc values indexed (as %) to amount measured when Hb So2 = 56%. (D) RBC SNO content (black) decreased in tandem with Hb O2 desaturation (dashed blue). Note that as SNO content in RBCs fell, extraerythrocytic SNOAC (yellow symbols) accumulated. Note also that SNOAC levels were below the limits of detection when the Hb O2 saturation was above 80%. These data indicate that the O2‐responsive vasoactivity demonstrated for human RBCs (see Fig. 3) can involve transfer of NO groups by RBCs to extraerythrocytic low‐mass thiols.

Figure 11. Figure 11.

The red blood cell (RBC) membrane furnishes a platform for O2‐regulated NO processing and export 226. (A) NO group ligation (Fe‐ or S‐) and disposition (cytosol or membrane) after exposure of intact RBCs to NO. Note that FeNO (representing HbFeNO) is predominantly cytosolic, whereas S‐nitrosothiol (SNO) is largely membrane associated. (B) The intrinsic membrane protein AE‐1 (band 3 protein) is trans‐S‐nitrosylated by S‐nitroso‐hemoglobin. SNO content of AE‐1 immunoprecipitates derived either from inside‐out RBC membrane vesicles (IOVs) incubated with free (B1) or sepharose‐bound SNO‐Hb (B2) or from membrane extracts of RBCs treated with NO (B3). Note that the cytoplasmic domain of AE‐1 contains two reactive cysteine thiols, which are exposed on the outer surface of RBC‐derived IOVs. (C) Membrane SNO is necessary for RBC‐mediated vasodilation. Representative traces display tension generated by aortic rings at high or low (hypoxic) pO2. At 95% O2, RBC IOVs S‐nitrosylated by Hb [as in (B)] above elicit relaxation, whereas NO‐treated intact RBCs [as in (A) above] elicit contraction. At ∼1% O2 (representative of tissue pO2), NO‐treated RBCs elicit relaxation, which is abrogated by the AE‐1 inhibitor DIDS. Addition of membranes derived from NO‐treated RBCs produces relaxation, whereas an equivalent aliquot of cytosol (containing HbFeNO) elicits contraction. (D) Cartoon illustrating a juxta‐membrane Hb population in which R → T transition promotes docking of deoxy‐Hb (via the β‐cleft or 2,3‐DPG binding cavity) to the cytoplasmic domain of AE‐1. Concomitantly, NO is transferred from βCys93 to a cysteine thiol within AE‐1. In addition, R‐to‐T transition may facilitate a shift in the intraerythrocytic equilibrium between SNO‐Hb and S‐nitroso‐glutathione (GSNO).

Figure 12. Figure 12.

O2‐responsive generation of S‐nitrosothiols (SNOs) from red blood cells (RBCs) signals the ventilatory response to hypoxia 178. After implantation of a cannula proximal to the brainstem nucleus tractus soliatarius (NTS), minute ventilation () was monitored in conscious rats, using whole‐body plethysmography. (A) during (shaded) and following a brief period of hypoxia demonstrated the characteristic hypoxic ventilatory response. (B) Injection into the NTS (at arrow) of the low‐mass SNO S‐nitrosocysteinylglycine (CGSNO, 1 nM) resulted in a marked increase in , with onset and decay characteristics identical to those observed in (A). (C) Injection of the low‐mass fraction from deoxygenated blood‐derived plasma (at arrow, black tracing) recapitulates the increase observed in (A) and (B); this response was absent when the low‐mass fraction from oxygenated blood was injected (gray). (D) Glutathione (GSH) reacts with deoxygenated but not oxygenated blood to form S‐nitroso‐glutathione (GSNO). Liquid chromatography/mass spectrometry was performed on plasma from blood to which 400 μM of GSH had been added. The GSNO peak (eluted at 5.82 min; blue) in the deoxygenated blood‐derived plasma was attenuated after ultraviolet treatment (violet), demonstrating SNO ligation and is not evident in the oxygenated blood‐derived plasma (red). These observations indicate that deoxygenated but not oxygenated blood reacts with reduced thiol species to form SNO (also see Fig. 8). (E) Mean differences in before and after the injection of the deoxygenated fractions (black) analyzed in (D); oxygenated fractions (gray) had no effect. Reproduced with permission from 178.



Figure 1.

Local vascular reflexes support maintenance of O2 delivery to tissue in the setting of progressive hypoxia. In a classic paper 244, Guyton demonstrated regional autoregulation of systemic blood flow in normal dogs (following spinal anesthesia) by observing variation in blood flow during constant pressure blood perfusion of the femoral artery, while reducing the hemoglobin oxygen saturation (Hb So2%) from 100% to 0% in the perfusing blood. (A) Stepwise reduction in Hb So2% caused a progressive increase in blood flow through the leg. (B) These data demonstrate that autoregulation of blood flow occurs at a local level and this regulation serves to improve oxygen supply when blood oxygen content falls. In addition, effects on blood flow were replicated by injecting partially deoxygenated versus oxygenated red blood cells into the artery, demonstrating that effects could be elicited during arteriovenous transit (<1 s). Reproduced with permission from 244.



Figure 2.

Red blood cells (RBCs) transduce regional O2 gradients in tissue to control nitric oxide (NO) bioactivity in plasma by trapping or delivering NO groups as a function of hemoglobin (Hb) O2 saturation. (A) In this fashion, circulating NO groups are processed by Hb into the highly vasoactive (thiol‐based) NO congener, S‐nitrosothiol (SNO). By exporting SNOs as a function of Hb deoxygenation, RBCs precisely dispense vasodilator bioactivity in direct proportion to regional blood flow lack. (B) O2 delivery homeostasis requires biochemical coupling of vessel tone to environmental cues that matches perfusion sufficiency to metabolic demand. Because oxy‐ and deoxy‐Hb process NO differently (see text), allosteric transitions in Hb conformation afford context‐responsive (O2‐coupled) control of NO bioavailability, thereby linking the sensor and effector arms of this system. Specifically, Hb conformation governs the equilibria among deoxy‐HbFeNO (A; NO sink), SNO‐oxy‐Hb (B; NO store), and acceptor thiols including the membrane protein SNO‐AE‐1 (C; bioactive NO source). Direct SNO export from RBCs or S‐transnitrosylation from RBCs to plasma thiols (D) or to endothelial cells directly (not shown) yields vasoactive SNOs, which influence resistance vessel caliber and close this signaling loop. Thus, RBCs either trap (A) or export (D) NO groups to optimize blood flow. (C) NO processing in RBCs (A and B) couples vessel tone to tissue pO2; this system subserves hypoxic vasodilation in the arterial periphery and thereby calibrates blood flow to regional tissue hypoxia. See Figures 10 and 11 and related discussion for more details.



Figure 3.

Hemoglobin oxygen saturation (Hb So2%) exerts coordinated governance of red blood cell (RBC) S‐nitrosothiol (SNO) content, RBC vasoactivity, and human peripheral blood flow. (A) Human blood gas measurements of SNO and O2 64. RBCs with (black) or without (red) added extracellular glutathione (GSH) were deoxygenated under inert gas. The natural logarithm of SNO content in RBCs (SNO RBC) is linearly related to Hb So2. GSH accelerated SNO RBC decay, consistent with O2‐linked export of NO groups to extraerythrocytic thiols. (B) RBCs induce graded relaxation of systemic arteries (aortic ring bioassay) that is inversely related to Hb So2 across the physiological range, recapitulating hypoxic vasodilation (HVD) 196. Hb So2 spanned from red (oxy, >90% Hb So2) to blue (deoxy, <40% Hb So2). (C) Leg vascular conductance increases as blood O2 content falls (HVD) 99. Hb So2 and thus arterial blood O2 content were manipulated by CO exposure ± varying Fio2 (fraction of inspired O2; hypoxia versus hyperoxia). Neither vascular conductance nor blood flow correlated with blood pO2 per se. Note the similarity between observations in panels A, B, and C and Guyton's original observations 244 (Fig. 1). Panel (C) Reproduced with permission from 99.



Figure 4.

Hypoxic vasodilation (1% O2) by native (untreated) human red blood cells (RBCs) of mouse aortic segments requires neither eNOS nor intact endothelium 62. (A) Hypoxiacoupled vasodilation by RBCs is preserved despite endothelial denudation, inhibition of NOS activity with L‐NAME, or eNOS deletion (eNOS‐/‐). RBC‐induced vasorelaxation was greatly attenuated by the guanylate cyclase inhibitor ODQ. (n = 9 to 34; * P < 0.05). (B, C) Representative tracings from the vascular ring preparation show that removal of endothelium eliminates acetylcholine (ACh)‐induced vasorelaxation but has no effect on RBC‐induced vasorelaxation [N.B. vessel rings are precontracted with phenylephrine (PE)].



Figure 5.

Hypoxic vasodilation (HVD) by native human red blood cells (RBCs) of rabbit aortic segments is S‐nitrosothiol (SNO) based 62. (A) RBC‐induced vasorelaxation was potentiated by cysteine and by N‐acetylcysteine but was unaffected by nitrite (NO2) or the prostaglandin synthesis inhibitors indomethacin and aspirin (all agents added 2 min before RBCs) (n = 5 to 11; * P < 0.05). (BD), Representative tracings illustrate the augmentation of relaxation by cysteine and the attenuation of relaxation by S‐nitroso‐hemoglobin depletion (∼80% depletion). (E) Addition of NO2 to the bath (even at supraphysiologic concentrations of 1 μM) before (NO2 → RBC), simultaneously with the addition of RBCs (RBC + NO2), or near the termination of RBC‐induced vasorelaxation (RBC → NO2), had no discernible effect on the magnitude or duration of RBC‐induced HVD.



Figure 6.

Conformation‐specific binding between hemoglobin (Hb) and the red blood cell (RBC) membrane protein AE‐1 affords O2 responsive control of NO trapping 111. (A) Under oxygenated conditions (normoxic RBC, at left), the RBC membrane constitutes a significant barrier to NO entry via tight association between the submembrane cytoskeleton and the cytoplasmic domain of the band 3 (AE‐1) membrane protein (ankyrin successfully competes with oxy‐Hb for binding to AE‐1). Upon RBC deoxygenation (hypoxic RBC), oxy‐Hb R‐state is converted to deoxy‐Hb T‐state, which now successfully competes with ankyrin for the AE‐1 cytoplasmic domain (affinity for AE‐1 ranks as follows: deoxy‐Hb > ankyrin > oxy‐Hb). Proximal apposition of heme to the membrane and diminished cytoskeleton‐membrane interaction allows increased NO entry and affords intraerythrocytic Hb greater access to extraerythrocytic NO. Moreover, if deoxy‐Hb encounters high concentrations of NO, “super‐T” Hb‐α‐Fe(II)NO may form [in which NO bound to the α‐subunit disrupts normal heme‐globin linkage, locking Hb in the deoxy‐ or T‐conformation 211,312; see text for details]. (B) Increasing the proportion of intraerythrocytic T‐state Hb [by forming either deoxy‐Hb or α‐Fe(II)NO, both of which are T‐state tetramers and bind avidly to AE‐1] increases NO consumption by intact RBCs, as measured by a competition assay. In this plot, increased consumption in treated vs. control RBCs appears as an increased KRBC/KRBC control ratio (y‐axis) (+ P < 0.05, n = 4 to 8). (C) Increased NO uptake by NO‐pretreated hypoxic RBCs correlates with the formation of α‐Fe(II)NO (i.e., “super‐T” Hb) (+ P < 0.05, n = 4 for each sample). Reproduced with permission from 111.



Figure 7.

O2 exerts allosteric control over hemoglobin (Hb) S‐nitrosylation 149. (A) O2 loading accelerates S‐nitrosylation of Hb. Rates of Hb S‐nitrosylation by S‐nitrosothiol (SNO)‐cysteine are faster in the oxy conformation [HbFe(II)O2] than in the deoxy state [HbFe(II)]. (B) Deoxygenation accelerates denitrosylation of S‐nitroso‐hemoglobin (SNO‐Hb). Rates of SNO decomposition (and transnitrosylative transfer) are much faster in the deoxy conformation [SNO‐HbFe(II)] than in the oxy state [SNO‐HbFe(II)O2]. The decomposition of SNO‐HbFe(II) is further accelerated by the presence of excess glutathione (GSH) (Note: parallel to data in Figure 3). Within the dead time of the assay system (∼15 s), a major fraction of SNO‐HbFe(II) was converted to S‐nitroso‐glutathione (dashed line).



Figure 8.

Crystal structures and models of S‐nitrosothiol hemoglobin (SNO‐Hb). A crystal structure of R‐state SNO‐Hb has been published by Arnone and colleagues 38. SNO‐Hb did not crystallize in T‐state, consistent with thermodynamic predictions of instability [Note: subsequent work by Zhao and Houk suggests that SNO‐Cysβ93 in Hb crystals is a radical species 315]. Structural models of Hb and SNO‐Hb demonstrate that positioning of Cysβ93 and SNO‐βCys93 is dependent upon Hb conformation, consistent with crystallographic findings 262. (A) Deoxy‐Hb: Cysβ93 is exposed on the protein surface, above the external His146‐Asp94 salt bridge; Tyrβ145 packs alongside its side chain. The γ‐sulfur of Cysβ93 (yellow) is accessible to solvent in a cavity formed by the COOH‐terminus of β‐helix F, the COOH‐terminus of the β‐subunit, and helix C of the α2‐subunit. The α‐ and β‐carbons of Cysβ93 are gray. (B) Oxy‐Hb: Cysβ93 rotates in and away from solvent (and the broken salt bridge); Tyrβ145 is positioned over the top of its side chain. The γ‐sulfur of Cysβ93 is buried below its β‐carbon (gray) and is further shielded by structural changes. (C) SNO‐deoxy‐Hb: In the T‐structure, SNO is highly exposed to solvent (favoring NO group donation). The SNO is positioned either above or below the His146‐Asp94 salt bridge; if positioned up, the sulfur (yellow) and nitrogen (blue) are exposed, and if positioned down, the sulfur and oxygen (red) are exposed. (D) SNO‐oxy‐Hb: In the R‐structure, SNO is protected from solvent (disfavoring NO group donation). The NO group (blue and red) is accommodated behind the β‐heme, under Tyrβ145. The β‐carbon (gray) buries the sulfur (yellow). The SNO is also shielded by the backbone and side chain conformations.



Figure 9.

O2‐dependent variation in S‐nitrosothiol hemoglobin (SNO‐Hb) (▪) and Hb[FeNO] (□) demonstrate the association between Hb conformation and intramolecular heme → thiol migration of NO groups in red blood cells 196. (A‐D) Moles NO per mole Hb tetramer in arterial and mixed venous blood from humans breathing either 21% O2 at 1 ATA (absolute atmospheres) (A and C), 21% O2 at 0.56 ATA (equivalent to ∼12% O2 at 1 ATA) (B), or 100% O2 at 3 ATA (D). Total Hb‐bound NO equals the sum of the two bars for each condition. These data demonstrate O2‐dependent shuttling of Hb‐bound NO groups between heme and βCys93. (E and F) SNO content of blood Hb, presented as the fraction of NO‐Hb (% SNO), correlates with Hb O2 saturation (E) but not with pO2 (F). * P < 0.05, paired t‐test, indicates amounts of SNO‐Hb significantly different from zero; ** P < 0.05, paired t‐test, indicates a significant arterial‐venous difference in SNO‐Hb. Data represent the mean (s.e.m.) from 8‐11 individuals. See also Figure 3 showing similar results in human blood across a wide range of Hb O2 saturations.



Figure 10.

pO2‐regulated export of NO groups from red blood cells (RBCs) can occur via NO group transfer from S‐nitrosothiol hemoglobin (SNO‐Hb) to an extraerythrocytic thiol reactant 223. Circulatory transit was simulated for human whole blood in a thin‐film tonometer (under 5% CO2, balance N2, pH 7.4) after spiking the sample with the nonnative, low‐mass thiol, N‐acetylcysteine (NAC, 100 μM in plasma). The concentration of S‐nitroso‐N‐acetylcysteine (SNOAC) that formed in plasma was measured by mass spectrometry and confirmed by mass labeling with 15N. The conversion of extraerythrocytic NAC to SNOAC correlated with RBC O2 and SNO content. Serum SNOAC formed as a function of Hb So2. (A) Liquid chromatogram demonstrating coelution of RBC‐generated SNOAC with the 15N‐labeled SNOAC standard. (B) Mass spectrum demonstrating paired signals from RBC‐generated SNOAC and the 15N‐labeled SNOAC standard (m/z 194). (C) Extraerythrocytic SNOAC concentration follows oxy‐Hb desaturation (by co‐oximetry). “♦” ‐ extraerythrocytic SNOAc values indexed (as %) to amount measured when Hb So2 = 56%. (D) RBC SNO content (black) decreased in tandem with Hb O2 desaturation (dashed blue). Note that as SNO content in RBCs fell, extraerythrocytic SNOAC (yellow symbols) accumulated. Note also that SNOAC levels were below the limits of detection when the Hb O2 saturation was above 80%. These data indicate that the O2‐responsive vasoactivity demonstrated for human RBCs (see Fig. 3) can involve transfer of NO groups by RBCs to extraerythrocytic low‐mass thiols.



Figure 11.

The red blood cell (RBC) membrane furnishes a platform for O2‐regulated NO processing and export 226. (A) NO group ligation (Fe‐ or S‐) and disposition (cytosol or membrane) after exposure of intact RBCs to NO. Note that FeNO (representing HbFeNO) is predominantly cytosolic, whereas S‐nitrosothiol (SNO) is largely membrane associated. (B) The intrinsic membrane protein AE‐1 (band 3 protein) is trans‐S‐nitrosylated by S‐nitroso‐hemoglobin. SNO content of AE‐1 immunoprecipitates derived either from inside‐out RBC membrane vesicles (IOVs) incubated with free (B1) or sepharose‐bound SNO‐Hb (B2) or from membrane extracts of RBCs treated with NO (B3). Note that the cytoplasmic domain of AE‐1 contains two reactive cysteine thiols, which are exposed on the outer surface of RBC‐derived IOVs. (C) Membrane SNO is necessary for RBC‐mediated vasodilation. Representative traces display tension generated by aortic rings at high or low (hypoxic) pO2. At 95% O2, RBC IOVs S‐nitrosylated by Hb [as in (B)] above elicit relaxation, whereas NO‐treated intact RBCs [as in (A) above] elicit contraction. At ∼1% O2 (representative of tissue pO2), NO‐treated RBCs elicit relaxation, which is abrogated by the AE‐1 inhibitor DIDS. Addition of membranes derived from NO‐treated RBCs produces relaxation, whereas an equivalent aliquot of cytosol (containing HbFeNO) elicits contraction. (D) Cartoon illustrating a juxta‐membrane Hb population in which R → T transition promotes docking of deoxy‐Hb (via the β‐cleft or 2,3‐DPG binding cavity) to the cytoplasmic domain of AE‐1. Concomitantly, NO is transferred from βCys93 to a cysteine thiol within AE‐1. In addition, R‐to‐T transition may facilitate a shift in the intraerythrocytic equilibrium between SNO‐Hb and S‐nitroso‐glutathione (GSNO).



Figure 12.

O2‐responsive generation of S‐nitrosothiols (SNOs) from red blood cells (RBCs) signals the ventilatory response to hypoxia 178. After implantation of a cannula proximal to the brainstem nucleus tractus soliatarius (NTS), minute ventilation () was monitored in conscious rats, using whole‐body plethysmography. (A) during (shaded) and following a brief period of hypoxia demonstrated the characteristic hypoxic ventilatory response. (B) Injection into the NTS (at arrow) of the low‐mass SNO S‐nitrosocysteinylglycine (CGSNO, 1 nM) resulted in a marked increase in , with onset and decay characteristics identical to those observed in (A). (C) Injection of the low‐mass fraction from deoxygenated blood‐derived plasma (at arrow, black tracing) recapitulates the increase observed in (A) and (B); this response was absent when the low‐mass fraction from oxygenated blood was injected (gray). (D) Glutathione (GSH) reacts with deoxygenated but not oxygenated blood to form S‐nitroso‐glutathione (GSNO). Liquid chromatography/mass spectrometry was performed on plasma from blood to which 400 μM of GSH had been added. The GSNO peak (eluted at 5.82 min; blue) in the deoxygenated blood‐derived plasma was attenuated after ultraviolet treatment (violet), demonstrating SNO ligation and is not evident in the oxygenated blood‐derived plasma (red). These observations indicate that deoxygenated but not oxygenated blood reacts with reduced thiol species to form SNO (also see Fig. 8). (E) Mean differences in before and after the injection of the deoxygenated fractions (black) analyzed in (D); oxygenated fractions (gray) had no effect. Reproduced with permission from 178.

References
 1. Abman SH, Kinsella JP. Inhaled nitric oxide therapy for pulmonary disease in pediatrics. Curr Opin Pediatr 10: 236–242, 1998.
 2. Abraham E. Neutrophils and acute lung injury. Crit Care Med 31: S195–S199, 2003.
 3. Abraham E, Matthay MA, Dinarello CA, Vincent JL, Cohen J, Opal SM, Glauser M, Parsons P, Fisher F, Repine JE. Consensus conference definitions for sepsis, septic shock, acute lung injury, and acute respiratory distress syndrome: Time for a reevaluation. Crit Care Med 28: 232–235, 2000.
 4. Agarwal R, Aggarwal AN, Gupta D, Behera D, Jindal SK. Etiology and outcomes of pulmonary and extrapulmonary acute lung injury/ARDS in a respiratory ICU in north India. Chest 130: 724–729, 2006.
 5. Alayash AI. Oxygen therapeutics: Can we tame haemoglobin? Nat Rev Drug Discov 3: 152–159, 2004.
 6. Allen BW, Piantadosi CA. How do red blood cells cause hypoxic vasodilation? The SNO‐hemoglobin paradigm. Am J Physiol Heart Circ Physiol 291: H1507–H1512, 2006.
 7. Angelo M, Hausladen A, Singel DJ, Stamler JS, Robert KP. Interactions of NO with hemoglobin: From microbes to man. In: Methods in Enzymology Globins and Other Nitric Oxide‐Reactive Proteins, Part A. Elsevier Academic Press, San Diego, CA, 2008, p. 131–168.
 8. Angelo M, Singel DJ, Stamler JS. An S‐nitrosothiol (SNO) synthase function of hemoglobin that utilizes nitrite as a substrate. Proc Natl Acad Sci U S A 103: 8366–8371, 2006.
 9. Angus DC, Linde‐Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR. Epidemiology of severe sepsis in the United States: Analysis of incidence, outcome, and associated costs of care. Crit Care Med 29: 1303–1310, 2001.
 10. Arciero JC, Carlson BE, Secomb TW. Theoretical model of metabolic blood flow regulation: Roles of ATP release by red blood cells and conducted responses. Am J Physiol Heart Circ Physiol 295: H1562–H1571, 2008.
 11. Arnelle DR, Stamler JS. NO+, NO, and NO− donation by S‐nitrosothiols: Implications for regulation of physiological functions by S‐nitrosylation and acceleration of disulfide formation. Arch Biochem Biophys 318: 279–285, 1995.
 12. Arnold WP, Mittla CK, Katsuki S, Murad F. Nitric oxide activates guanylate cyclase and increases guanosine 3′:5′‐cyclic monophosphate levels in various tissue preparations. Proc Natl Acad Sci U S A 74: 3203–3207, 1977.
 13. Azarov I, Huang KT, Basu S, Gladwin MT, Hogg N, Kim‐Shapiro DB. Nitric oxide scavenging by red blood cells as a function of hematocrit and oxygenation. J Biol Chem 280: 39024–39032, 2005.
 14. Barry PH, Diamond JM. Effects of unstirred layers on membrane phenomena. Physiol Rev 64: 763–872, 1984.
 15. Barvitenko NN, Adragna NC, Weber RE. Erythrocyte signal transduction pathways, their oxygenation dependence and functional significance. Cell Physiol Biochem 15: 1–18, 2005.
 16. Bennett‐Guerrero E, Veldman TH, Doctor A, Telen MJ, Ortel TL, Reid TS, Mulherin MA, Zhu H, Buck RD, Califf RM, McMahon TJ. Evolution of adverse changes in stored RBCs. Proc Natl Acad Sci U S A 104: 17063–17068, 2007.
 17. Besarab A. The effects of normal as compared to low hematocrit values in patients with cardiac disease who are receiving hemodialysis and epoetin. N Engl J Med 339: 584–590, 1998.
 18. Beuhler PW, Layash AI. All hemoglobin based oxygen carriers are not created equally. Biochim Biophys Acta 1784: 1378–1381, 2008.
 19. Bhattacharya J, Staub NC. Direct measurement of microvascular pressures in the isolated perfused dog lung. Science 210: 327–328, 1980.
 20. Bonaventura C, Taboy CH, Low PS, Stevens RD, Lafon C, Crumbliss AL. Heme redox properties of S‐nitrosated hemoglobin Ao and hemoglobin S. Implications for interactions of nitric oxide with normal and sickle red blood cells. J Biol Chem 277: 14557–14563, 2002.
 21. Bone RC. Toward an epidemiology and natural history of SIRS (systemic inflammatory response syndrome). JAMA 268: 3452–3455, 1992.
 22. Bone RC. Immunologic dissonance: A continuing evolution in our understanding of the systemic inflammatory response syndrome (SIRS) and the multiple organ dysfunction syndrome (MODS). Ann Intern Med 125: 680–687, 1996.
 23. Bone RC, Grodzin CJ, Balk RA. Sepsis: A new hypothesis for pathogenesis of the disease process. Chest 112: 235–243, 1997.
 24. Bosworth CA, Toledo JC, Zmijewski JW, Li Q, Lancaster JR. Dinitrosyl iron complexes and the mechanism(s) of cellular protein nitrosothiol formation from nitric oxide. Proc Natl Acad Sci U S A 106: 4671–4676, 2009.
 25. Brower RG, Gottlieb J, Wise RA, Permutt S, Sylvester JT. Locus of hypoxic vasoconstriction in isolated ferret lungs. J Appl Physiol 63: 58–65, 1987.
 26. Brunner F, Leonhard B, Kukovetz WR, Mayer B. Role of endothelin, nitric oxide and l‐arginine release in ischaemia/reperfusion injury of rat heart. Cardiovasc Res 36: 60–66, 1997.
 27. Brunner F, Stessel H, Kukovetz WR. Novel guanylyl cyclase inhibitor, ODQ reveals role of nitric oxide, but not of cyclic GMP in endothelin‐1 secretion. FEBS Lett 376: 262–266, 1995.
 28. Brunori M, Taylor JF, Antonini E, Wyman J, Rossi‐Fanelli A. Studies on the oxidation‐reduction potentials of heme proteins. VI. Human hemoglobin treated with various sulfhydryl reagents. J Biol Chem 242: 2295–2300, 1967.
 29. Buehler PW, Alayash AI. All hemoglobin‐based oxygen carriers are not created equally. Biochim Biophys Acta 1784: 1378–1381, 2008.
 30. Bunn HF, Nathan DG, Dover GJ, Hebbel RP, Platt OS, Rosse WF, Ware RE. Pulmonary hypertension and nitric oxide depletion in sickle cell disease. Blood 116(5): 687–692, 2010.
 31. Butler AR, Megson IL, Wright PG. Diffusion of nitric oxide and scavenging by blood in the vasculature. Biochim Biophys Acta 1425: 168–176, 1998.
 32. Cannon RO III, Schechter AN, Panza JA, Ognibene FP, Pease‐Fye ME, Waclawiw MA, Shelhamer JH, Gladwin MT. Effects of inhaled nitric oxide on regional blood flow are consistent with intravascular nitric oxide delivery. J Clin Invest 108: 279–287, 2001.
 33. Carlsen E, Comroe JH. The rate of uptake of carbon monoxide and of nitric oxide by normal human erythrocytes and experimentally produced spherocytes. J Gen Physiol 42: 83–107, 1958.
 34. Cedergvist B, Persson MG, Gustafsson LE. Direct demonstration of NO formation in vivo from organic nitrites and nitrates, and correlation to effects on blood pressure and to in vitro effects. Biochem Pharmacol 47: 1047–1053, 1994.
 35. Ceron PÃIB, Cremonez DC, Bendhack LM, Tedesco AC. The relaxation induced by S‐nitroso‐glutathione and S‐nitroso‐N‐acetylcysteine in rat aorta is not related to nitric oxide production. J Pharmacol Exp Ther 298: 686–694, 2001.
 36. Chakraborty S, Balakotaiah V, Bidani A. Diffusing capacity reexamined: Relative roles of diffusion and chemical reaction in red cell uptake of O2, CO, CO2, and NO. J Appl Physiol 97: 2284–2302, 2004.
 37. Chan NL, Kavanaugh JS, Rogers PH, Arnone A. Crystallographic analysis of the interaction of nitric oxide with quaternary‐T human hemoglobin. Biochemistry 43: 118–132, 2004.
 38. Chan NL, Rogers PH, Arnone A. Crystal structure of the S‐nitroso form of liganded human hemoglobin. Biochemistry 37: 16459–16464, 1998.
 39. Chen X, Jaron D, Barbee KA, Buerk DG. The influence of radial RBC distribution, blood velocity profiles, and glycocalyx on coupled NO/O2 transport. J Appl Physiol 100: 482–492, 2006.
 40. Chetrite G, Cassoly R. Affinity of hemoglobin for the cytoplasmic fragment of human erythrocytemembrane band 3. Equilibrium measurements at physiological pH using matrix‐bound proteins: The effects of ionic strength, deoxygenation and of 2,3‐diphosphoglycerate. J Mol Biol 185: 639–644, 1985.
 41. Chiancone E, Currell DL, Vecchini P, Antonini E, Wyman J. Kinetics of the reaction of the “masked” and “free” sulfhydryl groups of human hemoglobin with p‐mercuribenzoate. J Biol Chem 245: 4105–4111, 1970.
 42. Chu H, Breite A, Ciraolo P, Franco RS, Low PS. Characterization of the deoxyhemoglobin binding site on human erythrocyte band 3: Implications for O2 regulation of erythrocyte properties. Blood 111: 932–938, 2008.
 43. Cirillo M, Laurenzi M, Trevisan M, Stamler J. Hematocrit, blood pressure, and hypertension. The Gubbio Population Study. Hypertension 20: 319–326, 1992.
 44. Clough AV, Haworth ST, Ma W, Dawson CA. Effects of hypoxia on pulmonary microvascular volume. Am J Physiol Heart Circ Physiol 279: H1274–1282, 2000.
 45. Cosby K, Partovi KS, Crawford JH, Patel RP, Reiter CD, Martyr S, Yang BK, Waclawiw MA, Zalos G, Xu X, Huang KT, Shields H, Kim‐Shapiro DB, Schechter AN, Cannon RO III, Gladwin MT. Nitrite reduction to nitric oxide by deoxyhemoglobin vasodilates the human circulation. Nat Med 9: 1498–1505, 2003.
 46. Crawford JH, Chacko BK, Kevil CG, Patel RP. The red blood cell and vascular function in health and disease. Antioxid Redox Signal 6: 992–999, 2004.
 47. Crawford JH, Chacko BK, Pruitt HM, Piknova B, Hogg N, Patel RP. Transduction of NO‐bioactivity by the red blood cell in sepsis: Novel mechanisms of vasodilation during acute inflammatory disease. Blood 104: 1375–1382, 2004.
 48. Crawford JH, Isbell TS, Huang Z, Shiva S, Chacko BK, Schechter AN, Darley‐Usmar VM, Kerby JD, Lang JD Jr, Kraus D, Ho C, Gladwin MT, Patel RP. Hypoxia, red blood cells, and nitrite regulate NO‐dependent hypoxic vasodilation. Blood 107: 566–574, 2006.
 49. Crawford JH, White CR, Patel RP. Vasoactivity of S‐nitrosohemoglobin: Role of oxygen, heme, and NO oxidation states. Blood 101: 4408–4415, 2003.
 50. Creagh‐Brown B, Griffiths M, Evans T. Bench‐to‐bedside review: Inhaled nitric oxide therapy in adults. Crit Care 13: 221, 2009.
 51. Daiber A, Schildknecht S, Muller J, Kamuf J, Bachschmid MM, Ullrich V. Chemical model systems for cellular nitros(yl)ation reactions. Free Radic Biol Med 47: 458–467, 2009.
 52. Dalsgaard T, Simonsen U, Fago A. Nitrite‐dependent vasodilation is facilitated by hypoxia and is independent of known NO‐generating nitrite reductase activities. Am J Physiol Heart Circ Physiol 292: H3072–H3078, 2007.
 53. Datta B, Tufnell‐Barrett T, Bleasdale RA, Jones CJ, Beeton I, Paul V, Frenneaux M, James P. Red blood cell nitric oxide as an endocrine vasoregulator: A potential role in congestive heart failure. Circulation 109: 1339–1342, 2004.
 54. Davies P, Burke G, Reid L. The structure of the wall of the rat intraacinar pulmonary artery: An electron microscopic study of microdissected preparations. Microvasc Res 32: 50–63, 1986.
 55. Davison GW, George L, Jackson SK, Young IS, Davies B, Bailey DM, Peters JR, Ashton T. Exercise, free radicals, and lipid peroxidation in type 1 diabetes mellitus. Free Radic Biol Med 33: 1543–1551, 2002.
 56. Davisson RL, Travis MD, Bates JN, Lewis SJ. Hemodynamic effects of l‐ and d‐S‐nitrosocysteine in the rat. Stereoselective S‐nitrosothiol recognition sites. Circ Res 79: 256–262, 1996.
 57. Dawson CA, Grimm DJ, Linehan JH. Influence of hypoxia on the longitudinal distribution of pulmonary vascular resistance. J Appl Physiol 44: 493–498, 1978.
 58. Deem S, Bishop MJ, Alberts MK. Effect of anemia on intrapulmonary shunt during atelectasis in rabbits. J Appl Physiol 79: 1951–1957, 1995.
 59. Deem S, Kim JU, Manjula BN, Acharya AS, Kerr ME, Patel RP, Gladwin MT, Swenson ER. Effects of S‐nitrosation and cross‐linking of hemoglobin on hypoxic pulmonary vasoconstriction in isolated rat lungs. Circ Res 91: 626–632, 2002.
 60. Deem S, Kim SS, Min JH, Eveland R, Moulding J, Martyr S, Wang X, Swenson ER, Gladwin MT. Pulmonary vascular effects of red blood cells containing S‐nitrosated hemoglobin. Am J Physiol Heart Circ Physiol 287(6): H2561‐H2568, 2004.
 61. Deem S, Swenson ER, Alberts MK, Hedges RG, Bishop MJ. Red blood cell augmentation of hypoxic pulmonary vasoconstriction: Hematocrit dependence and the importance of nitric oxide. Am J Respir Crit Care Med 157: 1181–1186, 1998.
 62. Diesen DL, Hess DT, Stamler JS. Hypoxic vasodilation by red blood cells: Evidence for an S‐nitrosothiol‐based signal. Circ Res 103: 545–553, 2008.
 63. Doctor A, Gaston B, Kim‐Shapiro DB. Detecting physiologic fluctuations in the S‐nitrosohemoglobin micropopulation: Triiodide versus 3C. Blood 108: 3225–3227, 2006.
 64. Doctor A, Platt R, Sheram ML, Eischeid A, McMahon T, Maxey T, Doherty J, Axelrod M, Kline J, Gurka M, Gow A, Gaston B. Hemoglobin conformation couples erythrocyte S‐nitrosothiol content to O2 gradients. Proc Natl Acad Sci U S A 102: 5709–5714, 2005.
 65. Doerschuk CM. Mechanisms of leukocyte sequestration in inflamed lungs. Microcirculation 8: 71–88, 2001.
 66. Doughty LA, Kaplan SS, Carcillo JA. Inflammatory cytokine and nitric oxide responses in pediatric sepsis and organ failure. Crit Care Med 24: 1137–1143, 1996.
 67. Doyle MP, Hoekstra JW. Oxidation of nitrogen oxides by bound dioxygen in hemoproteins. J Inorg Biochem 14: 351–358, 1981.
 68. Doyle MP, Pickering RA, DeWeert TM, Hoekstra JW, Pater D. Kinetics and mechanism of the oxidation of human deoxyhemoglobin by nitrites. J Biol Chem 256: 12393–12398, 1981.
 69. Duke H, Killick E. Pulmonary vasomotor responses of isolated perfused cat lungs to anoxia. J Physiol 117: 303–315, 1952.
 70. Duling BR, Berne RM. Longitudinal gradients in periarteriolar oxygen tension. A possible mechanism for the participation of oxygen in local regulation of blood flow. Circ Res 27: 669–678, 1970.
 71. Duling BR, Berne RM. Oxygen and the local regulation of blood flow; possible significance of longitudinal gradients in arterial blood oxygen tension. Circ Res 28 (Suppl 1): 65–69, 1971.
 72. Ellsworth ML, Ellis CG, Goldman D, Stephenson AH, Dietrich HH, Sprague RS. Erythrocytes: Oxygen sensors and modulators of vascular tone. Physiology 24: 107–116, 2009.
 73.From the bench to the bedside: The future of sepsis research. Executive summary of an American College of Chest Physicians, National Institute of Allergy and Infectious Disease, and National Heart, Lung, and Blood Institute Workshop. Chest 111: 744–753, 1997.
 74. Ferranti P, Malorni A, Mamone G, Sannolo N, Marino G. Characterisation of S‐nitrosohaemoglobin by mass spectrometry. FEBS Lett 400: 19–24, 1997.
 75. Ferranti P, Mamone G, Malorni A. Preparation and mass spectometric analysis of S‐nitrosohemoglobin. Methods Mol Biol 146: 147–165, 2000.
 76. Fischer SR, Deyo DJ, Bone HG, McGuire R, Traber LD, Traber DL. Nitric oxide synthase inhibition restores hypoxic pulmonary vasoconstriction in sepsis. Am J Respir Crit Care Med 156: 833–839, 1997.
 77. Foresti R, Clark JE, Green CJ, Motterlini R. Thiol compounds interact with nitric oxide in regulating heme oxygenase‐1 induction in endothelial cells. Involvement of superoxide and peroxynitrite anions. J Biol Chem 272: 18411–18417, 1997.
 78. Fortune JB, FeustelP.J, Saifi J, Stratton HH, Newell JC, Shah DM. Influence of hematocrit on cardiopulmonary function after acute hemorrhage. J Trauma 27: 243–249, 1987.
 79. Foster MW, McMahon TJ, Stamler JS. S‐Nitrosylation in health and disease. Trends Mol Med 9: 160–168, 2003.
 80. Fox GA, McCormack DG. A new look at the pulmonary circulation in acute lung injury. Thorax 47: 743–747, 1992.
 81. Fox‐Robichaud A, Payne D, Hasan SU, Ostrovsky L, Fairhead T, Reinhardt P, Kubes P. Inhaled NO as a viable antiadhesive therapy for ischemia/reperfusion injury of distal microvascular beds. J Clin Invest 101: 2497–2505, 1998.
 82. Fox‐Robichaud A, Payne D, Kubes P. Inhaled NO reaches distal vasculatures to inhibit endothelium—but not leukocyte‐dependent cell adhesion. Am J Physiol Lung Cell Mol Physiol 277: L1224–L1231, 1999.
 83. Frandsen U, Bangsbo J, Sander M, Hoffner L, Betak A, Saltin B, Hellsten Y. Exercise‐induced hyperaemia and leg oxygen uptake are not altered during effective inhibition of nitric oxide synthase with N(G)‐nitro‐l‐arginine methyl ester in humans. J Physiol 531: 257–264, 2001.
 84. Frei AC, Guo Y, Jones DW, Pritchard KA Jr, Fagan KA, Hogg N, Wandersee NJ. Vascular dysfunction in a murine model of severe hemolysis. Blood 112: 398–405, 2008.
 85. Fujii M, Hori H, Miyazaki G, Morimoto H, Yonetani T. The porphyrin‐iron hybrid hemoglobins. Absence of the Fe‐His bonds in one type of subunits favors a deoxy‐like structure with low oxygen affinity. J Biol Chem 268: 15386–15393, 1993.
 86. Funai EF, Davidson A, Seligman SP, Finlay TH. S‐Nitrosohemoglobin in the fetal circulation may represent a cycle for blood pressure regulation. Biochem Biophys Res Commun 239: 875–877, 1997.
 87. Furchgott RF, Zawadzki JV. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature 288: 373–376, 1980.
 88. Gaston B. Nitric oxide and thiol groups [Review]. Biochim Biophys Acta 1411: 323–333, 1999.
 89. Gaston B, Reilly J, Drazen JM, Fackler J, Ramdev P, Arnelle D, Mullins ME, Sugarbaker DJ, Chee C, Singel DJ. Endogenous nitrogen oxides and bronchodilator S‐nitrosothiols in human airways. Proc Natl Acad Sci U S A 90: 10957–10961, 1993.
 90. Gaston B, Singel D, Doctor A, Stamler JS. S‐Nitrosothiol signaling in respiratory biology. Am J Respir Crit Care Med 173: 1186–1193, 2006.
 91. Gattinoni L, Pelosi P, Suter PM, Pedoto A, Vercesi P, Lissoni A. Acute respiratory distress syndrome caused by pulmonary and extrapulmonary disease, different syndromes? Am J Respir Crit Care Med 158: 3–11, 1998.
 92. Gerbino AJ, McKinney S, Glenny RW. Correlation between ventilation and perfusion determines V A/Q heterogeneity in endotoxemia. J Appl Physiol 88: 1933–1942, 2000.
 93. Gladwin MT. Evidence mounts that nitrite contributes to hypoxic vasodilation in the human circulation. Circulation 117: 594–597, 2008.
 94. Gladwin MT, Ognibene FP, Pannell LK, Nichols JS, Pease‐Fye ME, Shelhamer JH, Schechter AN. Relative role of heme nitrosylation and beta‐cysteine 93 nitrosation in the transport and metabolism of nitric oxide by hemoglobin in the human circulation. Proc Natl Acad Sci U S A 97: 9943–9948, 2000.
 95. Gladwin MT, Shelhamer JH, Schechter AN, Pease‐Fye ME, Waclawiw MA, Panza JA, Ognibene FP, Cannon RO III. Role of circulating nitrite and S‐nitrosohemoglobin in the regulation of regional blood flow in humans. Proc Natl Acad Sci U S A 97: 11482–11487, 2000.
 96. Gladwin MT, Wang X, Reiter CD, Yang BK, Vivas EX, Bonaventura C, Schechter AN. S‐nitrosohemoglobin is unstable in the reductive erythrocyte environment and lacks O2/NO‐linked allosteric function. J Biol Chem 277: 27818–27828, 2002.
 97. Gonzalez‐Alonso J, Mortensen SP, Dawson EA, Secher NH, Damsgaard R. Erythrocytes and the regulation of human skeletal muscle blood flow and oxygen delivery: Role of erythrocyte count and oxygenation state of haemoglobin. J Physiol 572: 295–305, 2006.
 98. Gonzalez‐Alonso J, Olsen DB, Saltin B. Erythrocyte and the regulation of human skeletal muscle blood flow and oxygen delivery: Role of circulating ATP. Circ Res 91: 1046–1055, 2002.
 99. Gonzalez‐Alonso J, Richardson RS, Saltin B. Exercising skeletal muscle blood flow in humans responds to reduction in arterial oxyhaemoglobin, but not to altered free oxygen. J Physiol Online 530: 331–341, 2001.
 100. Gow A, Doctor A, Mannick J, Gaston B. S‐Nitrosothiol measurements in biological systems. J Chromatogr B 851: 140–151, 2007.
 101. Gow AJ, Buerk DG, Ischiropoulos H. A novel reaction mechanism for the formation of S‐nitrosothiol in vivo. J Biol Chem 272: 2841–2845, 1997.
 102. Gow AJ, Farkouh CR, Munson DA, Posencheg MA, Ischiropoulos H. Biological significance of nitric oxide‐mediated protein modifications. Am J Physiol Lung Cell Mol Physiol 287: L262–L268, 2004.
 103. Gow AJ, Luchsinger BP, Pawloski JR, Singel DJ, Stamler JS. The oxyhemoglobin reaction of nitric oxide. Proc Natl Acad Sci U S A 96: 9027–9032, 1999.
 104. Gow AJ, Stamler JS. Reactions between nitric oxide and haemoglobin under physiological conditions. Nature 391: 169–173, 1998.
 105. Granillo OM, Brahmajothi MV, Li S, Whorton AR, Mason SN, McMahon TJ, Auten RL. Pulmonary alveolar epithelial uptake of S‐nitrosothiols is regulated by L‐type amino acid transporter. Am J Physiol Lung Cell Mol Physiol 295: L38–L43, 2008.
 106. Guastadisegni C, Minghetti L, Nicolini A, Polazzi A, Levi G. Prostaglandin E2 synthesis is differentially affected by reactive nitrogen intermediates in cultured rat microglia and RAW 264.7 cells. FEBS Lett 413: 314–318, 1997.
 107. Gurney AM. Multiple sites of oxygen sensing and their contributions to hypoxic pulmonary vasoconstriction. Respir Physiol Neurobiol 132: 43–53, 2002.
 108. Guz A, Kurland GS, Freedberg AS. Relation of coronary flow to oxygen supply. Am J Physiol 199: 179–182, 1960.
 109. Hammerman SI, Klings ES, Hendra KP, Upchurch GR Jr, Rishikof DC, Loscalzo J, Farber HW. Endothelial cell nitric oxide production in acute chest syndrome. Am J Physiol Heart Circ Physiol 277: H1579–H1592, 1999.
 110. Han TH, Liao JC. Erythrocyte nitric oxide transport reduced by a submembrane cytoskeletal barrier. Biochim Biophys Acta 1723: 135–142, 2005.
 111. Han TH, Qamirani E, Nelson AG, Hyduke DR, Chaudhuri G, Kuo L, Liao JC. Regulation of nitric oxide consumption by hypoxic red blood cells. Proc Natl Acad Sci U S A 100: 12504–12509, 2003.
 112. Hanssen H, Brunini TM, Conway M, Banning AP, Roberts NB, Mann GE, Ellory JC, Mendes RAC. Increased l‐argenine transport in human erythrocytes in chronic heart failure. Clin Sci (Lond) 94: 43–48, 1998.
 113. Harada S, Tokunaga S, Momohara M, Masaki H, Tagawa T, Imaizumi T, Takeshita A. Inhibition of nitric oxide formation in the nucleus tractus solitarius increases renal sympathetic nerve activity in rabbits. Circ Res 72: 511–516, 1993.
 114. Hare JM, Stamler JS. NO/redox disequilibrium in the failing heart and cardiovascular system. J Clin Invest 115: 509–517, 2005.
 115. Hart RG, Kanter MC. Hematologic disorders and ischemic stroke. Stroke 21: 1111–1121, 1990.
 116. Hausladen A, Rafikov R, Angelo M, Singel DJ, Nudler E, Stamler JS. Assessment of nitric oxide signals by triiodide chemiluminescence. Proc Natl Acad Sci U S A 104: 2157–2162, 2007.
 117. Hebbel RP, Morgan WT, Eaton JW, Hedlund BE. Accelerated autooxidation and heme loss due to instability of sickle hemoglobin. Proc Natl Acad Sci U S A 85: 237–241, 1988.
 118. Hebert PC, Wells G, Blajchamn MA, Marshall J. A multicenter, randomized, controlled clinical trial of transfusion requirements in critical care. N Engl J Med 340: 409–417, 1999.
 119. Herold S, Exner M, Nauser T. Kinetic and mechanistic studies of the NO‐mediated oxidation of oxymyoglobin and oxyhemoglobin. Biochemistry 40: 3385–3395, 2001.
 120. Herold S, Rock G. Reactions of deoxy‐, oxy‐, and methemoglobin, with nitrogen monoxide. mechanistic studies of the S‐nitrosothiol formation under different mixing conditions. J Biol Chem 278: 6623–6634, 2002.
 121. Herrera M, Garvin JL. Novel role of AQP‐1 in NO‐dependent vasorelaxation. Am J Physiol Renal Physiol 292: F1443–F1451, 2007.
 122. Herrera M, Hong NJ, Garvin JL. Aquaporin‐1 transports NO across cell membranes. Hypertension 48: 157–164, 2006.
 123. Herring N, Paterson DJ. Neuromodulators of peripheral cardiac sympatho‐vagal balance. Exp Physiol 94: 46–53, 2009.
 124. Hess DT, Foster MW, Stamler JS. Assays for S‐nitrosothiols and S‐nitrosylated proteins and mechanistic insights into cardioprotection. Circulation 120: 190–193, 2009.
 125. Hess DT, Matsumoto A, Kim SO, Marshall HE, Stamler JS. Protein S‐nitrosylation: Purview and parameters. Nat Rev Mol Cell Biol 6: 150–166, 2005.
 126. Hickey MJ, Sihota E, Amrani A, Santamaria P, Zbytnuik LD, Ng ES, Ho W, Sharkey KA, Kubes P. Inducible nitric oxide synthase (iNOS) in endotoxemia: Chimeric mice reveal different cellular sources in various tissues. FASEB J 16: 1141–1143, 2002.
 127. Hildebrandt W, Alexander S, Bartsch P, Droge W. Effect of N‐acetyl‐cysteine on the hypoxic ventilatory response and erythropoietin production: Linkage between plasma thiol redox state and O2 chemosensitivity. Blood 99: 1552–1555, 2002.
 128. Hille R, Olson JS, Palmer G. Spectral transitions of nitrosyl hemes during ligand binding to hemoglobin. J Biol Chem 254: 12110–12120, 1979.
 129. Hillier SC, Graham JA, Hanger CC, Godbey PS, Glenny RW, Wagner WW Jr. Hypoxic vasoconstriction in pulmonary arterioles and venules. J Appl Physiol 82: 1084–1090, 1997.
 130. Ho J, Sibbald WJ, Chin‐Yee IH. Effects of storage on efficacy of red cell transfusion: When is it not safe? Crit Care Med 31: S687–S697, 2003.
 131. Hogg N, Singh RJ, Konorev E, Joseph J, Kalyanaraman B. S‐Nitrosoglutathione as a substrate for γ‐glutamyl transpeptidase. Biochem J 323: 477–481, 1997.
 132. Holland RA, Shibata H, Scheid P, Piiper J. Kinetics of O2 uptake and release in a stopped flow apparatus: Effects of the unstirred layer. Respir Physiol 59: 71–91, 1985.
 133. Hrinczenko BW, Schechter AN, Wojtkowski TL, Pannell LK, Cashon RE, Alayash AI. Nitric oxide‐mediated heme oxidation and selective [beta]‐globin nitrosation of hemoglobin from normal and sickle erythrocytes. Biochem Biophys Res Commun 275: 962–967, 2000.
 134. Huang KT, Han TH, Hyduke DR, Vaughn MW, Van Herle H, Hein TW, Zhang C, Kuo L, Liao JC. Modulation of nitric oxide bioavailability by erythrocytes. Proc Natl Acad Sci U S A 98: 11771–11776, 2001.
 135. Huang KT, Huang Z, Kim‐Shapiro DB. Nitric oxide red blood cell membrane permeability at high and low oxygen tension. Nitric Oxide 16: 209–216, 2007.
 136. Huang Z, Shiva S, Kim‐Shapiro DB, Patel RP, Ringwood LA, Irby CE, Huang KT, Ho C, Hogg N, Schechter AN, Gladwin MT. Enzymatic function of hemoglobin as a nitrite reductase that produces NO under allosteric control. J Clin Invest 115: 2099–2107, 2005.
 137. Hudson LD, Milberg JA, Anardi D, Maunder RJ. Clinical risks for development of the acute respiratory distress syndrome. Am J Respir Crit Care Med 151: 293–301, 1995.
 138. Huxley VH, Kutchai H. Effect of diffusion boundary layers on the initial uptake of O2 by red cells. Theory versus experiment. Microvasc Res 26: 89–107, 1983.
 139. Ignarro L. Biosynthesis and metabolism of endothelium‐derived nitric oxide. Annu Rev Pharmacol Toxicol 30: 535–560, 1990.
 140. Ignarro LJ. Nitric Oxide: Biology and Pathobiology. Academic Press, San Diego, CA, 2000.
 141. Ignarro LJ, Buga GM, Wood KS, Byrns RE, Chaudhuri G. Endothelium‐derived relaxing factor produced and released from artery and veins is nitric oxide. Proc Natl Acad Sci U S A 1987: 9265–9269, 1987.
 142. Isbell TS, Koenitzer JR, Crawford JH, White CR, Kraus D, Patel RP. Assessing NO‐dependent vasodilatation using vessel bioassays at defined oxygen tensions. Methods Enzymol 396: 553–568, 2005.
 143. Isbell TS, Sun CW, Wu LC, Teng X, Vitturi DA, Branch BG, Kevil CG, Peng N, Wyss JM, Ambalavanan N, Schwiebert L, Ren J, Pawlik KM, Renfrow MB, Patel RP, Townes TM. SNO‐hemoglobin is not essential for red blood cell‐dependent hypoxic vasodilation. Nat Med 14: 773–777, 2008.
 144. Jacob, C, Knight I, Winyard PG. Aspects of the biological redox chemistry of cysteine: From simple redox responses to sophisticated signalling pathways. Biol Chem 387: 1385–1397, 2006.
 145. Jagger JE, Bateman RM, Ellsworth ML, Ellis CG. Role of erythrocyte in regulating local O2 delivery mediated by hemoglobin oxygenation. Am J Physiol Heart Circ Physiol 280: H2833–H2839, 2001.
 146. James PE, Lang D, Tufnell‐Barret T, Milsom AB, Frenneaux MP. Vasorelaxation by red blood cells and impairment in diabetes: Reduced nitric oxide and oxygen delivery by glycated hemoglobin. Circ Res 94: 976–983, 2004.
 147. James PE, Tufnell‐Barret T, Milsom AB, Frenneaux MP, Lang D. Red blood cell‐mediated hypoxic vasodilatation: A balanced physiological viewpoint. Circ Res 95: e8–e9, 2004.
 148. Jeffers A, Gladwin MT, Kim‐Shapiro DB. Computation of plasma hemoglobin nitric oxide scavenging in hemolytic anemias. Free Radic Biol Med 41: 1557–1565, 2006.
 149. Jia L, Bonaventura C, Bonaventura J, Stamler JS. S‐Nitrosohaemoglobin: A dynamic activity of blood involved in vascular control. Nature 380: 221–226, 1996.
 150. Jiang J, Corbett J, Hogg N, Mason RP. An electron paramagnetic resonance investigation of the oxygen dependence of the arterial‐venous gradient of nitrosyl hemoglobin in blood circulation. Free Radic Biol Med 43: 1208–1215, 2007.
 151. Johnson D, Mayers I. Multiple organ dysfunction syndrome: A narrative review. Can J Anesth 48: 502–509, 2001.
 152. Jones DP, Go YM, Anderson CL, Ziegler TR, Kinkade JM Jr, Kirlin WG. Cysteine/cystine couple is a newly recognized node in the circuitry for biologic redox signaling and control. FASEB J 18: 1246–1248, 2004.
 153. Jourd'heuil D, Gray L, Grisham MB. S‐Nitrosothiol formation in blood of lipopolysaccharide‐treated rats. Biochem Biophys Res Commun 273: 22–26, 2000.
 154. Kantrow SP, Huang YC, Whorton AR, Grayck EN, Knight JM, Millington DS, Piantadosi CA. Hypoxia inhibits nitric oxide synthesis in isolated rabbit lung. Am J Physiol Lung Cell Mol Physiol 272: L1167–L1173, 1997.
 155. Kato M, Staub NC. Response of small pulmonary arteries to unilobar hypoxia and hypercapnia. Circ Res 19: 426–440, 1966.
 156. Kaye DM, Wiviott SD, Kobzik L, Kelly RA, Smith TW. S‐Nitrosothiols inhibit neuronal norepinephrine transport. Am J Physiol 272: H875–H883, 1997.
 157. Ketcham EM, Cairns CB. Hemoglobin based oxygen carriers: Development and clinical potential. Ann Emerg Med 33: 326–337, 1999.
 158. Kilbourn RG, Joly G, Cashon B, DeAngelo J, Bonaventura J. Cell‐free hemoglobin reverses the endotoxin‐mediated hyporesponsivity of rat aortic rings to alpha‐adrenergic agents. Biochem Biophys Res Commun 199: 155–162, 1994.
 159. Kim S, Kong RL, Popel AS, Intaglietta M, Johnson PC. Temporal and spatial variations of cell‐free layer width in arterioles. Am J Physiol Heart Circ Physiol 293: H1526–H1535, 2007.
 160. Kim SF, Huri DA, Snyder SH. Inducible nitric oxide synthase binds, S‐nitrosylates, and activates cyclooxygenase‐2. Science 310: 1966–1970, 2005.
 161. Kim‐Shapiro DB, Schechter AN, Gladwin MT. Unraveling the reactions of nitric oxide, nitrite, and hemoglobin in physiology and therapeutics. Arterioscler Thromb Vasc Biol 26: 697–705, 2006.
 162. Kosaka H. Nitric oxide and hemoglobin interactions in the vasculature. Biochem Biophys Acta 1411: 370–377, 1999.
 163. Kosaka H, Sawai Y, Sakaguchi H, Kumura E, Harada N, Watanabe M, Shiga T. ESR spectral transition by arteriovenous cycle in nitric oxide hemoglobin of cytokine‐treated rats. Am J Physiol Cell Physiol 266: C1400–C1405, 1994.
 164. Kosaka H, Seiyama A. Elevation of oxygen release by nitroglycerin without an increase in blood flow in the hepatic microcirculation. Nat Med 3: 456–459, 1997.
 165. Kosaka H, Watanabe M, Yoshihara H, Harada N, Shiga T. Detection of nitric oxide production on lipopolysaccharide‐treated rats by ESR using carbon monoxide hemoglobin. Biochem Biophys Res Commun 184: 1119–1124, 1992.
 166. Kowaluk EA, Fung HL. Spontaneous liberation of nitric oxide cannot account for in vitro vascular relaxation by S‐nitrosothiols. J Pharmacol Exp Ther 255: 1256–1264, 1990.
 167. Kruszyna R, Kruszyna H, Smith RP, Wilcox DE. Generation of valency hybrids and nitrosylated species of hemoglobin in mice by nitric oxide vasodilators. Toxicol Appl Pharmacol 94: 458–465, 1988.
 168. Kubes P, Payne D, Grisham MB, Jourd'heuil D, Fox‐Robichaud A. Inhaled NO impacts vascular but not extravascular compartments in postischemic peripheral organs. Am J Physiol 277: H676–H682, 1999.
 169. Lancaster JR. Simulation of the diffusion and reaction of endogenously produced nitric oxide. Proc Natl Acad Sci U S A 91: 8137–8141, 1994.
 170. Landry DW, Oliver JA. The pathogenesis of vasodilatory shock. N Engl J Med 345: 588–595, 2001.
 171. Leal‐Noval SR, Jara‐Lopez I, Garcia‐Garmendia JL, Marin‐Niebla A, Herruzo‐Aviles AMD, Camacho‐Larana P, Loscertales J. Influence of erythrocyte concentrate storage time on postsurgical morbidity in cardiac surgery patients. Anesthesiology 98: 815–822, 2003.
 172. Leal‐Noval SR, Munoz‐Gomez M, Arellano‐Orden V, Marin‐Caballos A, Amaya‐villar R, Marin A, Puppo‐Moreno A, Ferrandiz‐millon C, Flores‐cordero JM, Murillo‐Cabezas F. Impact of age of transfused blood on cerebral oxygenation in male patients with severe traumatic brain injury. Crit Care Med 36: 1290–1296, 2008.
 173. Leone AM, Palmer RM, Knowles RG, Francis PL, Ashton DS, Moncada S. Constitutive and inducible nitric oxide synthases incorporate molecular oxygen into both nitric oxide and citrulline. J Biol Chem 266: 23790–23795, 1991.
 174. Li S, Whorton AR. Identification of stereoselective transporters for S‐nitroso‐l‐cysteine: Role of LA1 and LA1 in biological activity of nitrosothiols. J Biol Chem 280: 20102–20110, 2005.
 175. Li S, Whorton AR. Functional characterization of two S‐nitroso‐l‐cysteine transporters, which mediate movement of NO equivalents into vascular cells. Am J Physiol Cell Physiology 292: C1263–C1271, 2007.
 176. Liao JC, Hein TW, Vaughn MW, Huang KT, Kuo L. Intravascular flow decreases erythrocyte consumption of nitric oxide. Proc Natl Acad Sci U S A 96: 8757–8761, 1999.
 177. Lima B, Lam GKW, Xie L, Diesen DL, Villamizar N, Nienaber J, Messina E, Bowles D, Kontos CD, Hare JM, Stamler JS, Rockman HA. Endogenous S‐nitrosothiols protect against myocardial injury. Proc Natl Acad Sci U S A 106: 6297–6302, 2009.
 178. Lipton AJ, Johnson MA, Macdonald T, Lieberman MW, Gozal D, Gaston B. S‐Nitrosothiols signal the ventilatory response to hypoxia. Nature 413: 171–174, 2001.
 179. Lipton SA. Physiology: Nitric oxide and respiration. Nature 413: 118–121, 2001.
 180. Liu L, Hausladen A, Zeng M, Que L, Heitman J, Stamler JS. A metabolic enzyme for S‐nitrosothiol conserved from bacteria to humans. Nature 410: 490–494, 2001.
 181. Liu L, Hausladen A, Zeng M, Que L, Heitman J, Stamler JS, Steverding D. Nitrosative stress: Protection by glutathione‐dependent formaldehyde dehydrogenase. Redox Rep 6: 209–210, 2001.
 182. Liu L, Yan Y, Zeng M, Zhang J, Hanes MA, Ahearn G, McMahon TJ, Dickfeld T, Marshall HE, Que LG, Stamler JS. Essential roles of S‐nitrosothiols in vascular homeostasis and endotoxic shock. Cell 116: 617–628, 2004.
 183. Liu X, Miller MJ, Joshi MS, Sadowska‐Krowicka H, Clark DA, Lancaster JR. Diffusion‐limited reaction of free nitric oxide with erythrocytes. J Biol Chem 273: 18709–18713, 1998.
 184. Luchsinger BP, Rich EN, Gow AJ, Williams EM, Stamler JS, Singel DJ. Routes to S‐nitroso‐hemoglobin formation with heme redox and preferential reactivity in the beta subunits. Proc Natl Acad Sci U S A 100: 461–466, 2003.
 185. Luchsinger BP, Rich EN, Yan Y, Williams EM, Stamler JS, Singel DJ. Assessments of the chemistry and vasodilatory activity of nitrite with hemoglobin under physiologically relevant conditions. J Inorg Biochem 99: 912–921, 2005.
 186. Mamone G, Sannolo N, Malorni A, Ferranti P. In vitro formation of S‐nitrosohemoglobin in red cells by inducible nitric oxide synthase. FEBS Lett 462: 241–245, 1999.
 187. Marik PE, Sibbald WJ. Effect of stored blood transfusion on oxygen delivery in patients with sepsis. JAMA 269: 3024–3029, 1993.
 188. Maritim AC, Sanders RA, Watkins JB. Diabetes, oxidative stress, and antioxidants. J Biochem Mol Toxicol 17: 24–38, 2003.
 189. Martinez‐Ruiz A, Lamas S. Nitrosylation of thiols in vascular homeostasis and disease. Curr Atheroscler Rep 7: 213–218, 2005.
 190. Matsuda T, Bates JN, Lewis SJ, Abboud FM, Chapleau MW. Modulation of baroreceptor activity by nitric oxide and S‐nitrosocysteine. Circ Res 76: 426–433, 1995.
 191. Mayer B, Pfeiffer S, Schrammel. A, Koesling. D, Schmidt K, Brunner F. A new pathway of nitric oxide cyclic GMP signaling involving S‐nitrosoglutathione. J Biol Chem 273: 3264–3270, 1998.
 192. McCann UG, Schiller HJ, Gatto LA, Steinberg JM, Carney DE, Nieman GF. Alveolar mechanics alter hypoxic pulmonary vasoconstriction. Crit Care Med 30: 1315–1321, 2002.
 193. McMahon TJ, Ahearn GS, Moya MP, Gow AJ, Huang YC, Luchsinger BP, Nudelman R, Yan Y, Krichman AD, Bashore TM, Califf RM, Singel DJ, Piantadosi CA, Tapson VF, Stamler JS. A nitric oxide processing defect of red blood cells created by hypoxia: Deficiency of S‐nitrosohemoglobin in pulmonary hypertension. Proc Natl Acad Sci U S A 102: 14801–14806, 2005.
 194. McMahon TJ, Doctor A. Extrapulmonary effects of inhaled nitric oxide: Role of reversible S‐nitrosylation of erythrocytic hemoglobin. Proc Am Thorac Soc 3: 153–160, 2006.
 195. McMahon TJ, Exton SA, Bonaventura J, Singel DJ, Solomon SJ. Functional coupling of oxygen binding and vasoactivity in S‐nitrosohemoglobin. J Biol Chem 275: 16738–16745, 2000.
 196. McMahon TJ, Moon RE, Luschinger BP, Carraway MS, Stone AE, Stolp BW, Gow AJ, Pawloski JR, Watke P, Singel DJ, Piantadosi CA, Stamler JS. Nitric oxide in the human respiratory cycle. Nat Med 8: 711–717, 2002.
 197. McMahon TJ, Stamler JS. Concerted nitric oxide/oxygen delivery by hemoglobin. Methods Enzymol 301: 99–114, 1999.
 198. McMurtry IF, Hookway BW, Roos SD. Red blood cells but not platelets prolong vascular reactivity of isolated rat lungs. Am J Physiol Heart Circ Physiol 234: H186–H191, 1978.
 199. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Crit Care Med 20: 864–874, 1992.
 200. Mendes Ribeiro AC, Brunini TMC, Ellory JC, Mann GE. Abnormalities in l‐arginine transport and nitric oxide biosynthesis in chronic renal and heart failure. Cardiovasc Res 49: 697–712, 2001.
 201. Meyer DJ, Kramer H, Ozer N, Coles B, Ketterer B. Kinetics and equilibria of S‐nitrosothiol‐thiol exchange between glutathione, cysteine, penicillamines and serum albumin. FEBS Lett 345: 177–180, 1994.
 202. Milsom AB, Jones CJ, Goodfellow J, Frenneaux MP, Peters JR, James PE. Abnormal metabolic fate of nitric oxide in type I diabetes. Diabetologia 45(11): 1515–1512, 2002.
 203. Motterlini R, Foresti R, Bassi R, Calabrese V, Clark JE, Green CJ. Endothelial heme oxygenase‐1 induction by hypoxia. Modulation by inducible nitric oxide synthase and S‐nitrosothiols. J Biol Chem 275: 13613–13620, 2000.
 204. Moudgil R, Michelakis ED, Archer SL. Hypoxic pulmonary vasoconstriction. J Appl Physiol 98: 390–403, 2005.
 205. Moya MP, Gow AJ, Califf RM, Goldberg RN, Stamler JS. Inhaled ethyl nitrite gas for persistent pulmonary hypertension of the newborn. Lancet 360: 141–143, 2002.
 206. Moya MP, Gow AJ, McMahon TJ, Toone EJ, Cheifetz IM, Goldberg RN, Stamler JS. S‐Nitrosothiol repletion by an inhaled gas regulates pulmonary function. Proc Natl Acad Sci U S A 98: 5792–5797, 2001.
 207. Murad F. Cyclic guanosine monophosphate as a mediator of vasodilation. J Clin Invest 78: 1–5, 1986.
 208. Myers PR, Minor RL, Guerra R, Bates JN, Harrison DG. Vasorelaxant properties of the endothelium‐derived relaxing factor more closely resemble S‐nitrosocysteine than nitric oxide. Nature 345: 161–163, 1990.
 209. Nagababu E, Ramasamy S, Abernethy DR, Rifkind JM. Active nitric oxide produced in the red cell under hypoxic conditions by deoxyhemoglobin‐mediated nitrite reduction. J Biol Chem 278: 46349–46356, 2003.
 210. Nagababu E, Ramasamy S, Rifkind JM. S‐Nitrosohemoglobin: A mechanism for its formation in conjunction with nitrite reduction by deoxyhemoglobin. Nitric Oxide 15: 20–29, 2006.
 211. Nagatomo S, Nagai M, Tsuneshige A, Yonetani T, Kitagawa T. UV resonance Raman studies of alpha‐nitrosyl hemoglobin derivatives: Relation between the alpha 1‐2 subunit interface interactions and the Fe‐histidine bonding of alpha heme. Biochemistry 38: 9659–9666, 1999.
 212. Nakai S, Sakuma I, Togashi H, Yoshoika M. S‐Nitrosylated polyethylene glycol‐conjugated hemoglobin derivative as a candidate material for oxygen therapeutics. Adv Exp Med Biol 519: 216, 2003.
 213. Nakamura H, Nakamura K, Yoidi J. Redox regulation of cellular activation. Annu Rev Immunol 15: 351–369, 1997.
 214. Nozic‐Grayck E, McMahon T, Huang YCT, Dieterle C, Stamler JS, Piantadosi C. Pulmonary vasoconstriction by serotonin is inhibited by S‐nitrosoglutathione. Am J Physiol 282: L1057–L1065, 2001.
 215. Nozik‐Grayck E, Whalen EJ, Stamler JS, McMahon TJ, Chitano P, Piantadosi CA. S‐Nitrosoglutathione inhibits {alpha}1‐adrenergic receptor‐mediated vasoconstriction and ligand binding in pulmonary artery. Am J Physiol Lung Cell Mol Physiol 290: L136–L143, 2006.
 216. Nwose EU, Jelinek HF, Richards RS, Kerr PG. Erythrocyte oxidative stress in diabetes and its cardiovascular complications. Br J Biomed Sci 64: 35–43, 2007.
 217. Okishio M, Ohikawa S, Ichimori Y, Kondo K. Interaction between endothelium‐derived relaxing factors, S‐nitrosothiols, and endothelin‐1 on Ca2+ mobilization in rat vascular smooth muscle cells. Biochem Biophys Res Commun 183: 849–855, 1992.
 218. Ottesen LH, Harry D, Frost M, Davies S, Khan K, Halliwell B, Moore K. Increased formation of S‐nitrothiols and nitrotyrosine in cirrhotic rats during endotoxemia. Free Radic Biol Med 31: 790–798, 2001.
 219. Owen JA, Bates JN, Lewis SJ. Endogenous nitrosyl factors may inhibit the desensitization of 5‐HT3 receptors on vagal cardiopulmonary afferents. Brain Res 1059: 167–172, 2005.
 220. Paddenberg R, Konig P, Faulhammer P, Goldenberg A, Pfeil U, Kummer W. Hypoxic vasoconstriction of partial muscular intra‐acinar pulmonary arteries in murine precision cut lung slices. Respir Res 7: 93, 2006.
 221. Padron J, Peiro C, Cercas E, Llergo JL, Sanchez‐Ferrer CF. Enhancement of S‐nitrosylation in glycosylated hemoglobin. Biochem Biophys Res Commun 271: 217–221, 2000.
 222. Paiva M, Engel LA. Model analysis of intra‐acinar gas exchange. Respir Physiol 62: 257–272, 1985.
 223. Palmer LA, Doctor A, Chhabra P, Sheram ML, Laubach VE, Karlinsey MZ, Forbes MS, Macdonald T, Gaston B. S‐Nitrosothiols signal hypoxia‐mimetic vascular pathology. J Clin Invest 117: 2592–2601, 2007.
 224. Palmer LA, Doctor A, Gaston B. SNO‐hemoglobin and hypoxic vasodilation. Nat Med 14: 1009, 2008.
 225. Patel RP. Biochemical aspects of the reaction of hemoglobin and NO: Implications for Hb‐based blood substitutes. Free Radic Biol Med 28: 1525, 2000.
 226. Pawloski JR, Hess DT, Stamler JS. Export by red blood cells of nitric oxide bioactivity. Nature 409: 622–626, 2001.
 227. Pawloski JR, Hess DT, Stamler JS. Impaired vasodilation by red blood cells in sickle cell disease. Proc Natl Acad Sci U S A 102: 2531–2536, 2005.
 228. Pawloski JR, Swaminathan RV, Stamler JS. Cell‐free and erythrocytic S‐nitrosohemoglobin inhibits human platelet aggregation. Circulation 97: 263–267, 1998.
 229. Pelosi P, Gattinoni L. Acute respiratory distress syndrome of pulmonary and extrapulmonary origin. Intensive Care Med 27: 457–460, 2001.
 230. Pezacki JP, Ship NJ, Kluger R. Release of nitric oxide from S‐nitrosohemoglobin. Electron transfer as a response to deoxygenation. J Am Chem Soc 123: 4615–4616, 2001.
 231. Purdy FR, Tweeddale MG, Merrick PM. Association of mortality with age of blood transfused in septic ICU patients. Can J Anesth 44: 1256–1261, 1997.
 232. Que LG, Liu L, Yan Y, Whitehead GS, Gavett SH, Schwartz DA, Stamler JS. Protection from experimental asthma by an endogenous bronchodilator. Science 308: 1618–1621, 2005.
 233. Rask‐Madsen C, King GL. Mechanisms of disease: Endothelial dysfunction in insulin resistance and diabetes. Nat Clin Pract Endocrinol Metab 3: 46–56, 2007.
 234. Rassaf T, Bryan NS, Maloney RE, Specian V, Kelm M, Kalyanaraman B, Rodriguez J, Feelisch M. NO adducts in mammalian red blood cells: Too much or too little? Nat Med 9: 481–482, 2003.
 235. Reischl E, Dafre AL, Franco JL, Wilhelm Filho D. Distribution, adaptation and physiological meaning of thiols from vertebrate hemoglobins. Comp Biochem Physiol C Toxicol Pharmacol 146: 22–53, 2007.
 236. Reiter CD, Wang X, Tanus‐Santos JE, Hogg N, Cannon RO, Schecter AN, Gladwin MT. Cell‐free hemoglobin limits nitric oxide bioavailability in sickle‐cell disease. Nat Med 8: 1383–1389, 2002.
 237. Rengasamy A, Johns RA. Determination of Km for oxygen of nitric oxide synthase isoforms. J Pharmacol Exp Ther 276: 30–33, 1996.
 238. Reutov V, Sorokina E. NO‐synthase and nitrite‐reductase components of nitric oxide cycle. Biochemistry (Mosc) 63: 874–884, 1998.
 239. Reynolds JD, Ahearn GS, Angelo M, Zhang J, Cobb F, Stamler JS. S‐Nitrosohemoglobin deficiency: A mechanism for loss of physiological activity in banked blood. Proc Natl Acad Sci U S A 104: 17058–17062, 2007.
 240. Rifkind JM, Ramasamy S, Manoharan PT, Nagababu E, Mohanty JG. Redox reactions of hemoglobin. Antioxid Redox Signal 6: 657–666, 2004.
 241. Rigobello MP, Scutari G, Boscolo R, Bindoli A. Oxidation of adrenaline and its derivatives by S‐nitrosoglutathione. Nitric Oxide 5: 39–46, 2001.
 242. Roccatello D, Mengozzi G, Alfieri V, Pignone E, Menegatti E, Cavalli G, Cesano G, Rossi D, Fromica M, Inconis T, Martina G, Paradisi L, Sena L, Piccoli G. Early increase in blood nitric oxide, detected by electron paramagnetic resonance as nitrosylhaemoglobin, in haemodialysis. Nephrol Dial Transplant 12: 292–297, 1997.
 243. Romeo AA, Capobianco JA, English AM. Superoxide dismutase targets NO from GSNO to Cysbeta93 of oxyhemoglobin in concentrated but not dilute solutions of the protein. J Am Chem Soc 125: 14370–14378, 2003.
 244. Ross JM, Fairchild HM, Weldy J, Guyton AC. Autoregulation of blood flow by oxygen lack. Am J Physiol Legacy 202: 21–24, 1962.
 245. Rother RP, Bell L, Hillmen P, Gladwin MT. The clinical sequelae of intravascular hemolysis and extracellular plasma hemoglobin: A novel mechanism of human disease. JAMA 293: 1653–1662, 2005.
 246. Ryter SW, Morse D, Choi AMK. Carbon monoxide: To boldly go where NO has gone before. Sci STKE 2004: re6, 2004.
 247. Sand AE, Andersson AE, Fried G. Effects of nitric oxide donors and inhibitors of nitric oxide signalling on endothelin‐ and serotonin‐induced contractions in human placental arteries. Acta Physiol Scand 174: 217–223, 2003.
 248. Satoh S, Kimura T, Toda M, Miyazaki H, Ono S, Narita H, Murayama T, Nomura Y. NO donors stimulate noradrenaline release from rat hippocampus in a calmodulin‐dependent manner in the presence of l‐cysteine. J Cell Physiol 169: 87–96, 2003.
 249. Scharfstein JS, Keaney JF, Slivka A, Welch GN, Vita JA, Stamler JS, Loscalzo J. In vivo transfer of nitric oxide between a plasma protein‐bound reservoir and low molecular weight thiols. J Clin Invest 94: 1432–1439, 1994.
 250. Shimazutsu K, Uemura K, Auten K, Baldwin M, Belknap S, La Banca F, Jones M, McClaine D, Eubanks W, Stamler J, Reynolds J. Inclusion of a nitric oxide congener in the insufflation gas repletes S‐nitrosohemoglobin and stabilizes physiologic status during prolonged carbon dioxide pneumoperitoneum. Clin Transl Sci 2: 405–412, 2009.
 251. Shirai M, Sada K, Ninomiya I. Effects of regional alveolar hypoxia and hypercapnia on small pulmonary vessels in cats. J Appl Physiol 61: 440–448, 1986.
 252. Singel DJ, Stamler JS. Blood traffic control. Nature 430: 297–297, 2004.
 253. Singel DJ, Stamler JS. Chemical physiology of blood flow regulation by red blood cells: Role of nitric oxide and S‐nitrosohemoglobin. Annu Rev Physiol 67: 99–145, 2005.
 254. Sommer N, Dietrich A, Schermuly RT, Ghofrani HA, Gudermann T, Schulz R, Seeger W, Grimminger F, Weissmann N. Regulation of hypoxic pulmonary vasoconstriction: Basic mechanisms. Eur Respir J 32: 1639–1651, 2008.
 255. Sonveaux P, Kaz AM, Snyder SA, Richardson RA, Cardenas‐Navia LI, Braun RD, Pawloski JR, Tozer GM, Bonaventura J, McMahon TJ, Stamler JS, Dewhirst MW. Oxygen regulation of tumor perfusion by S‐nitrosohemoglobin reveals a pressor activity of nitric oxide. Circ Res 96: 1119–1126, 2005.
 256. Spencer NY, Zeng H, Patel RP, Hogg N. Reaction of S‐nitrosoglutathione with the heme group of deoxyhemoglobin. J Biol Chem 275: 36562–36567, 2000.
 257. Staab CA, Lander J, Morgenstern R, Grafström RC, Höög JO. The Janus face of alcohol dehydrogenase 3. Chem Biol Interact 178: 29–35, 2009.
 258. Stamler JS. Redox signaling: Nitrosylation and related target interactions of nitric oxide. Cell 78: 931–936, 1994.
 259. Stamler JS. S‐Nitrosothiols and the bioregulatory actions of nitrogen oxides through reactions with thiol groups. Curr Top Microbiol Immunol 196: 19–36, 1995.
 260. Stamler JS. S‐Nitrosothiols in the blood: Roles, amounts, and methods of analysis. Circ Res 94: 414–417, 2004.
 261. Stamler JS, Hausladen A. Oxidative modifications in nitrosative stress. Nat Struct Biol 5: 247–249, 1998.
 262. Stamler JS, Jia L, Eu JP, McMahon TJ, Demchenko IT, Bonaventura J, Gernert K, Piantadosi CA. Blood flow regulation by S‐nitrosohemoglobin in the physiological oxygen gradient. Science 276: 2034–2037, 1997.
 263. Stamler JS, Lamas S, Fang FC. Nitrosylation. The prototypic redox‐based signaling mechanism. Cell 106: 675–683, 2001.
 264. Stamler JS, Meissner G. Physiology of nitric oxide in skeletal muscle. Physiol Rev 81: 209–237, 2001.
 265. Stamler JS, Simon DI, Osborne JA, Mullins ME, Jaraki O, Michel T, Singel DJ, Loscalzo J. S‐Nitrosylation of proteins with nitric oxide: Synthesis and characterization of biologically active compounds. Proc Natl Acad Sci U S A 89: 444–448, 1992.
 266. Stamler JS, Singel DJ, Loscalzo J. Biochemistry of nitric oxide and its redox‐activated forms. Science 258: 1898–1902, 1992.
 267. Stamler JS, Singel D, Piantadosi C. SNO‐hemoglobin and hypoxic vasodilation. Nat Med 14: 1009, 2008.
 268. Stamler JS, Toone EJ, Lipton SA, Sucher NJ. (S)NO signals: Translocation, regulation, and a consensus motif. Neuron 18: 691–696, 1997.
 269. Staub NC. Site of hypoxic pulmonary vasoconstriction. Chest 88: 240S–245S, 1985.
 270. Stephansson O, Dickman PW, Johansson A, Cnattingius S. Maternal hemoglobin concentration during pregnancy and risk of stillbirth. JAMA 284: 2611–2617, 2000.
 271. Strand OA, Leone A, Giercksky KE, Kirkeboen KA. Nitric oxide indices in human septic shock. Crit Care Med 28: 2779–2785, 2000.
 272. Subasinghe W, Spence DM. Simultaneous determination of cell aging and ATP release from erythrocytes and its implications in type 2 diabetes. Anal Chim Acta 618: 227–233, 2008.
 273. Sugerman HJ, Davidson DT, Vibul S, Delivoria‐Papadopoulos M, Miller LD, Oski FA. The basis of defective oxygen delivery from stored blood. Surg Gynecol Obstet 131: 733–741, 1970.
 274. Suzuki K, Naoki K, Kudo H, Nishio K, Sato N, Aoki T, Suzuki Y, Takeshita KEI, Miyata A, Tsumura H, Yamakawa Y, Yamaguchi K. Impaired hypoxic vasoconstriction in intraacinar microvasculature in hyperoxia‐exposed rat lungs. Am J Respir Crit Care Med 158: 602–609, 1998.
 275. Swan A, Hunter P, Tawhai M. Pulmonary gas exchange in anatomically‐based models of the lung. In: Integration in Respiratory Control. 2008, p. 184–189.
 276. Sylvester JT, Mitzner W, Ngeow Y, Permutt S. Hypoxic constriction of alveolar and extra‐alveolar vessels in isolated pig lungs. J Appl Physiol 54: 1660–1666, 1983.
 277. Szabo C. Role of nitrosative stress in the pathogenesis of diabetic vascular dysfunction. Br J Pharmacol 156: 713–727, 2009.
 278. Tabuchi A, Mertens M, Kuppe H, Pries AR, Kuebler WM. Intravital microscopy of the murine pulmonary microcirculation. J Appl Physiol 104: 338–346, 2008.
 279. Tawhai MH, Burrowes KS, Hoffman EA. Computational models of structure‐function relationships in the pulmonary circulation and their validation. Exp Physiol 91: 285–293, 2006.
 280. Tejedor C, Lorente JA, Delgado MA, Fernandez‐Segoviano P, De PM, Tobalina R, Alonso M, Moscoso A, Soto F, Blazquez J, Esteban A. Interaction between hemoglobin and glutathione in the regulation of blood flow in normal and septic pigs. Crit Care Med 30: 2493–2500, 2002.
 281. Tinmouth A, Fergusson D, Yee IC, Hebert PC. Clinical consequences of red cell storage in the critically ill. Transfusion 46: 2014–2027, 2006.
 282. Titheradge MA. Nitric oxide in septic shock. Biochim Biophys Acta 1411: 437–455, 1999.
 283. Tsai AG, Cabrales P, Intaglietta M. Microvascular perfusion upon exchange transfusion with stored red blood cells in normovolemic anemic conditions. Transfusion 44: 1626–1634, 2004.
 284. Tse CM, Fincham DA, Ellory JC, Young JD. Use of membrane vesicles to estimate the numbers of system y+ and system L amino acid transporters in human erythrocytes. Biochem J 277: 565–568, 1991.
 285. Tsoukias NM. Nitric oxide bioavailability in the microcirculation: Insights from mathematical models. Microcirculation 15: 813–834, 2008.
 286. Ullrich R, Bloch KD, Ichinose F, Steudel W, Zapol WM. Hypoxic pulmonary blood flow redistribution and arterial oxygenation in endotoxin‐challenged NOS2‐deficient mice. J Clin Invest 104: 1421–1429, 1999.
 287. Uncles DR, Daugherty MO, Frank DU, Roos CM, Rich GF. Nitric oxide modulation of pulmonary vascular resistance is red blood cell dependent in isolated rat lungs. Anesth Analg 83: 1212–1217, 1996.
 288. Valeri CR, Hirsch NM. Restoration in vivo of erythrocyte adenosine triphosphate 2,3‐diphosphoglycerate potassium ion and sodium ion concentrations following the transfusion of acid‐citrate‐dextrose stored human red blood cells. J Lab Clin Med 73: 722–733, 1969.
 289. Valtis DJ. Defective gas transport function of stored red blood cells. Lancet 266: 119–124, 1954.
 290. Vanhoutte PM. Endothelium‐derived free radicals: For worse and for better. J Clin Invest 107: 23–25, 2001.
 291. Vaughn MW, Huang KT, Kuo L, Liao JC. Erythrocytes possess an intrinsic barrier to nitric oxide consumption. J Biol Chem 275: 2342–2348, 2000.
 292. Vaughn MW, Kuo L, Liao JC. Effective diffusion distance of nitric oxide in the microcirculation. Am J Physiol 274: H1705–H1714, 1998.
 293. Vidwans AS, Uliasz TF, Hewett JA, Hewett SJ. Differential modulation of prostaglandin H synthase‐2 by nitric oxide‐related species in intact cells. Biochemistry 40: 11533–11542, 2001.
 294. Villagra J, Shiva S, Hunter LA, Machado RF, Gladwin MT, Kato GJ. Platelet activation in patients with sickle disease, hemolysis‐associated pulmonary hypertension, and nitric oxide scavenging by cell‐free hemoglobin. Blood 110: 2166–2172, 2007.
 295. Vincent JL, Baron JF, Reinhart K, Gattinoni L, Thijs L, Webb A, Meier‐Hellmann A, Nollet G, Peres‐Bota D; ABC (Anemia and Blood Transfusion in Critical Care) Investigators. Anemia and blood transfusion in critically ill patients. JAMA 288: 1499–1507, 2002.
 296. Volk T, Kox WJ. Endothelium function in sepsis. Inflamm Res 49: 185–198, 2000.
 297. Walder JA, Chatterjee R, Steck TL, Low PS, Musso GF, Kaiser ET, Rogers PH, Arnone A. The interaction of hemoglobin with the cytoplasmic domain of band 3 of the human erythrocyte membrane. J Biol Chem 259: 10238–10246, 1984.
 298. Wang X, Tanus‐Santos JE, Reiter CD, Dejam A, Shiva S, Smith RD, Hogg N, Gladwin MT. Biological activity of nitric oxide in the plasmatic compartment. Proc Natl Acad Sci U S A 101: 11477–11482, 2004.
 299. Ward JP, Aaronson PI. Mechanisms of hypoxic pulmonary vasoconstriction: Can anyone be right? Respir Physiol 115: 261–271, 1999.
 300. Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med 342: 1334–1349, 2000.
 301. Weissmann N, Grimminger F, Olschewski A, Seeger W. Hypoxic pulmonary vasoconstriction. Am J Physiol 281: L314–L317, 2001.
 302. Weissmann N, Grimminger F, Walmrath D, Seeger W. Hypoxic vasoconstriction in buffer‐perfused rabbit lungs. Respir Physiol 100: 159–169, 1995.
 303. Weissmann N, Sommer N, Schermuly RT, Ghofrani HA, Seeger W, Grimminger F. Oxygen sensors in hypoxic pulmonary vasoconstriction. Cardiovasc Res 71: 620–629, 2006.
 304. Wheeler AP, Bernard GR. Treating patients with severs sepsis. N Engl J Med 340: 207–214, 1999.
 305. Wink DA, Cook JA, Kim SY, Vodovotz Y, Pacelli R, Krishna MC, Russo A, Mitchell JB, Jourd'heuil D, Miles AM, Grisham MB. Superoxide modulates the oxidation and nitrosation of thiols by nitric oxide‐derived reactive intermediates. J Biol Chem 272: 11147–11151, 1997.
 306. Winslow RM. Red cell substitutes. Semin Hematol 44: 51–59, 2007.
 307. Wood KC, Granger DN. Sickle cell disease: Role of reactive oxygen and nitrogen metabolites. Clin Exp Pharmacol Physiol 34: 926–932, 2007.
 308. Wu WC, Rathore SS, Wang Y, Radford MJ, Krumholz HM. Blood transfusion in elderly patients with acute myocardial infarction. N Engl J Med 345: 1230–1236, 2001.
 309. Xue Y, Liu Z, Gao X, Jin C, Wen L, Yao X, Ren J. GPS‐SNO: Computational prediction of protein S‐nitrosylation sites with a modified GPS algorithm. PLoS ONE 5: e11290, 2010.
 310. Yamaguchi K, Suzuki K, Naoki K, Nishio K, Sato N, Takeshita K, Kudo H, Aoki T, Suzuki Y, Miyata A, Tsumura H. Response of intra‐acinar pulmonary microvessels to hypoxia, hypercapnic acidosis, and isocapnic acidosis. Circ Res 82: 722–728, 1998.
 311. Yeo TW, Lampah DA, Gitawati R, Tjitra E, Kenangalem E, McNeil YR, Darcy CJ, Granger DL, Weinberg JB, Lopansri BK, Price RN, Duffull SB, Celermajer DS, Anstey NM. Impaired nitric oxide bioavailability and l‐arginine reversible endothelial dysfunction in adults with Falciparum malaria. J Exp Med 204: 2693–2704, 2007.
 312. Yonetani T, Tsuneshige A, Zhou Y, Chen X. Electron paramagnetic resonance and oxygen binding studies of alpha‐nitrosyl hemoglobin. J Biol Chem 273: 20323–20333, 1998.
 313. Zallen G, Offner PJ, Moore EE, Blackwell J, Ciesla DJ, Gabriel J, Denny C, Silliman CC. Age of transfused blood is an independent risk factor for postinjury multiple organ failure. Am J Surg 178: 570–572, 1999.
 314. Zhang Y, Hogg N. The mechanism of transmembrane S‐nitrosothiol transport. Proc Natl Acad Sci U S A 101: 7891–7896, 2004.
 315. Zhao YL, Houk KN. Thionitroxides, RSNHO: The structure of the SNO moiety in “S‐nitrosohemoglobin,” a possible NO reservoir and transporter. J Am Chem Soc 128: 1422–1423, 2006.
 316. Zimmet JM, Hare JM. Nitroso‐redox interactions in the cardiovascular system. Circulation 114: 1531–1544, 2006.
 317. Zingarelli B, Day BJ, Crapo JD, Salzman AL, Szabo C. The potential role of peroxynitrite in the vascular contractile and cellular energetic failure in endotoxic shock. Br J Pharmacol 120: 259–267, 1997.
Further Reading
 1. Singel DJ, Stamler JS. Chemical physiology of blood flow regulation by red blood cells: Role of nitric oxide and S‐nitrosohemoglobin. Annu Rev Physiol 67: 99–145, 2005.
 2. Stamler JS, Singel DJ, Loscalzo J. Biochemistry of nitric oxide and its redox‐activated forms. Science 258: 1898–1902, 1992.
 3. Gaston B, Singel D, Doctor A, Stamler JS. S‐Nitrosothiol signaling in respiratory biology. Am J Respir Crit Care Med 173: 1186–1193, 2006.
 4. Zimmet JM, Hare JM. Nitroso‐redox interactions in the cardiovascular system. Circulation 114: 1531–1544, 2006.
 5. Hess DT, Matsumoto A, Kim SO, Marshall HE, Stamler JS. Protein S‐nitrosylation: Purview and parameters. Nat Rev Mol Cell Biol 6: 150–166, 2005.
 6. Lima B, Forrester MT, Hess DT, Stamler JS. S‐Nitrosylation in cardiovascular signaling. Circ Res. 106: 633–46, 2010.

Further Reading
1.      Singel DJ and Stamler JS. Chemical Physiology of Blood Flow Regulation by Red Blood Cells: Role of Nitric Oxide and S-Nitrosohemoglobin. Annual Review of Physiology 67: 99-145,2005.
2.      Stamler JS, Singel DJ and Loscalzo J. Biochemistry of nitric oxide and its redox-activated forms. Science 258: 1898-1902, 1992.
3.      Gaston B, Singel D, Doctor A and Stamler JS. S-Nitrosothiol Signaling in Respiratory Biology. Am J Respir Crit Care Med 173: 1186-1193,2006.
4.      Zimmet JM and Hare JM. Nitroso-Redox Interactions in the Cardiovascular System. Circulation 114: 1531-1544, 2006.
5.      Hess DT, Matsumoto A, Kim SO, Marshall HE and Stamler JS. Protein S-Nitrosylation: Purview and Parameters. Nat Rev Mol Cell Biol 6:150-166,2005.
6.      Lima B, Forrester MT, Hess DT, Stamler JS. S-nitrosylation in cardiovascular signaling. Circ Res. 106:633-46, 2010.


Related Articles:

Blood Oxygen Transport
Spatial Distribution of Ventilation and Perfusion: Mechanisms and Regulation
Interaction of Cardiovascular Reflexes in Circulatory Control
The Pulmonary Microcirculation

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Allan Doctor, Jonathan S. Stamler. Nitric Oxide Transport in Blood: A Third Gas in the Respiratory Cycle. Compr Physiol 2011, 1: 541-568. doi: 10.1002/cphy.c090009