A REMARKABLY exciting field of research has developed since nitric oxide (NO) was identified in 1987 as a key endothelium-derived relaxing factor (EDRF). 1,2The awarding of the 1998 Nobel prize in physiology or medicine to three seminal researchers in the field of NO biology provided the most recent evidence for the emerging prominence of this area of study. 3The understanding of the roles of NO in the cardiovascular, immune, and nervous systems; the isolation and localization of NO synthases (NOS); the manipulation of the genes for NOS, including their cloning and selective transfer or knock-out; and the therapeutic use of inhaled NO gas have revolutionized many fields of physiologic research and are influencing clinical therapy.

Many insights into the mechanisms of action of NO have been gained. Since the reported applications of inhaled NO in the laboratory 4and in adult patients with primary pulmonary hypertension in 1991, 5hundreds of studies have been conducted to determine the clinical applicability of inhaled NO. In subgroups of severely ill and hypoxic children and adults, inhaled NO improves arterial oxygenation and selectively decreases pulmonary arterial hypertension (PAH). NO inhalation therapy, in combination with conventional 6,7or high-frequency oscillatory ventilation, 8can reduce the need for extracorporeal membrane oxygenation (ECMO), an expensive and invasive procedure in newborn patients with hypoxic respiratory failure. 6,7However, it remains uncertain whether NO inhalation improves survival rates in adults or children with severe lung injury.

New applications for NO inhalation have been discovered. Recent studies indicate that inhaled NO may decrease intestinal ischemia–reperfusion injury 9and may be useful to treat thrombotic disorders. 10,11By increasing the oxygen affinity of sickle cell hemoglobin, 12inhaled NO may prevent or treat sickle cell crisis. This article reviews the relevant physiologic effects, therapeutic uses, side-effects, and toxicity of NO inhalation. The first portion of this article concentrates on the chemistry, biochemistry, toxicology, and biology of NO; the second portion summarizes the results of NO inhalation studies to date in experimental settings and the results of clinical studies in newborns, children, and adults.

Nitric oxide is a colorless, almost odorless gas that is slightly soluble in water (2 or 3 mM). 13Environmental NO arises from combustion processes (e.g. , fossil fuel combustion and tobacco smoke) and lightning. 14Atmospheric concentrations of NO usually range between 10 and 500 parts per billion (ppb), but can exceed 1.5 parts per million (ppm) in areas of heavy traffic. 15Concentrations of NO produced in the hot cone of a glowing cigarette can reach 1,000 ppm in a 40-ml puff. 16The Occupational Safety and Health Administration has set 8-h time-weighted average exposure limits in the workplace at 25 ppm for NO breathing and at 5 ppm for nitrogen dioxide (NO2). 17Commercially, NO is manufactured from the reaction of sulfur dioxide with nitric acid, from the reaction of sodium nitrite and sulfuric acid, or by the oxidation of ammonia over a platinum catalyst at a high temperature (> 500°C). 18In an anaerobic environment (i.e. , in highly purified nitrogen), NO can be stored for several years.

Reaction of Nitric Oxide with Oxygen 

In the gaseous phase, NO reacts with molecular oxygen to form NO2. The conversion rate of NO to NO2can be described by the relation

formula

where k  is the rate constant for conversion of NO to NO2. The rate constant has been reported to be between 0.79 × 10−9to 2.26 × 10−9· ppm−2· min−1, dependent on experimental conditions. 19Approximately half of a 10,000-ppm NO mixture in air is converted into NO2within 24 s, whereas 50% of a 10-ppm NO mixture in air is converted into NO2within 7 h at 20°C. 21In aqueous solution, NO2decomposes to give equal amounts of nitrite (NO2) and nitrate (NO3). 13 

The pathologic effects of NO2inhalation have been studied in various animal species. High levels of inhaled NO2(> 10 ppm) induce pulmonary edema, alveolar hemorrhage, changes in the surface tension activities of surfactant, hyperplasia of type 2 alveolar epithelial cells, intrapulmonary accumulation of fibrin, neutrophils, and macrophages, and death. 22,23Lower inhaled NO2concentrations (< 2 ppm) can alter surfactant function, produce alveolar cell hyperplasia, and alter the epithelium of the terminal bronchioles. 24Inhalation of 2 ppm NO2in humans increases alveolar permeability 25and airway reactivity. 26–28Inhalation of 0.5–1.5 ppm NO2for 9 weeks caused focal degeneration of pulmonary interstitial cells, with mild emphysematous changes, in rats. 29 

Reaction of Nitric Oxide with Superoxide 

Nitric oxide and superoxide (O2) readily react to form peroxynitrite (OONO) at nearly a diffusion-limited rate. 13During physiologic conditions, O2is scavenged by endogenous O2scavengers (e.g. , superoxide dismutase) and formation of OONO is minimal. During pathologic conditions, such as in the presence of increased concentrations of O2or after O2scavengers are exhausted, significant concentrations of OONO may be produced. 30Peroxynitrite directly causes oxidation, peroxidation, and nitration of biologically important molecules (e.g. , lipids, proteins, DNA; for review articles see Szabo et al.  31,32). The cytotoxic effects of OONO provide protective functions if they are directed by inflammatory cells against invading microorganisms or tumor cells.

An important example of a reaction caused by OONO is the nitration of tyrosine. Tyrosine nitration inhibits tyrosine phosphorylation, alters the dynamics of assembly and disassembly of cytoskeletal proteins, and inhibits tyrosine hydroxylase, thereby reducing dopamine production by neurons and inhibiting cytoskeletal movements of endothelial cells. 31Nitrotyrosine has been detected in lung tissue sections from patients with lung injury, 33,34in atherosclerotic lesions, 35,36and in lungs after ischemia–reperfusion injury. 37 

Exposure of surfactant to high concentrations of OONO in vitro  reduced its minimum surface tension. 38Peroxynitrite exposure impaired pulmonary surfactant function, because of peroxidation of surfactant lipids, and decreased the ability of the major hydrophilic surfactant, protein A, to aggregate lipids and act synergistically with other surfactant proteins to reduce the minimum surface tension. 39,40These changes of surfactant protein A were associated with nitrotyrosine formation. 39A mixture of surfactant proteins B and C exposed to OONO was incapable of reducing phospholipid minimum surface tension during dynamic compression. 41 

Peroxynitrite can cause cell apoptosis by DNA strand breakage, activation of poly-adenosine-diphosphate-ribosyltransferase and by inhibition of mitochondrial respiratory enzymes. 31,32Peroxynitrite rapidly reacts with carbon dioxide to form an adduct that participates in nitration and oxidation reactions. 42Interestingly, in a model of thrombin or hydrogen peroxide (H2O2)–induced vascular injury of the rat mesenteric endothelium and in an ischemia–reperfusion model of the rat heart, infusion of OONO significantly reduced neutrophil adhesion to the endothelium and expression of adhesion molecules, suggesting that OONO exerts inhibitory effects on neutrophil adhesion in inflammatory processes. 43 

In summary, OONO is more cytotoxic than NO in a variety of experimental systems, 32and the balance of NO, O2, and O2OONO scavenging systems determines whether biologically relevant OONO concentrations will occur in tissues. 30 

Reaction of Nitric Oxide with Heme Proteins and Metals 

Nitric oxide binds to intracellular iron and heme-containing proteins. Examples of heme proteins that are directly affected by NO are oxyhemoglobin, soluble guanylate cyclase (sGC), cyclooxygenase, and cytochrome p450. Guanylate cyclase is stimulated by NO; cyclooxygenase is stimulated by low NO concentrations 44and inhibited by high NO concentrations. 45The cytochrome p450 system is inhibited by NO. 46,47The ratio of rates of uptake and release of NO for ferrous (Fe2+) hemoglobin is 105–106times larger than for oxygen. 48Different from other iron–heme ligands, such as carbon monoxide or oxygen, NO can bind with the ferric (Fe3+) and Fe2+oxidation state of hemoglobin. The vasodilating effects of NO in vivo  are limited by its rapid reaction with oxyhemoglobin or oxymyoglobin to form nitrosylhemoglobin or nitrosylmyoglobin. Methemoglobin (Fe3+hemoglobin) is produced when the heme iron is oxidized from Fe2+to Fe3+and NO3is released. 49Most of the methemoglobin is reduced back to Fe2+hemoglobin by NADH-cytochrome b5/cytochrome b5methemoglobin reductase within erythrocytes. In addition, reduced glutathione reduces methemoglobin. 50 

Reaction of Nitric Oxide with Thiols 

Nitric oxide can nitrosate thiol groups to form S-nitrosothiols. Common, naturally occurring S-nitrosothiols include S-nitrosocysteine, S-nitrosohomocysteine, and S-nitrosoglutathione. 51S-nitrosothiols have similar platelet-inhibitory and vasorelaxant activities to NO, which are mediated through guanylate cyclase activation, 52but which differ in other important physiologic characteristics from gaseous NO. 53–55 

In addition to binding to the iron–heme center of hemoglobin, NO participates in transnitrosation reactions with the sulfhydryl group of hemoglobin to form S-nitrosohemoglobin. Such reactions may serve as important steps in the uptake and distribution of NO in the systemic circulation. In vivo  analysis reveals that arterial blood samples from normal rats contained larger concentrations of S-nitrosohemoglobin than did venous samples, 56suggesting that S-nitrosylation is regulated by hemoglobin oxygenation and changes with erythrocyte transit through the lungs. Stamler et al.  57demonstrated that hemoglobin cysteines (Cysβ93) participate in the binding and release of NO. When deoxygenated hemoglobin with a high oxygen affinity enters the pulmonary circulation, the affinity of the hemoglobin thiol groups for NO is high and NO uptake occurs. In the peripheral circulation, where oxygenated hemoglobin with a low oxygen affinity releases oxygen to tissues at a low partial pressure of oxygen (PO2), release of NO is enhanced. Such localized release of NO permits vasodilation and increased oxygen delivery to occur in tissues with reduced PO2. 57 

Effect of Nitric Oxide on DNA 

Nitric oxide can alter DNA by the formation of mutagenic nitrosamines, 58,59by direct modification and strand breakage of DNA from the formation of radical nitrogen oxide species (e.g. , OONO), 60–62and by inhibition of enzyme systems that are necessary to repair DNA lesions. 63–65NO deaminates desoxynucleosides and desoxynucleotides in mammalian cell preparations and in aerobic solutions of nucleic acids 66and causes dose-dependent DNA strand breakage. 67In contrast, NO can abate DNA damage caused by xanthine oxidase and H2O2. 68–70 

Tumoricidal and tumor-promoting effects of NO have been reported. 65Nitric oxide derived from the inducible NOS (iNOS) of macrophages, Kupffer cells, natural killer cells, and endothelial cells produces tumoricidal effects against many types of tumors, 71–79reduces the viability of several tumor cell lines, 80and inhibits angiogenesis, tumor growth, and metastasis. 81Transfection of iNOS into metastatic melanoma cells reduces their potential for metastasis. 82NO inhibits tumor cell adhesion 83and decreases the metastatic activity of colon cancer cells. 84In other studies, NO mediates tumor growth through NO-mediated control of angiogenesis and of growth factors. 65,85,86Wink et al.  65recently concluded that the role of NO in carcinogenesis is multidimensional. Tissues that are exposed for prolonged durations to high NO concentrations in combination with long-term inflammation and production of reactive oxygen species may accumulate mutations caused by the direct or indirect effects of NO. As a tumor develops, NO produced from iNOS can kill tumor cells through cytostatic and cytotoxic activity. As the tumor progresses, NO may inhibit or support angiogenesis, may limit leukocyte infiltration, and may limit metastasis or kill tumor cells through the induction of apoptosis. 65 

Effect of Nitric Oxide on Lipids 

Nitric oxide has contrasting effects on lipids, particularly on the oxidation of low-density lipoproteins in the pathogenesis of atherosclerotic lesions (for review see Rubbo et al.  87). NO inhibits lipid peroxidation by inhibiting radical chain propagation reactions via  radical–radical reaction with lipid peroxyl and alkoxyl groups. 87,88As a ligand to iron (and other transition metals), NO modulates the prooxidant effects of iron and thereby limits the formation of hydroxyl radicals and iron-dependent electron-transfer reactions. 87NO inhibits cell and OONO-mediated lipoprotein oxidation in macrophage and endothelial cell systems. 89However, NO-induced OONO formation can oxidize low-density lipoproteins to potentially atherogenic species. 90,91The antioxidant versus  prooxidant outcome of these reactions appears to depend on the relative concentration of the various reactive molecules. 88,92 

Endogenous Nitric Oxide Synthesis 

Nitric oxide synthase catalyzes a partially tetrahydrobiopterin-dependent five-electron oxidation of the terminal guanidino nitrogen of L-arginine. 93The reaction stoichiometrically consumes oxygen and nicotinamide adenine dinucelotide hydrogen phosphate (NADPH), requires the cofactors flavin adenine dinucleotide, flavin mononucleotide, and calmodulin, and produces L-citrulline and NO. NOS does not produce detectable levels of NO unless superoxide dismutase is present. 94During conditions of L-arginine depletion, NOS generates O2. 95,96NOS is homologous to the cytochrome p450 reductase enzyme containing iron–protoporphyrin IX. 97,98 

Three NOS isoforms have been identified and classified based on the tissue in which they were first identified, the regulation of their activity, and their substrate–inhibitor profile. Constitutive neuronal NOS (nNOS, NOS1) initially was discovered in nerve tissue. Inducible NOS (iNOS, NOS2), a cytokine-inducible isoform, is expressed in a variety of inflammatory cells. Constitutive endothelial NOS (eNOS, NOS3) was originally described in vascular endothelial cells. More recent studies have shown that expression of the constitutive NOS isoforms (nNOS and eNOS) is regulated, 99–102and that the inducible isoform (iNOS2) is constitutively present without previous stimulation. 103NOS isoforms are expressed in many different cell types and intracellular organelles, and most cells are able to synthesize NO. 104,105Altered NOS expression and endogenous NO synthesis have been reported in a large variety of ischemic, traumatic, neoplastic, inflammatory, and infectious diseases. 106–115In addition to enzymatic generation of NO by NOS isoforms, nonenzymatic formation of NO in vivo  during reduced and acidotic conditions (e.g. , organ ischemia) has been reported 116and can contribute to NO production during pathologic conditions.

Neuronal NOS. 

Neuronal NOS fulfills a myriad of disparate functions in a wide variety of tissues. In the peripheral nervous system, NO acts as a neurotransmitter, regulating smooth muscle relaxation in the gastrointestinal, urogenital, and respiratory tracts via  nonadrenergic noncholinergic nerves containing nNOS. 114Neuronal NOS expression is also present in vasodilator nerves that innervate large cerebral vessels. 117In the central nervous system, NO is essential in neuronal plasticity to modulate information storage in the brain 118and has effects on brain development, memory function, behavior, and pain perception. 114In human skeletal muscle, nNOS modulates contractile force, myocyte development, myofiber differentiation, and myotube innervation. 114Other nNOS expression sites include cardiac nerve terminals that regulate the release of catecholamines in the heart, 119and the retina, where nNOS is involved in NO production in photoreceptors and bipolar cells. 120 

Inducible NOS. 

Inflammatory cells (e.g. , macrophages and granulocytes), among many other cell types, express iNOS in response to a variety of infectious and inflammatory stimuli. Inducible NOS produces effects that are beneficial and critical for survival during important bacterial and parasitic infections (e.g. , Mycobacterium tuberculosis, Toxoplasma gondii ) and in the response to inflammation (e.g. , decrease of neutrophil adhesion in endotoxemia, increase of wound closure, and neovascularization of wounds), as shown in murine models of congenital iNOS deficiency. 115In contrast, increased iNOS expression has been associated with the worsening of other infectious diseases (e.g. , influenza pneumonitis) and inflammatory states (e.g. , endotoxin-induced hypotension, autoimmune vasculitis). 115Enhanced expression of iNOS and increased vascular NO synthesis and release have been associated with systemic arterial vasodilation and the “low-tone state” in sepsis. 121 

Endothelial NOS. 

The 1998 Nobel prize in physiology or medicine was awarded to three researchers who discovered endothelium-derived relaxing factor and demonstrated that NO, generated from eNOS in vascular endothelial cells, is endothelium-derived relaxing factor. 1,2,122Endothelial NOS activity is increased by acetylcholine, bradykinin, and other mediators that increase intracellular calcium concentrations. 122Endothelial NOS activity modulates systemic 122and pulmonary vascular tone 123and plays important roles in lung development and disease. Endothelial NOS expression in the fetal lung changes with lung maturation. 124,125NO production and eNOS expression by endothelial cells is increased by vascular shear stress. 126,127For example, pulmonary vascular eNOS expression is reduced in patients with chronic pulmonary hypertension. 128Congenital absence of eNOS in mice results in pulmonary hypertension and increased right ventricular remodeling if the mouse is stressed by long-term hypoxic breathing. 129Endothelial NOS can be reversibly inhibited by NO 130and eNOS expression can be upregulated by cyclic guanosine monophosphate (cGMP). 102In cardiac endothelial cells, eNOS activity inhibits contractile tone and proliferation of the underlying vascular smooth muscle cells, reduces platelet aggregation and monocyte adhesion, promotes diastolic relaxation, and decreases the oxygen consumption rate of cardiac muscle. 119Endothelial NOS is constitutively expressed in cardiac myocytes, where its activity opposes the inotropic action of catecholamines after muscarinic cholinergic or β-adrenergic receptor stimulation. 119 

Smooth Muscle Relaxation by Inhaled NO 

Soluble guanylate cyclase (sGC) mediates many of the biologic effects of NO and is responsible for conversion of guanosine-5′-triphosphate to cGMP (fig. 1). Cyclic 3′:5′ GMP is an important second messenger in a variety of cell types and is found in the cytosol of almost all mammalian cells. A variety of nitrovasodilators (e.g. , nitroglycerin, sodium nitroprusside) stimulate cGMP synthesis, which, in turn, is responsible for the smooth muscle relaxation mediated by these drugs. 131The common mechanism of action of these drugs is attributed to the release of NO. 132 

Fig. 1. Nitric oxide (NO)–cyclic guanosine monophosphate (cGMP) signal transduction pathway. NO, formed by endothelial cells (  left ) or administered by inhalation (  right ), diffuses to vascular smooth muscle cells (  lower ). NO activates soluble guanylate cyclase, which in turn catalyzes the production of cGMP. Through cGMP-dependent protein kinase mediated effects, increased intracellular concentration of cGMP relaxes smooth muscle and inhibits leukocyte adhesion, platelet adhesion, and cellular proliferation. The action of cGMP is limited by phosphodiesterases, which convert cGMP to GMP. 

Fig. 1. Nitric oxide (NO)–cyclic guanosine monophosphate (cGMP) signal transduction pathway. NO, formed by endothelial cells (  left ) or administered by inhalation (  right ), diffuses to vascular smooth muscle cells (  lower ). NO activates soluble guanylate cyclase, which in turn catalyzes the production of cGMP. Through cGMP-dependent protein kinase mediated effects, increased intracellular concentration of cGMP relaxes smooth muscle and inhibits leukocyte adhesion, platelet adhesion, and cellular proliferation. The action of cGMP is limited by phosphodiesterases, which convert cGMP to GMP. 

Close modal

Soluble guanylate cyclase is a heme-containing protein composed of an α and β subunit. The heme moiety in sGC is essential for activation of the enzyme. The presence of heme results in a 100-fold increase of enzyme activity after stimulation with NO, whereas basal enzyme activity is low without heme and does not change with the addition of NO. 133Effects of cGMP on vascular tone, cardiac function, and intestinal water and ion transport by protein kinase-dependent and -independent mechanisms have been reviewed in detail by others. 134,135 

The physiologic action of cGMP is limited by its hydrolysis to GMP by a family of cyclic nucleotide phosphodiesterases. Of the seven known phosphodiesterase isozymes, phosphodiesterases 1 and 5 hydrolyze cGMP. Phosphodiesterase 1 catalyzes cyclic adenosine monophosphate and cGMP hydrolysis and is found in high concentrations in the brain, the heart, the lung, and the testis. Phosphodiesterase 5 is cGMP specific and has been found in lung tissue, platelets, vascular smooth muscle, and the kidney. It has a high affinity for cGMP and can be inhibited by the selective phosphodiesterase 5 inhibitors zaprinast, sildenafil, and dipyridamole. Inhibition of phosphodiesterase 5 enhances endothelium-dependent vasorelaxation, reduces pulmonary vascular tone, and enhances the hypotensive effects of nitrovasodilators. 136,137 

Selective Pulmonary Vasodilation 

Alveolar Hypoxia. 

The ability of NO to selectively dilate the pulmonary vasculature was evaluated in an awake lamb model of alveolar hypoxia. Alveolar hypoxia produces reversible pulmonary vasoconstriction mediated by an unknown mechanism. During normoxia, inhalation of 80 ppm NO did not alter the normally low mean pulmonary artery pressure (MPAP) and pulmonary vascular resistance (PVR). With alveolar hypoxia (inspired fraction of oxygen [FIO2], 0.1), pulmonary vasoconstriction increased MPAP from 17 mmHg to 28 mmHg. With NO inhalation (40 ppm), MPAP decreased to 20 mmHg and further to 18 mmHg (80 ppm NO) within 3 min of NO breathing. 4Cardiac output and systemic arterial pressure were not affected by NO inhalation. These results were confirmed in mechanically ventilated sheep, 138in awake healthy volunteers breathing low oxygen concentrations at ambient pressure 139(fig. 2), and in volunteers at high altitude (at 4,559 m, hypobaric hypoxia). 140 

Fig. 2. Physiologic effects of inhaled NO (40 ppm) during hypoxia in nine healthy volunteers. Note the decrease of mean pulmonary artery pressure and pulmonary vascular resistance achieved by NO inhalation. Values are mean ± SE. LVSWI = left ventricular stroke work index; PaO2= arterial partial pressure of oxygen, PaCO2= arterial partial pressure of carbon dioxide; PCWP = pulmonary capillary wedge pressure; PPA = mean pulmonary artery pressure; PVR = pulmonary vascular resistance; RVSWI = right ventricular stroke work index. Reprinted with permission from Frostell  et al. 139 

Fig. 2. Physiologic effects of inhaled NO (40 ppm) during hypoxia in nine healthy volunteers. Note the decrease of mean pulmonary artery pressure and pulmonary vascular resistance achieved by NO inhalation. Values are mean ± SE. LVSWI = left ventricular stroke work index; PaO2= arterial partial pressure of oxygen, PaCO2= arterial partial pressure of carbon dioxide; PCWP = pulmonary capillary wedge pressure; PPA = mean pulmonary artery pressure; PVR = pulmonary vascular resistance; RVSWI = right ventricular stroke work index. Reprinted with permission from Frostell  et al. 139 

Close modal

Pulmonary Selectivity and Vascular Sites of Vasodilation 

The pulmonary selectivity of inhaled NO and its rapid inactivation by hemoglobin was first evaluated in an isolated perfused rabbit lung model. The effluent of the perfused lung was conducted to an isolated, pharmacologically preconstricted segment of aorta. The pulmonary vasculature was then preconstricted with U46619, a thromboxane analog. When the perfusate was a hemoglobin-free aqueous buffer, inhalation of NO first decreased MPAP and subsequently the tone of the sequentially perfused aorta. When erythrocytes were added to the perfusate, inhaled NO still caused pulmonary vasodilation, but its vasodilatory effect on the effluent-perfused aorta was abolished, 141suggesting that inhaled NO was inactivated by contact with hemoglobin. The pulmonary vascular selectivity of NO inhalation (i.e. , pulmonary vasodilation in the absence of systemic arterial vasodilation) has been confirmed in numerous subsequent studies. 142–147 

The longitudinal effects of inhaled NO within the pulmonary vasculature are important because increases of arterial or venous vascular tone differentially influence hydrostatic fluid exchange within the lung. Pulmonary venoconstriction increases pulmonary capillary pressure and promotes edema formation. 148,149If venous and arteriolar constriction both contribute to increased PVR, a vasodilator selectively acting on arterial tone may worsen edema formation by increasing mean pulmonary vascular surface area and pressure. Lindeborg et al.  150reported that inhalation of 5, 20, and 80 ppm NO decreased arterial, microvascular, and venous resistances to the same extent in an isolated rabbit lung model. Shirai et al.  151used an X-ray television system to visualize the in vivo  effects of NO inhalation on the internal diameter of pulmonary arteries and veins in a feline model. Inhaling 5–40 ppm NO caused a dose-dependent increase of the diameter of small arteries and veins during normoxic conditions. After induction of hypoxic vasoconstriction by lobar anoxia, NO inhalation dilated smaller constricted and larger nonconstricted arteries, as well as veins. These results suggest that the pulmonary vasodilator response to inhaled NO is similar in pulmonary arteries and veins.

Selective Vasodilation of Ventilated Areas 

The intrapulmonary distribution of blood flow and ventilation (ventilation–perfusion [V̇/Q̇] distribution) is a major determinant of transpulmonary oxygenation effectiveness, and the resulting partial pressure of oxygen in arterial blood (PaO2). In a normal, healthy lung, ventilated areas are well perfused. The shunt from the right to the left side of the circulation is mainly extrapulmonary (e.g. , bronchial veins) and is less than 5–8% of cardiac output. 152Local alveolar hypoxia constricts the vascular bed adjacent to hypoxic regions and redistributes blood flow to lung regions with better ventilation and a higher intraalveolar PO2. It has been proposed that inhaled NO amplifies this mechanism by increasing blood flow through well-ventilated lung areas. Pison et al.  138studied the effects of inhaled NO on distribution in an ovine model of acute hypoxia. Because they studied generalized alveolar hypoxia, no improvement of PaO2during NO inhalation was expected. However, they demonstrated increased blood flow to better ventilated (but still hypoxic) lung areas and a stable PaO2during NO inhalation.

The effects of NO inhalation on gas exchange have been assessed using lung injury models that induce mismatch. The mismatch induced by oleic acid injury in sheep was significantly improved by inhalation of 40 ppm NO 153and was augmented by the simultaneous use of continuous positive airway pressure to open collapsed alveoli. 154Hopkins et al.  155studied the effects of inhaled NO on gas exchange in dogs by selectively creating areas of shunt or areas with a low ratio. NO (80 ppm) decreased blood flow to shunting regions. In areas with a mismatch, NO produced an inconsistent response. When the PVR of the partially obstructed airway regions was decreased by NO inhalation, inequality was increased because blood flow to the relatively poorly ventilated areas was increased by vasodilation. When NO did not reach the lung regions distal to the partial obstruction, and thus did not reduce local PVR, matching was improved.

Bronchodilator Action 

Nitric oxide synthase inhibitors suppress the bronchodilator actions of nonadrenergic noncholinergic–mediated bronchodilation, suggesting that endogenous NO synthesis is involved in the control of bronchial tone. 156,157The expression of various NOS isoforms in peripheral nonadrenergic noncholinergic nerve endings 158and in human bronchial epithelium 159supports this finding. Dupuy et al.  160demonstrated that inhaled NO decreased airway resistance after bronchoconstriction with methacholine in guinea pigs, later confirmed in various experimental models using rabbits, 161,162dogs, 163–165and pigs. 166,167In human volunteers, however, inhaled NO only reduced airway resistance minimally after a methacholine challenge. 168Large airways appeared to be preferentially dilated by inhaled NO. 169 

Pulmonary Surfactant 

Surfactant synthesized by type 2 alveolar epithelial cells affects lung mechanics by reducing surface tension, modifies pulmonary gas exchange, and has antimicrobial functions. 40Isolated type 2 alveolar epithelial cells exposed to NO (generated by the NO donor drugs S-nitroso-N-penicillamine, spermine NONOate, or 3-morpholino-sydnonimine) in the presence of superoxide dismutase reduced their surfactant synthesis by approximately 60%. 170Exposure of surfactant to NO ex vivo  was not associated with changes of surface activity. 38,In vivo , a combination of high inspired oxygen concentrations and high inspired NO concentrations (100 ppm) inhaled by newborn piglets for 48 h significantly decreased the minimum surface tension of surfactant recovered by bronchoalveolar lavage. 38 

In lambs, high inhaled NO concentrations (80–200 ppm) resulted in abnormal surface activities and inhibition of surfactant protein A lipid aggregation. 171In addition, NO (from NO donor drugs) can decrease surfactant protein A gene expression by distal respiratory epithelial cells. 172In contrast, in an experimental model of acute lung injury, combining exogenous surfactant therapy with inhaled NO improved ventilation–perfusion matching and arterial oxygenation. 173Other interactions of NO and surfactant were recently summarized by Hallman and Bry. 40 

Metabolic Fate of Inhaled Nitric Oxide 

Nitric oxide is inactivated by reaction with its biologic target molecules. The half-life of NO in vivo  is only a few seconds. The main metabolic pathways are the binding of NO to O2and to the heme iron of hemoglobin with the subsequent release of NO3. The binding and release of NO to thiols presents another important metabolic pathway. Approximately 90% of NO is absorbed during a steady state inhalation. Almost 70% of the inhaled gas appears within 48 h as NO3in the urine. 174The remaining 30% of inhaled NO is recovered as NO2in the oral cavity through secretion from salivary glands. NO2is also partly converted to nitrogen gas in the stomach and some NO2in the intestine is reduced to ammonia, reabsorbed, and converted to urea. 175,176 

Models of Persistent Pulmonary Hypertension of the Newborn and Respiratory Distress Syndrome 

Hypoxia in the preterm and term newborn is usually characterized by severe PAH, extrapulmonary right-to-left shunting, hypoxemia, and acidosis. Roberts et al.  177studied the effects of inhaled NO in hypoxic and acidotic term newborn lambs delivered by cesarean section. Hypoxia associated with hypercapnia doubled PVR. In this model, inhaling 20 ppm NO during hypoxia completely abolished pulmonary vasoconstriction, despite the presence of a marked respiratory acidosis. Similar results were obtained in hypoxic, mechanically ventilated late-gestation ovine fetuses. 178In an experimental model of persistent pulmonary hypertension of the newborn (PPHN) in lambs (induced by ductal ligation), inhaled NO decreased PVR and markedly increased survival rates. 179,180 

The responsiveness of the premature lung to inhaled NO depends on gestational age and the maturity of the pulmonary vasculature. In the immature lung of the ovine fetus at 0.78 of term, initiation of mechanical ventilation caused maximal pulmonary vasodilation and the addition of NO (20 ppm) or 100% oxygen did not increase vasodilation further. If surfactant was administered and mechanical ventilation at 0.78 term continued with 100% oxygen for 2 h, PVR continued to increase and the initial beneficial effect of mechanical ventilation on PVR and oxygenation decreased. With initiation of NO inhalation 2 h after commencing mechanical ventilation, PVR was again decreased and oxygenation improved. 181At 0.86 of term, initiation of mechanical ventilation caused pulmonary vasodilation, which was further increased by NO inhalation but not by 100% oxygen. Near term (0.96), NO inhalation and 100% oxygen administration both further increased the pulmonary vasodilation caused by mechanical ventilation. 182 

Models of Acute Pulmonary Artery Hypertension and Lung Injury in Adult Animals 

Selective pulmonary vasodilation during NO inhalation has been shown in numerous animal models: after pharmacologic preconstriction of the pulmonary vasculature with U46619, a synthetic thromboxane analog 4; after a heparin–protamine reaction that induces thromboxane-mediated pulmonary vasoconstriction 183; after pulmonary oleic acid instillation, which induces endothelial and alveolar edema, cell necrosis, and PAH 184,185; and after bilateral lung lavage, which depletes surfactant. 186Inhaled NO is also an effective pulmonary vasodilator in endotoxin-induced PAH 187–189and after smoke inhalation injury. 190 

Lung Injury Induced by Neutrophil-derived Oxidants and by Molecular Oxygen 

Reactive oxygen species (e.g. , H2O2, O2) promote lung injury in various clinical settings. 191–194The use of high inspired concentrations of oxygen is sometimes necessary during the treatment of acute lung injury, but these high oxygen concentrations may cause or worsen lung injury. 195,196Thus, the effects of NO inhalation during lung injury caused by neutrophil oxidants or oxygen are of particular interest. Reaction of NO with oxygen or O2results in NO2or OONO formation, which may damage the lung. 33,34,96In contrast, NO may also protect against the cellular toxicity of H2O2, alkyl peroxides, and O2. 197 

Data from isolated lung studies suggest that the net physiologic effects of NO inhalation protects against tissue injury by typical neutrophil oxidants. In buffer-perfused isolated rabbit lungs, NO inhalation decreased H2O2-induced pulmonary edema, pulmonary vascular permeability, 198and edema formation after injury with O2(generated by the reaction of purine with xanthine oxidase). 199In hyperoxic lung injury, low-dose (10 ppm) but not high-dose (100 ppm) inhaled NO completely prevented pulmonary edema caused by inhalation of 100% oxygen for 40 h (fig. 3). 200In rats exposed for 60 h to 95% oxygen, pulmonary endothelial permeability, protein transfer, and type I alveolar epithelial cell injury were attenuated by inhalation of 20 ppm NO. 201,In vitro , however, using isolated microvascular endothelial cells and alveolar epithelial cells, simultaneous exposure to a NO donor drug and hyperoxia was associated with earlier cell death, as compared to hyperoxia alone. These in vitro  findings have been related to an increased production of OONO. 202 

Fig. 3. Effects of 100% oxygen exposure with or without inhalation of nitric oxide (NO; 10 and 100 ppm) on pulmonary edema in rats. (  Upper ) After 40 h of oxygen exposure, the increase of the wet to dry ratio (QW/QD) was prevented by inhalation of 10 ppm NO, but not by 100 ppm NO. (  Lower ) The increase of the alveolar–arterial oxygen difference was also prevented by inhalation of 10 ppm NO, but not by 100 ppm NO. Data are mean ± SD of six rats in each group. Reprinted with permission from Garat  et al. 200 

Fig. 3. Effects of 100% oxygen exposure with or without inhalation of nitric oxide (NO; 10 and 100 ppm) on pulmonary edema in rats. (  Upper ) After 40 h of oxygen exposure, the increase of the wet to dry ratio (QW/QD) was prevented by inhalation of 10 ppm NO, but not by 100 ppm NO. (  Lower ) The increase of the alveolar–arterial oxygen difference was also prevented by inhalation of 10 ppm NO, but not by 100 ppm NO. Data are mean ± SD of six rats in each group. Reprinted with permission from Garat  et al. 200 

Close modal

Inhibition of Neutrophil Adhesion by NO Inhalation 

Migration and adherence of neutrophils to the pulmonary vasculature and the local release of their oxidants are believed to be key events in oxidant lung injury. 203,204The effects of inhaled NO on neutrophil activity and neutrophil–endothelium interactions, in addition to direct effects of their oxidizing products, have been studied extensively. NO inhalation reduced pulmonary neutrophil accumulation after intestinal ischemia–reperfusion injury in rats 205and after dialysis in pigs. 206In an isolated rat lung model perfused with a mixture of human neutrophils and either N-formyl-L-methionyl-L-leucyl-L-phenylalanine 207or interleukin 1, 208inhalation of 50 ppm NO markedly decreased lung edema formation, neutrophil accumulation and neutrophil migration from the vascular into the alveolar space. Similarly, inhaled NO reduced pulmonary leukocyte sequestration in premature lambs with severe respiratory distress. 209In an in vivo  porcine model of Pseudomonas aeroginosa  sepsis, inhalation of NO (20 ppm) for 5 h after bacterial infusion had significant beneficial effects on pulmonary neutrophil sequestration and neutrophil oxidant activity. Inhaled NO reduced protein and neutrophil sequestration into the alveolar space. Neutrophil oxidant activity (stimulated O2production) and alveolar structural damage (assessed by electron microscopy) were also reduced in these septic lungs treated with inhaled NO. 210However, opposing effects also have been reported. Increased oxidant activity (production of O2and OONO) from intraalveolar neutrophils and increased protein sequestration into the alveolar space after NO inhalation have been observed. 211In a rat model of intratracheally administered endotoxin, inhaled NO (15 ppm) failed to prevent neutrophil sequestration and activation when inhalation was commenced 8 h after endotoxin challenge. 212One important difference in these studies was the time point when NO inhalation was begun; early NO inhalation (with respect to bacterial or endotoxin challenge) appeared to be associated with more effective inhibition of neutrophil activation and sequestration than did NO inhalation begun at a later time.

The effects of inhaled NO on pulmonary neutrophil sequestration may be mediated by modification of adhesion molecule expression and inhibition of the adherence of stimulated neutrophils to the endothelium and their migration through endothelial cell layers. In vitro  studies report that expression of a variety of endothelial and neutrophil adhesion molecules in response to inflammatory stimuli or ischemia–reperfusion injury is modified by molecular NO, NO donor drugs, or the inhibition of endogenous NO synthesis. 213–217NO also scavenges O2released from migrating neutrophils and thereby reduces neutrophil oxidant activity after adherence. 218,219Recent in vitro  data showed a dose-dependent effect of NO on neutrophils: exposure of isolated human neutrophils to an environment of 80% oxygen and 20 ppm NO increased cell death by DNA inhibition, whereas 5 ppm NO did not induce significant DNA fragmentation. 220Whether this apoptotic effect is significant in vivo  is unknown.

High-altitude Pulmonary Edema 

Severe (hypoxic) pulmonary vasoconstriction and hypertension characterize high-altitude pulmonary edema (HAPE). Scherrer et al.  140hypothesized that inhalation of NO would reduce MPAP and thus the severity of HAPE. NO (40 ppm), inhaled at high altitude (4,559 m), decreased MPAP both in subjects prone to HAPE and those with HAPE, but not in HAPE-resistant control subjects. In subjects with HAPE, perfusion scintigraphy showed that inhaled NO redistributed pulmonary blood flow from edematous to nonedematous lung regions. This was associated with an improved PaO2.

Models of Prolonged Hypoxia 

Prolonged exposure to hypoxia induces PAH, pulmonary vascular wall remodeling with neomuscularization, and right ventricular hypertrophy. 221,222Inhaled NO (10–20 ppm), added while breathing at FIO20.1 for 2–3 weeks, effectively prevented PAH, pulmonary vascular remodeling, and right ventricular hypertrophy in adult rats, 223,224in newborn rats, 225and in wild-type and eNOS-deficient mice. 129These salutary effects of inhaled NO may be mediated by direct vasodilatory mechanisms in the pulmonary vasculature and antiproliferative effects of NO on smooth muscle cells. 226 

Models of Pulmonary Embolism and Thrombosis 

Because inhaled NO can reduce reactivity and adhesion of circulating blood cells (e.g. , leukocytes, thrombocytes), it has been hypothesized that thrombus formation may be decreased by inhaled NO. In a rat model of collagen-induced pulmonary thrombosis, inhalation of 80 ppm NO reduced the MPAP increase associated with collagen injection and inhibited ex vivo  collagen-induced platelet aggregation. Rats treated with inhaled NO showed fewer platelet thrombi in small pulmonary vessels and a higher residual circulating platelet count. 11In an in vivo  porcine model of microsphere-induced pulmonary embolism, inhaled NO (5–80 ppm) reduced the increase of MPAP and increased the end-tidal carbon dioxide concentration. Platelet aggregation was increased with pulmonary embolism in control animals. Inhaled NO decreased the initial and maximum platelet aggregation. 227 

Experimental Models of Lung Transplantation 

A beneficial effect of NO inhalation on ischemia–reperfusion injury, graft function, PAH, and oxygenation after lung transplantation has been reported in various experimental studies. 228–232The shortage of suitable donor lungs allows only a small percentage of potential recipients to receive a lung transplant. 233,234It has been suggested to harvest donor lungs from non–heart-beating donors to increase the number of lungs available for transplantation. 235,236Bacha et al.  studied whether NO inhalation can improve the function of lungs harvested after cardiac arrest in the donor. In pig and rat models, they treated the donor (after cardiac arrest) and recipient with inhaled NO (30 ppm) and demonstrated a significant improvement of oxygenation and short-term graft survival after transplantation, as well as reduction of PAH and decreased pulmonary neutrophil accumulation. 237,238 

Bleeding Time 

Nitric oxide stimulates cGMP formation in platelets and, thus, NO inhalation may inhibit platelet function and augment a bleeding tendency in some species. Högman et al.  239reported that the bleeding time increased after rabbits inhaled NO. They noted that breathing 30 ppm NO for 15 min increased the bleeding time from 51 ± 5 to 72 ± 7 s (mean ± SE), and breathing 300 ppm NO for 15 min increased the bleeding time from 48 ± 12 to 78 ± 17 s. However, bleeding time was not altered in rats breathing 80 ppm NO for 1 h 240or in dogs breathing 20–200 ppm NO for 45 min. 10The reason for these differences among species is unknown.

Vascular Injury in the Systemic Circulation 

Inhaled NO may affect the systemic vasculature and cells circulating within the systemic circulation, possibly by reversibly binding to hemoglobin 57or other proteins, with subsequent transport and release of NO at distant sites, or by modification of leukocytes and platelets during their transit through the lung. In a rat model of carotid injury, which is associated with migration and proliferation of smooth muscle cells in the arterial intima, inhalation of 80 ppm for 2 weeks decreased the degree of neointimal formation. 240In a canine model of coronary artery thrombosis and lysis, NO inhalation decreased the cyclic flow variation frequency and increased the duration of periods of coronary artery patency. In the latter model, inhaled NO had no vasodilatory effects on the pharmacologically preconstricted coronary artery segment and did not change the bleeding time. Therefore, coronary patency after thrombolysis was increased by NO inhalation, independent of direct vasodilatory activity or increased bleeding time. 10In contrast to pulmonary vasodilation, these actions of inhaled NO cannot be mediated by any direct effects of gaseous NO on smooth muscle cell relaxation.

Further insights into the mechanisms of the systemic effects of NO inhalation were recently provided by Fox-Robichaub et al.  9In a cat model of intestinal ischemia and reperfusion, inhalation of 80 ppm attenuated the reduction of perfusion, increase of leukocyte rolling, adhesion and migration, and endothelial dysfunction. Changes in leukocyte activity and vessel size were directly visualized by in vivo  microscopy and were induced by inhalation of 80 ppm but not 20 ppm NO. These effects were independent of intrapulmonary modification of leukocyte adhesion molecules, suggesting that inhaled NO was bound to transport molecules and was released in the peripheral circulation. 9 

Sickle Cell Hemoglobin 

Homozygous sickle cell anemia is a genetic disease characterized by severe hemolytic anemia, frequent vasoocclusive events, and a reduced life expectancy. A single amino acid substitution from valine to glutamic acid of the hemoglobin β chain results in hemoglobin S (HbS) formation. At deoxygenation, an erythrocyte containing HbS changes its shape from a biconcave disk to a crescent sickle cell because of intracellular hemoglobin polymerization. Sickle cells can occlude the microcirculation. 241–243 

Hemoglobin S has a markedly decreased oxygen affinity, measured as a markedly increased P50(partial pressure of oxygen at half saturation of hemoglobin), compared with adult hemoglobin. In studies by Head et al. , 12inhalation of 80 ppm NO for 45 min by patients with homozygous sickle cell disease shifted the oxygen dissociation curve of their erythrocytes 4.6 ± 2.0 mmHg to the left, significantly decreasing the P50(fig. 4). Methemoglobin concentrations did not increase substantially. In five of seven volunteers with sickle cell disease, the effect persisted for at least 60 min after discontinuing NO. 12In normal volunteers, the P50was not affected by breathing NO. 12Precisely how inhaled NO alters sickle hemoglobin is unknown. One hypothesis is that the Cysβ93 residue of HbS is modified by NO, increasing HbS solubility and decreasing the tendency to polymerize during deoxygenation. 244,245 

Fig. 4. Effects of nitric oxide (NO) on oxygen affinity of erythrocytes. Exposure to NO (80 ppm for 15 min) shifted the oxygen dissociation curve of hemoglobin S erythrocytes to the left (  left ). The effect of NO exposure on P50of hemoglobin S erythrocytes was dose-dependent (  right ). Values are mean ± SE. Reprinted with permission from Head  et al. 12 by copyright permission of The American Society for Clinical Investigation. 

Fig. 4. Effects of nitric oxide (NO) on oxygen affinity of erythrocytes. Exposure to NO (80 ppm for 15 min) shifted the oxygen dissociation curve of hemoglobin S erythrocytes to the left (  left ). The effect of NO exposure on P50of hemoglobin S erythrocytes was dose-dependent (  right ). Values are mean ± SE. Reprinted with permission from Head  et al. 12 by copyright permission of The American Society for Clinical Investigation. 

Close modal

The first clinical studies of inhaled NO focused on whether the physiologic effects measured in animal models were reproducible in patients. Acute respiratory distress syndrome (ARDS) and persistent PPHN have been the most commonly studied clinical syndromes. The results of large multicenter studies of NO inhalation in the treatment of critically ill newborns and adults recently have been reported. Inhaled NO also has been tested clinically in other conditions, including chronic PAH, chronic obstructive pulmonary disease (COPD), and lung transplantation and heart surgery.

Respiratory Failure of the Newborn 

PPHN and Hypoxic Respiratory Failure. 

Persistent pulmonary hypertension of the newborn is a clinical syndrome characterized by sustained pulmonary hypertension and severe hypoxemia, resulting in cyanosis unresponsive to oxygen therapy. Persistent pulmonary hypertension of the newborn may be caused by a variety of etiologies (e.g. , aspiration) or can be idiopathic. 246Diagnostic confirmation of PPHN includes echocardiographic observation of a right-to-left shunt through the ductus arteriosus or foramen ovale, caused by increased PVR, in the absence of congenital heart disease. Conventional treatment strategies include breathing high inspired concentrations of oxygen, hyperventilation, and infusion of bicarbonate to produce alkalosis, inhalation treatments with bovine surfactant, and intravenous vasodilator therapy. ECMO may be used to treat hypoxemia. However, the anticoagulation and cannulation of large vessels required for ECMO is associated with hemorrhagic complications.

In 1992, Roberts et al.  247and Kinsella et al.  248reported that 80 ppm 247or 6–20 ppm 248inhaled NO improved oxygenation in patients with PPHN. Several large controlled, randomized multicenter trials of the effects of inhaled NO in near-term and term hypoxic newborn patients were reported in 1997 (tables 1 and 2). 6–8,249,250In the majority of patients with PPHN and hypoxic respiratory failure (in whom the decision to initiate ECMO were made by the clinical team on the basis of center-specific ECMO entry criteria and without knowledge of assignment of the patient to the treatment group or placebo), NO improved oxygenation and decreased the requirement for ECMO. Kinsella et al.  reported that NO inhalation and high-frequency oscillatory ventilation were an effective combination that may increase the rate of responsiveness to inhaled NO. 8In a 1- to 2-yr follow-up study of children who received inhaled NO treatment for PPHN, neurodevelopment scores, growth rates (growth percentiles for weight, length, and occipitofrontal circumference), the frequency of airway disease, and the need for supplemental oxygen were comparable to conventionally ventilated or ECMO-treated patients. 251In summary, NO improved oxygenation in many newborns and, although it did not change overall survival, it reduced the need for ECMO (P < 0.05). 6,7Inhaled NO therapy did not appear to impart any benefits, however, to newborns with congenital diaphragmatic hernia. 249 

Table 1. Multicenter Trials of Inhaled Nitric Oxide in Patients With PPHN 

Table 1. Multicenter Trials of Inhaled Nitric Oxide in Patients With PPHN 
Table 1. Multicenter Trials of Inhaled Nitric Oxide in Patients With PPHN 

Table 2. Short-term Effects of Inhaled Nitric Oxide on Systemic Oxygenation in Infants with Severe Hypoxemia and Persistent Pulmonary Hypertension 

Table 2. Short-term Effects of Inhaled Nitric Oxide on Systemic Oxygenation in Infants with Severe Hypoxemia and Persistent Pulmonary Hypertension 
Table 2. Short-term Effects of Inhaled Nitric Oxide on Systemic Oxygenation in Infants with Severe Hypoxemia and Persistent Pulmonary Hypertension 

Preterm Neonates with Respiratory Distress Syndrome. 

Respiratory distress syndrome (RDS), or hyaline membrane disease, of the premature newborn is characterized by deficiency or dysfunction of surfactant and is often associated with acute PAH. 181After promising preliminary studies of inhaled NO in the premature newborn with RDS, 252,253Skimming et al.  254studied the effect of inhaled NO at 5 and 20 ppm in preterm neonates (without systemic hypotension or congenital malformations and mechanically ventilated at FIO2> 0.5). They demonstrated that arterial oxygenation improved and systemic arterial blood pressure was unaffected during a 15 min NO inhalation trial. The conclusions of this study were limited, however, because MPAP was not measured, and only 7% of the initially evaluated premature infants were included in the study.

Acute Lung Injury and Acute Respiratory Distress Syndrome 

Selective Pulmonary Vasodilation. 

In severe ARDS, PAH augments pulmonary edema and may impede right ventricular function and decrease cardiac output. Rossaint et al.  144demonstrated in patients with severe ARDS that inhaled NO produced selective pulmonary vasodilation. This was later confirmed by larger studies. 142,147Occasionally, the NO-induced pulmonary vasodilation has been associated with improved right ventricular performance, as indicated by improvements in right ventricular ejection fraction and decreased right ventricular end-diastolic and end-systolic volumes. 255In children with ARDS, inhaled NO (20 ppm) decreased MPAP by 25% and increased cardiac index by 14%. 256Inhaled NO also effectively decreased MPAP associated with the use of permissive hypercapnia in patients with ARDS. 257 

Pulmonary Capillary Pressure. 

Inhaled NO (40 ppm) has been reported to decrease pulmonary capillary pressure 258and pulmonary transvascular albumin flux, 259partly caused by its effect on venous PVR in patients with acute lung injury. 258Such reductions of pulmonary venous and pulmonary capillary pressure should promote resolution of pulmonary edema, an important component of ARDS.

Arterial Oxygenation. 

Severe hypoxemia caused by extensive intrapulmonary right-to-left shunting is characteristic of ARDS. Common current strategies of management include lung recruitment by high levels of positive end-expiratory pressure, prone positioning, and ventilation with a high FIO2. Therapies that permit lower airway pressures and FIO2might reduce the risk of barotrauma and oxidant injury to the lung. Inhaling 18 ppm NO for 40 min reduced the shunt fraction by 5% and increased the PaO2/FIO2ratio by 30% in patients with ARDS. 144In a dose-ranging study of ARDS patients breathing NO, the ED50(the dose producing 50% of maximal effect) for increasing PaO2(10–100 ppb) was markedly less than the ED50producing pulmonary vasodilation (1–10 ppm, fig. 5). 260 

Fig. 5. Dose–response of inhaled nitric oxide (NO) for PaO2(  upper ) and mean pulmonary arterial pressure (  lower ) in 12 patients with acute respiratory distress syndrome. The estimated ED50for PaO2increase was 110 ppb and the estimated ED50for mean pulmonary artery pressure (PPA) decrease was 1.2 ppm. Values are mean ± SD. Reprinted with permission from Gerlach  et al. 260 

Fig. 5. Dose–response of inhaled nitric oxide (NO) for PaO2(  upper ) and mean pulmonary arterial pressure (  lower ) in 12 patients with acute respiratory distress syndrome. The estimated ED50for PaO2increase was 110 ppb and the estimated ED50for mean pulmonary artery pressure (PPA) decrease was 1.2 ppm. Values are mean ± SD. Reprinted with permission from Gerlach  et al. 260 

Close modal

In a phase 2 multicenter trial, the effects of NO inhalation on oxygenation were studied in 177 patients who met the criteria of ARDS (PaO2/FIO2< 200 mmHg within the last 72 h, bilateral chest infiltrates, pulmonary capillary wedge pressure (PCWP) < 18 mmHg, positive end-expiratory pressure requirement > 8 cm H2O and FIO2requirement > 0.5). Sixty-five percent of the patients who received inhaled NO (pooled results of patients receiving 1.25, 5, 20, 40, 40, or 80 ppm NO) had a significant (P = 0.0002 vs.  placebo) improvement in PaO2(defined as a 20% increase of PaO2after 4 h of therapy). Only 24% of the patients receiving placebo (nitrogen) responded similarly (fig. 6). 142The improved oxygenation induced by NO allowed physicians to reduce FIO2and positive end-expiratory pressure and thereby decreased the oxygenation index (FIO2× mean airway pressure × 100)/PaO2) for the first 4 days of therapy. The MPAP was slightly lower in the inhaled NO group, compared with placebo, for 2 days. Similar transient improvements of PaO2/FIO2during inhaled NO therapy in patients with ARDS have been shown in prospective studies conducted by Michael et al.  261and Troncy et al.  262The reasons for such transient effects remain unclear.

Fig. 6. Percentage of patients with acute respiratory distress syndrome who respond with a PaO2increase of 20% or more while receiving 0–40 ppm nitric oxide during a 4-h inhalation period. Bars from left to right within each dose group indicate progressive exposure periods of 30 min, 1 h, 2 h, and 4 h. Reprinted with permission from Dellinger  et al. 142 

Fig. 6. Percentage of patients with acute respiratory distress syndrome who respond with a PaO2increase of 20% or more while receiving 0–40 ppm nitric oxide during a 4-h inhalation period. Bars from left to right within each dose group indicate progressive exposure periods of 30 min, 1 h, 2 h, and 4 h. Reprinted with permission from Dellinger  et al. 142 

Close modal

Outcome. 

The phase 2 U.S. multicenter study of the effects of inhaled NO on ARDS patients (discussed previously) reported a mortality rate of 30% in NO-(all doses pooled) and placebo-treated patients both. 142The patients were assigned randomly to receive 1.25, 5, 20, 40, or 80 ppm NO or nitrogen placebo. Subgroups receiving the same dose of NO for the treatment period consisted of 8–34 patients. Mortality rates in the subgroups were 32%(7 of 22 patients) in the 1.25 ppm group, 24%(8 of 24 patients) in the 5 ppm group, 31%(9 of 29 patients) in the 20 ppm group, 30%(8 of 27 patients) in the 40 ppm group, and 38%(3 of 8 patients) in the 80 ppm group. A prospective randomized study reported by Troncy et al.  262similarly found no significant difference between ARDS patients receiving inhaled NO and controls with regard to 30-day mortality or days of mechanical ventilation. Several hypotheses should be addressed in the interpretation of these data and should be kept in mind for the design of future studies:

  1. The beneficial effects of NO inhalation (e.g. , improvement of gas exchange) may not alter the overall outcome because the survival of patients with ARDS may not be primarily dependent on gas exchange. The majority of patients dying with ARDS also have severe sepsis or multiple organ failure. The incidence of death primarily because of respiratory failure varies among studies. In a study by Montgomery et al.  published in 1985, the percentage of ARDS deaths specifically because of hypoxemia and respiratory failure was reported to be 16%. 263A study of patients treated at the LDS Hospital in Salt Lake City reported that 40% of deaths in ARDS patients were caused by respiratory failure. 264It is unknown whether this subgroup of patients would benefit from inhaled NO because such severely hypoxemic patients have been excluded from prospective studies.

  2. The beneficial effects of NO inhalation are offset by its toxic effects. As described previously, NO has several negative effects on biologic molecules and tissues, especially if OONO is formed. This hypothesis needs further testing in patients, e.g. , by analyzing lung specimens for evidence of NO or OONO toxicity after NO inhalation.

  3. NO inhalation has a narrow therapeutic range. Small doses, e.g. , 1–5 ppm, could be effective and improve survival, but smaller doses might be not effective, and larger doses could be toxic. The effective dose may change over time and vary among different patients and different disease states. 265It is necessary to establish improved dosing criteria. Larger patient groups receiving similar doses are necessary to discover statistically robust differences. Currently, a blinded, multicenter phase 3 study is being completed that investigated the effects of 5 ppm inhaled NO, compared with placebo, in patients with ARDS. Such a protocol assumes that the dose–response relation of inhaled NO is similar among ARDS patients and over time. This may not be true. 265 

Performing such trials is difficult and expensive. The incidence of ARDS is relatively low, and the precipitating events are often multifactorial. Usually a large number of centers must participate to recruit sufficient numbers of patients. There may be significant differences in response rates and outcomes among different centers, as reported for PPHN patients by Kinsella et al.  8Different treatment strategies and the experiences of individual institutions and caregivers may provide confounding variables. As a result, conclusive studies evaluating the effects of a drug such as inhaled NO, which at best has a modest effect on the survival of a diverse population of patients with ARDS, may not be economically viable.

Chronic Pulmonary Artery Hypertension 

The pathophysiology of chronic PAH includes a partially reversible increase of MPAP in the early stages of the disease, leading to a nonreactive and irreversibly remodeled pulmonary vasculature after long-standing PAH. 266Vascular remodeling is characterized by muscularization of previously nonmuscular small resistance arteries, medial hypertrophy of proximal pulmonary arteries, and a reduced number of arteries within the lung. Diagnosis and medical treatment of chronic PAH relies on vasodilator therapy. When medical treatment is no longer possible (e.g. , no vasodilator response) or ineffective (e.g. , tachyphylaxis or tolerance), lung transplantation remains as the last option to prolong life.

Evaluation. 

The determination of pulmonary vascular responsiveness is essential for prognosis and long-term treatment. Drugs commonly used to assess pulmonary vasodilatory responses include intravenous prostacyclin (PGI2), adenosine, and calcium channel blockers. 267After studying adult patients with inhaled NO and infused prostacyclin, Sitbon et al.  suggested using inhaled NO as the “gold-standard” to assess pulmonary vasoreactivity 268because inhaled NO was reported to selectively reduce MPAP in patients with pulmonary hypertension. 5A recent survey of long-term vasodilator treatment in approximately 800 patients with primary pulmonary hypertension reported that inhaled NO was used as the primary vasodilator to test pulmonary vascular responsiveness by 32% of the participating U.S. tertiary hospitals. 269 

As opposed to commonly used intravenous vasodilators, which can produce systemic hypotension, inhaled NO does not significantly affect systemic vascular resistance. This permits the rapid and safe evaluation of changes of biventricular function during a brief trial of pulmonary vasodilation and, therefore, provides an important diagnostic tool for the decision to begin medical treatment or to plan lung transplantation or combined heart and lung transplantation. 270 

Treatment. 

In chronic PAH, a positive response to a short-term vasodilator trial usually results in long-term drug treatment that may include a wide spectrum of systemic vasodilator drugs (e.g. , acetylcholine, α-adrenergic agonists, direct-acting vasodilators, angiotensin-converting enzyme inhibitors, calcium channel blockers, prostaglandins) and permanent anticoagulation. 267The effectiveness of current vasodilator treatment often is limited by systemic hypotension. Channick et al.  271tested an ambulatory NO delivery system consisting of an 80-ppm NO tank, a gas–pulsing device, and a nasal cannula in eight PAH patients with a pulmonary artery catheter. They reported that this technique produced effective pulmonary vasodilation without evidence of significant nitrogen dioxide formation. One of the eight patients was discharged from the hospital and treated for 9 months with inhaled NO without any apparent adverse events. Long-term domiciliary NO inhalation as an alternative or a bridge to lung transplantation 272requires investigation in larger patient groups, after consideration of the beneficial and toxic effects caused by long-term NO inhalation.

Obstructive Airway Disease 

Inhaled NO has been tested for use as a pulmonary vasodilator in COPD and as a bronchodilator in COPD and asthma. COPD is characterized by irreversible airway obstruction and is associated with irregular enlargement of alveoli and destruction of alveolar walls after chronic inflammation. Hypoxia produces pulmonary vasoconstriction, resulting in chronic PAH and right ventricular hypertrophy. Important characteristics of asthma include inflamed, hyperreactive airways with reversible bronchoconstriction.

Bronchodilator Action. 

Inhaled NO has been reported to be a bronchodilator in many experimental animal models. Högman et al.  169evaluated inhalation of 80 ppm NO in healthy volunteers, in patients with hyperreactive airways, bronchial asthma, and COPD. Inhaled NO caused mild bronchodilation in patients with asthma but not in patients with COPD. In other studies, however, the bronchodilator action of NO has been reported to be much weaker than commonly used inhaled β2-adrengeric agonists. 168,273,274 

Pulmonary Vasodilation. 

Hypoxemia in COPD is primarily caused by a mismatch and not by intrapulmonary right-to-left shunting (as in ARDS). Hypoxic vasoconstriction augments blood flow to better ventilated regions and improves oxygenation. Inhaled NO may oppose this physiologically useful mechanism by vasodilating poorly ventilated areas in the obstructed lung, and thus increasing blood flow to these areas, as reported by Hopkins et al.  155Indeed, transcutaneous arterial oxygen tension, 275PaO2, and V̇/Q̇ distribution 276were worsened by NO inhalation in air-breathing COPD patients. However, when NO was used in combination with modest oxygen enrichment, 277PaO2was improved to a greater extent than with oxygen therapy alone. The combination also more effectively decreased MPAP. Thus, the combined use of supplemental oxygen and inhaled NO (e.g. , via  an ambulatory inhalation device) may offer a valuable therapeutic strategy for improving oxygenation and providing pulmonary vasodilation in selected COPD patients.

Lung Transplantation 

Pulmonary artery hypertension frequently occurs in the immediate postoperative period after lung transplantation and has been effectively treated with inhaled NO. 278Inhaled NO has been reported to be effective in the treatment of post–lung transplant pulmonary dysfunction. In a retrospective study by Date et al. , 279243 patients undergoing lung transplantation over 6 yr were analyzed. Thirty-two patients had immediate severe graft dysfunction, as indicated by a PaO2/FIO2ratio < 150 mmHg. Comparing patients in whom NO treatment was not available with patients in whom NO treatment was begun after graft dysfunction was diagnosed, inhaled NO reduced MPAP and increased the PaO2/FIO2ratio within the first hour of treatment. The requirement for ECMO was similar in both patient groups. The rate of airway complications and hospital mortality (7% in NO group vs.  24% in control group) was markedly reduced in patients receiving NO therapy. As in all retrospective studies using historical controls, changes of treatment strategies and increased clinical experience over time must be considered. Although this study suggests that inhaled NO may decrease post–lung transplant organ dysfunction, it should be confirmed in prospective controlled studies.

Congenital Heart Disease 

The degree and reversibility of the increased PVR determine the various treatment options and outcome in children with congenital heart disease and PAH. 280Roberts et al.  281demonstrated that inhaled NO (80 ppm for 10 min) decreased MPAP without causing systemic vasodilation in children between 3 months and 7 yr of age with congenital cardiac lesions (e.g. , atrioseptal defect, ventricular septal defect, atrioventricular canal). The ability of inhaled NO to decrease the PVR of children with congenital cardiac defects has been confirmed by others. 282,283In preoperative patients with severe right-to-left shunting, inhaled NO increased pulmonary blood flow, decreased extrapulmonary shunt flow and improved oxygenation. 284Inhaled NO, therefore, might provide a therapeutically useful tool for the acute nonsurgical treatment of these patients.

Cardiac Surgery 

Transient PAH is common after repair of congenital cardiac lesions and has been related to damage to the pulmonary vascular endothelium, probably induced by the use of cardiopulmonary bypass. 285Inhaled NO has been reported to ameliorate the postoperative PAH of congenital heart disease 286–291and decrease the need for postoperative ECMO. 292 

Patients with left ventricular valvular disease may have preoperative PAH caused by an increased left atrial pressure with retrograde transmission of pressure into the pulmonary arterial circulation. Pulmonary vascular remodeling occurs as a result of chronic pulmonary venous hypertension and PAH. Pulmonary vascular remodeling and vasoconstriction may persist or slowly decrease with time after valve replacement. Treatment with NO after repair of the valvular disease might relieve the vasoconstrictor component of PAH in these patients.

Fullerton et al.  293reported that 20–40 ppm inhaled NO produced pulmonary vasodilation in patients after aortocoronary bypass. In patients after heart transplantation, inhalation of 20 ppm NO caused significant pulmonary vasodilation but also decreased systemic vascular resistance. 294The decreased SVR was most likely secondary to an improved cardiac output because systemic arterial pressure and PCWP remained unchanged. Intravenous administration of the phosphodiesterase inhibitor dipyridamole markedly augmented the pulmonary vasodilatory response to inhaled NO in some patients after aortic or mitral valve replacement. 295 

Hyporesponsiveness to Inhaled Nitric Oxide 

A considerable number of patients who receive inhaled NO therapy do not respond by either pulmonary vasodilation or improvement of systemic oxygenation. The reported rate of hyporesponders ranges from 30% to 45%, depending on the threshold value chosen to define hyporesponsiveness. 142,147Several hypotheses have been raised to explain the mechanisms of hyporesponsiveness. Manktelow et al.  147and Krafft et al.  296reported that ARDS patients with sepsis were less likely to respond to inhaled NO (60–70% of septic patients were hyporesponders). The presence of high levels of endogenously produced NO and the opposing pulmonary vasoconstrictor action of catecholamines used for the treatment of septic vasodilation have been suggested as possible reasons for the decreased response to inhaled NO. Holzmann et al.  297evaluated the effects of sepsis on NO responsiveness in an isolated rat lung model. They reported that hyporesponsiveness was associated with decreased pulmonary cGMP release (table 3), suggesting that signal transduction in the NO response pathway is downregulated in sepsis. This was attributed to increased phosphodiesterase activity and therefore increased cGMP breakdown. Bigatello et al.  reported that hyporesponsive patients with ARDS have a reduced accumulation of plasma cGMP during NO breathing. 298 

Table 3. Inhaled Nitric Oxide–stimulated Pulmonary cGMP Release in Control and Septic Isolated Perfused Rat Lungs (treated with LPS) before and after Ventilation with 40 ppm Nitric Oxide 

Table 3. Inhaled Nitric Oxide–stimulated Pulmonary cGMP Release in Control and Septic Isolated Perfused Rat Lungs (treated with LPS) before and after Ventilation with 40 ppm Nitric Oxide 
Table 3. Inhaled Nitric Oxide–stimulated Pulmonary cGMP Release in Control and Septic Isolated Perfused Rat Lungs (treated with LPS) before and after Ventilation with 40 ppm Nitric Oxide 

Increased vascular production of O2, as observed in systemic nitrate tolerance, also may contribute to hyporesponsiveness to inhaled NO. Munzel et al.  reported that long-term nitrate treatment of rabbits resulted in increased O2production by the aorta and hyporesponsiveness (tolerance) to acute nitroglycerin administration, related to an activated membrane-associated oxidase. 299,300Increased O2production in the pulmonary vasculature may have similar effects.

Lastly, Weimann et al.  301recently reported a linkage between ABO blood group distribution and hyporesponsiveness to inhaled NO, demonstrating that ARDS patients with the major blood groups A or O had a larger increase of PaO2/FIO2in response to NO inhalation than patients with blood groups B or AB. The underlying mechanism of these results is unknown, but they indicate that the pulmonary vascular response to inhaled NO may be determined or modified by genetic factors.

Strategies to Increase Responsiveness 

Phosphodiesterase Inhibition. 

Because cGMP is hydrolyzed by phosphodiesterase, inhibition of phosphodiesterase may increase the effectiveness and duration of the action of inhaled NO. In awake lambs with U46619-induced PAH, intravenous infusion of zaprinast, a phosphodiesterase 5–specific inhibitor, increased the duration of action of inhaled NO and accentuated the NO-induced reduction of PVR. With administration of zaprinast, the half-time of pulmonary vasodilation after discontinuing a 4-min 40-ppm NO inhalation trial was increased from 1 or 2 min to 10–12 min (fig. 7). 302Potentiation of the effects of NO by another phosphodiesterase inhibitor, dipyridamole, has been reported in the ovine fetal pulmonary circulation. 303,304 

Fig. 7. Influence of continuous intravenous zaprinast infusion (0.1 mg · kg−1· min−1) on the magnitude of peak decreases of mean pulmonary arterial pressure (  A ), percent changes of pulmonary vascular resistance (  B ), and half-times of vasodilating effects (  C ) in response to nitric oxide inhalation during pulmonary arterial hypertension induced by U46619 in awake lambs. Values are mean ± SE. *Significantly different from control value (  P < 0.05). Reprinted with permission from Ichinose  et al. 302 

Fig. 7. Influence of continuous intravenous zaprinast infusion (0.1 mg · kg−1· min−1) on the magnitude of peak decreases of mean pulmonary arterial pressure (  A ), percent changes of pulmonary vascular resistance (  B ), and half-times of vasodilating effects (  C ) in response to nitric oxide inhalation during pulmonary arterial hypertension induced by U46619 in awake lambs. Values are mean ± SE. *Significantly different from control value (  P < 0.05). Reprinted with permission from Ichinose  et al. 302 

Close modal

Inhibition of Vascular Superoxide Production. 

No experimental or clinical studies have yet determined whether inhibition of pulmonary vascular O2production may increase responsiveness to inhaled NO. Inhibition of the O2-generating membrane-associated oxidase in rabbit aorta by hydralazine normalized vascular O2production and restored the vasodilatory activity of nitroglycerin. 299,300 

Almitrine Infusion. 

Almitrine bismesylate acts as an agonist at peripheral arterial chemoreceptors and increases discharge of the carotid sinus nerve. 305Intravenous administration of low concentrations of almitrine has been reported to restore or enhance hypoxic vasoconstriction in the acutely injured lung. Inequalities and the PaO2in ARDS patients usually are improved by almitrine administration, which augments hypoxic vasoconstriction and redistributes pulmonary arterial blood flow toward better ventilated areas with a higher PAO2. The combination of almitrine infusion and NO inhalation, each having different mechanism, might synergistically reduce mismatch. Almitrine administration enhanced the oxygenation beneficial effects of inhaled NO on oxygenation 306and allowed a further reduction of FIO2in patients with ARDS. 307The relatively long in vivo  half-life (mean tissue half-life, 12 h) and possible toxic side effects of almitrine (including a slowly reversible peripheral sensory neuropathy) require careful investigation before this regimen can be recommended for routine clinical use.

Partial Liquid Ventilation. 

Perfluorocarbons are inert liquids that lower surface tension in surfactant-depleted lungs and dissolve large concentrations of respiratory gases. Zobel et al.  308demonstrated that inhaled NO enhanced the effects of perfluorocarbons on pulmonary gas exchange in a piglet model of acute lung injury induced by repeated bilateral lung lavage. An additive effect of inhaled NO and perfluorocarbons on pulmonary gas exchange in acute lung injury has been confirmed by others. 309,310 

Left Ventricular Function 

The risks of inhaled NO should be carefully considered in patients with markedly impaired left ventricular function (e.g. , heart transplant candidates). Inhalation of NO may vasodilate the pulmonary circulation and increase blood flow to the left ventricle, thereby acutely increasing left atrial pressure and PCWP 311–313and promoting pulmonary edema formation. 314Cardiac output, left atrial pressure, or PCWP should be monitored if NO is administered to patients with severe left ventricular dysfunction.

Discontinuation of Nitric Oxide Inhalation 

Rebound PAH, an increase of intrapulmonary right-to-left shunting and a decreased PaO2after acute NO discontinuation is well-described. 144Lavoie et al.  315reported four patients with severe acute respiratory failure in whom NO therapy was discontinued abruptly after a decision to discontinue life-extending measures. A sustained decrease of arterial oxygen saturation occurred immediately after NO discontinuation, which was reversed by restarting NO therapy. All four patients died within 24 h after NO discontinuation.

It has been suggested that downregulation of endogenous NO synthesis by NO inhalation is responsible for rebound PAH. 130,316–318However, recent data obtained in rats with hypoxic pulmonary hypertension suggest that inhibition of endogenous NO synthesis plays a minor role in rebound PAH: no changes of lung eNOS protein levels, NOS activity, endothelium-dependent and -independent vasodilation were reported after 3 weeks inhaling 20 ppm NO. Lung GC activity was transiently decreased after 1 week of NO inhalation, but GC activity was normal after 3 weeks of NO inhalation. 319 

To avoid rebound PAH, a slow stepwise reduction of the inhaled NO concentration with immediate control of any adverse effects (e.g. , reduced oxygen saturation or blood gas tensions, increased MPAP 144) are important to safely wean the patient from inhaled NO. In addition, administration of the phosphodiesterase 5 inhibitor dipyridamole has been reported to prevent rebound PAH in children after cardiac surgery. 320 

Bleeding Time 

Inhaled NO can inhibit platelet function and increased the bleeding time in rabbits. In ARDS patients receiving inhaled NO (3–100 ppm), NO decreased platelet aggregation and agglutination in vitro . However, the in vivo  bleeding time (Ivy bleeding time) was not altered. 321In neonates, a recent study reported that bleeding time doubled after 30 min of 40-ppm NO inhalation. 322However, in the multicenter studies in newborns, no difference in the frequency of bleeding events was observed in NO-treated compared with placebo-treated patients. 6–8,249 

Nitric oxide inhalation therapy should be instituted after careful consideration of potential acute and long-term toxicity. The major concerns are (1) methemoglobinemia, (2) NO2formation, and (3) cellular toxicity. Acute inhaled NO overdose (> 500–1,000 ppm) leads to rapid NO2formation, severe methemoglobinemia, pulmonary alveolar edema and hemorrhage, hypoxemia, and death within minutes to hours. 323 

Blood methemoglobin concentrations and inspired NO2concentrations have been regularly monitored in clinical trials of inhaled NO in adults and neonates. 6,8,142,249In the large number of patients studied in these trials (n = 471) receiving inhaled NO therapy at doses ranging from 1.25 to 80 ppm, significant methemoglobinemia or NO2formation was uncommon (table 4). If methemoglobin or NO2levels increased above predetermined limits, the inhaled NO concentration was decreased. Discontinuation of NO administration because of NO2or methemoglobin formation was only necessary in 3 of 471 patients (0.6%).

Table 4. Adverse Events in Multicenter Trials of Inhaled Nitric Oxide in Newborn and Adult Patients 

Table 4. Adverse Events in Multicenter Trials of Inhaled Nitric Oxide in Newborn and Adult Patients 
Table 4. Adverse Events in Multicenter Trials of Inhaled Nitric Oxide in Newborn and Adult Patients 

The most important requirements for safe NO inhalation therapy are (1) continuous analysis of NO and NO2concentrations (using chemiluminescence or electrochemical analyzers 324,325);(2) frequent calibration of the monitoring equipment;(3) frequent analysis of blood methemoglobin levels;(4) the use of certified tanks; and (5) administration of the lowest NO concentration required.

Little is known about the long-term sequalae of NO inhalation in humans. In 12 newborns receiving NO inhalation treatment (< 20 ppm) for up to 4 days, there were no signs of increased lipid peroxidation product, impaired surfactant activity, or changed cytokine profile. 326However, in two infants requiring prolonged ventilation with NO, nitrotyrosine residues were detected in airway specimens. 326The relative contribution of NO inhalation and endogenous NO formation to nitrotyrosine formation in the lung is unclear because nitrotyrosine formation has been demonstrated in acutely injured lungs without the exogenous administration of NO. 33,34Follow-up studies of adult patients 8 months after NO treatment for ARDS showed no obvious differences in pulmonary function compared to ARDS patients not treated with NO. 327 

In summary, reported data of clinical NO toxicity are sparse. Studies appropriately designed to detect long-lasting or irreversible pathologic effects of NO breathing will be necessary to predict the long-term effects of inhaled NO and establish time and dose limits.

Inhaled Prostacyclin 

Prostacyclin is a natural product of the cyclooxygenase pathway and a potent vasodilator. The initial clinical studies of NO inhalation in ARDS patients compared the pulmonary vasoactive properties of intravenously administered prostacyclin with inhaled NO. 144Although prostacyclin infusion decreased MPAP to the same degree as inhaled NO, prostacyclin decreased PaO2, presumably by reducing hypoxic pulmonary vasoconstriction and caused systemic hypotension. Such adverse effects are commonly observered during intravenous infusion of commonly used vasodilator drugs.

It was hypothesized that the administration of prostacyclin via  aerosol would limit its hemodynamic effects to the lung. This hypothesis was tested in ARDS patients, and the effectiveness of prostacyclin when used as a short-term inhaled aerosol was compared with inhaled NO. 328Similar effects, namely decreased MPAP, decreased intrapulmonary right-to-left shunting, and increased PaO2/FIO2were observed. Systemic vasodilation was not reported with either drug. These effects of inhaled prostacyclin have been confirmed in other studies. 329,330A disadvantage of aerosolized prostacyclin therapy is that systemic absorption can occur, which makes it difficult to maintain pulmonary vasodilation without producing systemic vasodilation for periods lasting more than a few hours. Larger studies, including randomized trials studying the effectiveness and responsiveness to inhaled prostacyclin for longer time periods, are necessary to support a useful therapeutic role.

Nitric Oxide Donor Drugs 

The use of inhaled NO donor drugs has been proposed as an alternative to NO inhalation. Administration of such drugs, which release a defined amount of NO over a prolonged time period, might permit intermittent NO dosing. Adrie et al.  compared the pulmonary vascular effects of inhaled sodium 1-(N,N-diethylamino) diazen-1-ium-1,2-diolate (DEA/NO), which spontaneously generates NO, with the inhalation of sodium nitroprusside and NO gas in awake sheep with pharmacologically induced pulmonary hypertension. 331DEA/NO caused nonselective vasodilation, and sodium nitroprusside was only selective for the pulmonary circulation at low inhaled concentrations, compared with the highly selective effect of NO gas. 331In a pig model of acute pulmonary hypertension, Brilli et al.  compared the effects of the aerosolized NO donors ethylputreanine NONOate (EP/NO) and 2-(dimethylamino) ethylputreanine NONOate (DMAEP/NO). 332They reported that 3-min DMAEP/NO aerosolization caused selective pulmonary vasodilation, which lasted for approximately 30–50 min without effects on systemic arterial pressure or cardiac output. 332Similarly, tracheal instillation of DMAEP/NO resulted in prolonged and pulmonary selective vasodilation. 333EP/NO, aerosolized or instilled, was less effective and the effects were inconsistent. 332,333 

The intravenous infusion of ultra–short-acting NO donor agents might also be an alternative to inhaled NO for producing selective pulmonary vasodilation. In awake, healthy sheep with pulmonary hypertension, the intravenous (systemic) infusion of PROLI/NO (C5H7N3O4Na2· CH3OH), an ultra–short-acting nucleophile/NO adduct that generates NO, produced selective pulmonary vasodilation without affecting the systemic circulation. The selective effect of this intravenous drug was caused by its short half-life in vivo , which resulted in complete NO release during transit of the pulmonary circulation and before reaching the systemic arterial circulation. 334Because such drugs are intravenously administered, oxygenation might be adversely affected in the injured lung through indiscriminant release of hypoxic pulmonary vasoconstriction. Nevertheless, such newly designed NO donor drugs appear promising as a selective pulmonary vasodilator and provide an alternative to NO inhalation.

Inhaled Phosphodiesterase Inhibitors 

The use of inhaled phosphodiesterase inhibitors has been investigated as an alternative or adjunct to NO inhalation. Inhalation of nebulized zaprinast induced selective pulmonary vasodilation and enhanced the effects of inhaled NO in awake lambs. 335However, at a zaprinast concentration (50 mg/ml) producing a similar decrease of MPAP to 20 ppm NO, significant systemic vasodilation was observed (fig. 8).

Fig. 8. Percentage change of mean pulmonary arterial pressdure and systemic arterial pressure (SAP) during inhalation of nitric oxide (NO; 5 and 20 ppm), aerosolized zaprinast (ZAP, 10–50 mg/ml), or both, in awake lambs. *Significantly different from values at pulmonary arterial hypertension (  P < 0.05). #Significantly different from the value at 5 ppm NO (  P < 0.05) and from 10 mg/ml zaprinast (  P < 0.01). Values are mean ± SD. Reprinted with permission from Ichinose  et al. 302 

Fig. 8. Percentage change of mean pulmonary arterial pressdure and systemic arterial pressure (SAP) during inhalation of nitric oxide (NO; 5 and 20 ppm), aerosolized zaprinast (ZAP, 10–50 mg/ml), or both, in awake lambs. *Significantly different from values at pulmonary arterial hypertension (  P < 0.05). #Significantly different from the value at 5 ppm NO (  P < 0.05) and from 10 mg/ml zaprinast (  P < 0.01). Values are mean ± SD. Reprinted with permission from Ichinose  et al. 302 

Close modal

Nitric oxide is produced by almost every healthy mammalian tissue. In health, NO has a myriad of functions that are essential for life. In disease, NO has many effects that can be both helpful and deleterious.

Inhaled NO has made a rapid journey from the laboratory bench to the bedside because of its unique selective pulmonary vasodilator activity and its ready availability. It is the first vasodilator described to provide truly selective pulmonary vasodilation. A large number of basic and clinical research studies have made great steps in delineating its physiology, side effects, and clinical efficacy. Nearly simultaneously, the clinical use of inhaled NO has become widespread. During the past 8 yr, inhaled NO has been used to treat pulmonary hypertension and hypoxemia in thousands of patients worldwide. Inhaled NO is an effective pulmonary vasodilator in many disease states characterized by pulmonary hypertension. In addition, inhaled NO decreases pulmonary venous admixture in diffuse lung injury, and therefore increases systemic oxygenation in many patients. Most importantly, randomized multicenter studies of NO inhalation have shown that this new therapy significantly reduces the necessity for ECMO in newborns with PPHN or hypoxic respiratory failure. NO inhalation has many additional effects that may be clinically beneficial. Inhaled NO has been reported to decrease pulmonary edema formation and lung injury. In the systemic circulation, NO inhalation inhibits peripheral vascular restenosis in animal models of arterial injury, augments the oxygen binding of sickle erythrocytes, and reduces cyclic coronary occlusion in a model of coronary injury and thrombolysis. However, important questions remain:

  1. Does the reduction of pulmonary artery pressure and increased PaO2caused by NO inhalation improve clinical outcome for patients with acute lung injury? In a relatively uniform and well-defined population of patients, newborns with hypoxic respiratory failure, NO inhalation effectively improves oxygenation and significantly reduces the use of ECMO. Avoiding ECMO, a complicated and expensive invasive procedure with limited availability, is an important clinical endpoint and undoubtedly would justify the use of inhaled NO. 6,7The clinical usefulness of inhaled NO in adults remains unclear. Clinical studies of ARDS in adults are complicated by the diverse nature of the patient population, the precipitating causes of lung injury, and the common occurrence of sepsis and multiple organ system failure. Current data from multicenter trials suggest that the mortality rate in moderate lung injury is not significantly changed by NO inhalation. Whether this is because of inappropriate study design, the complex nature and spectrum of ARDS, inefficacy of NO, inappropriate dosing, or counterbalancing toxic effects of NO is unknown.

  2. If available data suggest that survival is unchanged, should clinicians continue to study NO inhalation? Researchers concentrating on the cellular and subcellular effects of NO properly express concerns that NO inhalation may worsen lung injury and damage important structures. Initial clinical studies, however, suggest that toxicity, if present, is extremely low. The doses of NO now commonly used are less than those received with cigarette exposure and are nearly within the atmospheric background range of many urban areas. Many clinical scientists continue to evaluate inhaled NO and find it useful for short-term symptomatic treatment of hypoxemic respiratory failure and pulmonary vasoconstriction.

The pharmacologic and toxicologic profiles of NO inhalation are incomplete. It is necessary to delineate further (1) proper indications, (2) contraindications, (3) sound dosing criteria, (4) organ disease from cellular and subcellular toxicity, and (5) the causes of NO hyporesponsiveness. Randomized clinical studies of patients with carefully defined specific disease states characterized by pulmonary hypertension or hypoxemia (e.g. , pulmonary embolism, severe PAH, postpneumonectomy pulmonary edema, acute rejection after lung transplantation) and in premature newborns with respiratory failure remain to be completed. If such trials are carefully designed and conducted, we may define additional groups of patients that may benefit from, or may be harmed by, inhaled NO. The use of inhaled NO continues to be a unique and fascinating approach to studying and treating diseases as diverse as acute rejection of the transplanted lung and sickle cell crisis. In evaluating this complex field, it is critical that our view does not become colored by a single study or effectiveness in a particular disease state. As with most medical advances, it is the evolution of a wide-ranging body of research that will properly determine the place for NO inhalation therapy in our armamentarium.

The authors thank Dr. Joerg Weimann (Department of Anesthesia and Critical Care, University of Heidelberg, Germany), Dr. Kenneth D. Bloch (Cardiovascular Research Center, Massachusetts General Hospital, Boston), and Dr. Steven Tannenbaum (Massachusetts Institute of Technology, Cambridge) for helpful suggestions and critical discussions.

1.
Palmer RM, Ferrige AG, Moncada S: Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nature 1987; 327: 524–6
2.
Ignarro LJ, Buga GM, Wood KS, Byrns RE, Chaudhuri G: Endothelium-derived relaxing factor produced and released from artery and vein is nitric oxide. Proc Natl Acad Sci U S A 1987; 84: 9265–9
3.
Altman LK: Three americans awarded nobel for discoveries of how a gas affects the body. New York Times October 13, 1998; CXLVIII, No. 51,309:A14
4.
Frostell C, Fratacci MD, Wain JC, Jones R, Zapol WM: Inhaled nitric oxide. A selective pulmonary vasodilator reversing hypoxic pulmonary vasoconstriction. Circulation 1991; 83: 2038–47
5.
Pepke-Zaba J, Higenbottam TW, Dinh-Xuan AT, Stone D, Wallwork J: Inhaled nitric oxide as a cause of selective pulmonary vasodilatation in pulmonary hypertension. Lancet 1991; 338: 1173–4
6.
Roberts JD Jr, Fineman JR, Morin FC3, Shaul PW, Rimar S, Schreiber MD, Polin RA, Zwass MS, Zayek MM, Gross I, Heymann MA, Zapol WM: Inhaled nitric oxide and persistent pulmonary hypertension of the newborn. The Inhaled Nitric Oxide Study Group. N Engl J Med 1997; 336: 605–10
7.
Neonatal Inhaled Nitric Oxide Study Group: Inhaled nitric oxide in full-term and nearly full-term infants with hypoxic respiratory failure. N Engl J Med 1997; 336: 597–604
8.
Kinsella JP, Truog WE, Walsh WF, Goldberg RN, Bancalari E, Mayock DE, Redding GJ, deLemos RA, Sardesai S, McCurnin DC, Moreland SG, Cutter GR, Abman SH: Randomized, multicenter trial of inhaled nitric oxide and high-frequency oscillatory ventilation in severe, persistent pulmonary hypertension of the newborn. J Pediatr 1997; 131: 55–62
9.
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 1998; 101: 2497–505
10.
Adrie C, Bloch KD, Moreno PR, Hurford WE, Guerrero JL, Holt R, Zapol WM, Gold HK, Semigran MJ: Inhaled nitric oxide increases coronary artery patency after thrombolysis. Circulation 1996; 94: 1919–26
11.
Nong Z, Hoyaerst M, Van Pelt N, Collen D, Janssens S: Nitric oxide inhalation inhibits platelet aggregation and platelet-mediated pulmonary thrombosis in rats. Circ Res 1997; 81: 865–9
12.
Head CA, Brugnara C, Martinez-Ruiz R, Kacmarek RM, Bridges KR, Kuter D, Bloch KD, Zapol WM: Low concentrations of nitric oxide increase oxygen affinity of sickle erythrocytes in vitro and in vivo. J Clin Invest 1997; 100: 1193–8
13.
Fukuto JM: Chemistry of nitric oxide: Biologically relevant aspects, Nitric Oxide: Biochemistry, Molecular Biology and Therapeutic Implications. Edited by Ignarro L, Murad F. San Diego, Academic Press, 1995, pp 1–13
14.
Alberts WM: Indoor air pollution: NO, NO2, CO and CO2. J Allergy Clin Immunol 1994; 94: 524–8
15.
Mourgeon E, Levesque E, Duveau C, Law-Koune JD, Charbit B, Ternissien E, Coriat P, Rouby JJ: Factors influencing indoor concentrations of nitric oxide in a Parisian intensive care unit. Am J Respir Crit Care Med 1997; 156: 1692–5
16.
Norman V, Keith CH: Nitrogen oxides in tobacco smoke. Nature 1965; 205: 915–7
17.
NIOSH recommendations for occupational safety and health standards. MMWR Morb Mortal Wkly Rep 1988; 37: 1
18.
Body SC, Hartigan PM: Manufacture and measurement of nitrogen oxides, Inhaled Nitric Oxide: I. Edited by Hess D, Hurford WE. Philiadelphia, WB Saunders, 1997, pp 411–34
19.
Glasson WA, Tuesday CS: The atmospheric thermal oxidation of nitric oxide. J Am Chem Soc 1963; 85: 2901–6
20.
Nishimura M, Hess D, Kacmarek RM, Ritz R, Hurford WE: Nitrogen dioxide production during mechanical ventilation with nitric oxide in adults. Effects of ventilator internal volume, air versus  nitrogen dilution, minute ventilation, and inspired oxygen fraction. A NESTHESIOLOGY 1995; 82: 1246–54
21.
Rabson SR, Quilliam JH, Goldblatt E: Elimination of nitrous fumes from blasting gases. J South Afr Inst Min Metall 1960; 61: 152–99
22.
Shiel FOM: Morbid anatomical changes in the lungs of dogs after inhalation of higher oxides of nitrogen during anaesthesia. Br J Anaest 1967; 39: 413–24
23.
Stavert DM, Lehnert BE: Nitric oxide and nitrogen dioxide as inducers of acute pulmonary injury when inhaled at relatively high concentrations for brief periods. Inhalation Toxicol 1990; 2: 53–67
24.
Evans MJ, Stephens RJ, Cabral LJ, Freeman G: Cell renewal in the lungs of rats exposed to low levels of NO2. Arch Environ Health 1972; 24: 180–8
25.
Rasmussen TR, Kjaergaard SK, Tarp U, Pedersen OF: Delayed effects of NO2exposure on alveolar permeability and glutathione peroxidase in healthy humans. Am Rev Respir Dis 1992; 146: 654–9
26.
Bylin G, Hedenstierna G, Lindvall T, Sundin B: Ambient nitrogen dioxide concentrations increase bronchial responsiveness in subjects with mild asthma. Eur Respir J 1988; 1: 606–12
27.
Morrow PE, Utell MJ, Bauer MA, Smeglin AM, Frampton MW, Cox C, Speers DM, Gibb FR: Pulmonary performance of elderly normal subjects and subjects with chronic obstructive pulmonary disease exposed to 0.3 ppm nitrogen dioxide. Am Rev Respir Dis 1992; 145: 291–300
28.
Stephens RJ, Freeman G, Evans MJ: Early response of lungs to low levels of nitrogen dioxide. Light and electron microscopy. Arch Environ Health 1972; 24: 160–79
29.
Mercer RR, Costa DL, Crapo JD: Effects of prolonged exposure to low doses of nitric oxide or nitrogen dioxide on the alveolar septa of the adult rat lung. Lab Invest 1995; 73: 20–8
30.
Darley-Usmar V, Halliwell B: Blood radicals: Reactive nitrogen species, reactive oxygen species, transition metal ions, and the vascular system. Pharm Res 1996; 13: 649–62
31.
Szabo C: The pathophysiological role of peroxynitrite in shock, inflammation, and ischemia-reperfusion injury. Shock 1996; 6: 79–88
32.
Szabo C: DNA strand breakage and activation of poly-ADP ribosyltransferase: A cytotoxic pathway triggered by peroxynitrite. Free Radic Biol Med 1996; 21: 855–69
33.
Haddad IY, Pataki G, Hu P, Galliani C, Beckman JS, Matalon S: Quantitation of nitrotyrosine levels in lung sections of patients and animals with acute lung injury. J Clin Invest 1994; 94: 2407–13
34.
Kooy NW, Royall JA, Ye YZ, Kelly DR, Beckman JS: Evidence for in vivo peroxynitrite production in human acute lung injury. Am J Respir Crit Care Med 1995; 151: 1250–4
35.
Beckman JS: Oxidative damage and tyrosine nitration from peroxynitrite. Chem Res Toxicol 1996; 9: 836–44
36.
Beckman JS, Koppenol WH: Nitric oxide, superoxide, and peroxynitrite: The good, the bad, and ugly. Am J Physiol 1996; 271: 1424–37
37.
Ischiropoulos H, al-Mehdi AB, Fisher AB: Reactive species in ischemic rat lung injury: Contribution of peroxynitrite. Am J Physiol 1995; 269: L158–64
38.
Robbins CG, Davis JM, Merritt TA, Amirkhanian JD, Sahgal N, Morin FC, Horowitz S: Combined effects of nitric oxide and hyperoxia on surfactant function and pulmonary inflammation. Am J Physiol 1995; 269: L545–50
39.
Haddad IY, Crow JP, Hu P, Ye Y, Beckman J, Matalon S: Concurrent generation of nitric oxide and superoxide damages surfactant protein A. Am J Physiol 1994; 267: L242–9
40.
Hallman M, Bry K: Nitric oxide and lung surfactant. Semin Perinatol 1996; 20: 173–85
41.
Haddad IY, Ischiropoulos H, Holm BA, Beckman JS, Baker JR, Matalon S: Mechanisms of peroxynitrite-induced injury to pulmonary surfactants. Am J Physiol 1993; 265: L555–64
42.
Denicola A, Freeman BA, Trujillo M, Radi R: Peroxynitrite reaction with carbon dioxide/bicarbonate: Kinetics and influence on peroxynitrite-mediated oxidations. Br J Pharmacol 1997; 121: 485–90
43.
Lefer DJ, Scalia R, Campbell B, Nossuli T, Hayward R, Salamon M, Grayson J, Lefer AM: Peroxynitrite inhibits leukocyte-endothelial cell interactions and protects against ischemia-reperfusion injury in rats. J Clin Invest 1997; 99: 684–91
44.
Corbett JA, Kwon G, Turk J, McDaniel ML: IL-1 beta induces the coexpression of both nitric oxide synthase and cyclooxygenase by islets of Langerhans: Activation of cyclooxygenase by nitric oxide. Biochemistry 1993; 32: 13767–70
45.
Stadler J, Harbrecht BG, Di Silvio M, Curran RD, Jordan ML, Simmons RL, Billiar TR: Endogenous nitric oxide inhibits the synthesis of cyclooxygenase products and interleukin-6 by rat Kupffer cells. J Leukoc Biol 1993; 53: 165–72
46.
Stadler J, Trockfeld J, Schmalix WA, Brill T, Siewert JR, Greim H, Doehmer J: Inhibition of cytochromes P4501A by nitric oxide. Proc Natl Acad Sci U S A 1994; 91: 3559–63
47.
Wink DA, Osawa Y, Darbyshire JF, Jones CR, Eshenaur SC, Nims RW: Inhibition of cytochromes P450 by nitric oxide and a nitric oxide-releasing agent. Arch Biochem Biophys 1993; 300: 115–23
48.
Doyle MP, Hoekstra JW: Oxidation of nitrogen oxides by bound dioxygen in hemoproteins. J Inorg Biochem 1981; 14: 351–8
49.
Curry S: Methemoglobinemia. Ann Emerg Med 1982; 11: 214–21
50.
Mansouri A, Lurie AA: Concise review: Methemoglobinemia. Am J Hematol 1993; 42: 7–12
51.
Upchurch GR, Welch GN, Loscalzo J: The vascular biology of S-nitrosothiols, nitrosated derivatives of thiols. Vasc Med 1996; 1: 25–33
52.
Ignarro LJ, Lippton H, Edwards JC, Baricos WH, Hyman AL, Kadowitz PJ, Gruetter CA: Mechanism of vascular smooth muscle relaxation by organic nitrates, nitrites, nitroprusside and nitric oxide: Evidence for the involvement of S-nitrosothiols as active intermediates. J Pharmacol Exp Ther 1981; 218: 739–49
53.
Stamler JS, Slivka A: Biological chemistry of thiols in the vasculature and in vascular-related disease. Nutr Rev 1996; 54: 1–30
54.
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 1996; 79: 256–62
55.
Do KQ, Benz B, Grima G, Gutteck-Amsler U, Kluge I, Salt TE: Nitric oxide precursor arginine and S-nitrosoglutathione in synaptic and glial function. Neurochem Int 1996; 29: 213–24
56.
Jia L, Bonaventura J, Stamler JS: S-nitrosohaemoglobin: a dynamic activity of blood involved in vascular control. Nature 1996; 380: 221–6
57.
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 1997; 276: 2034–7
58.
Tamir S, Burney S, Tannenbaum SR: DNA damage by nitric oxide. Chem Res Toxicol 1996; 9: 821–7
59.
Tamir S, DeRojas-Walker T, Wishnok JS, Tannenbaum SR: DNA damage and genotoxicity by nitric oxide. Methods Enzymol 1996; 269: 230–43
60.
Denicola A, Rubbo H, Rodriguez D, Radi R: Peroxynitrite-mediated cytotoxicity to Trypanosoma cruzi. Arch Biochem Biophys 1993; 304: 279–86
61.
Bonfoco E, Krainc D, Ankarcrona M, Nicotera P, Lipton SA: Apoptosis and necrosis: Two distinct events induced, respectively, by mild and intense insults with N-methyl-D-aspartate or nitric oxide/superoxide in cortical cell cultures. Proc Natl Acad Sci U S A 1995; 92: 7162–6
62.
Lin KT, Xue JY, Nomen M, Spur B, Wong PY: Peroxynitrite-induced apoptosis in HL-60 cells. J Biol Chem 1995; 270: 16487–90
63.
Kwon NS, Stuehr DJ, Nathan CF: Inhibition of tumor cell ribonucleotide reductase by macrophage-derived nitric oxide. J Exp Med 1991; 174: 761–7
64.
Lepoivre M, Fieschi F, Coves J, Thelander L, Fontecave M: Inactivation of ribonucleotide reductase by nitric oxide. Biochem Biophys Res Commun 1991; 179: 442–8
65.
Wink DA, Vodovotz Y, Laval J, Laval F, Dewhirst MW, Mitchell JB: The multifaceted roles of nitric oxide in cancer. Carcinogenesis 1998; 19: 711–21
66.
Wink DA, Kasprzak KS, Maragos CM, Elespuru RK, Misra M, Dunams TM, Cebula TA, Koch WH, Andrews AW, Allen JS: DNA deaminating ability and genotoxicity of nitric oxide and its progenitors. Science 1991; 254: 1001–3
67.
Nguyen T, Brunson D, Crespi CL, Penman BW, Wishnok JS, Tannenbaum SR: DNA damage and mutation in human cells exposed to nitric oxide in vitro. Proc Natl Acad Sci U S A 1992; 89: 3030–4
68.
Yoshie Y, Ohshima H: Nitric oxide synergistically enhances DNA strand breakage induced by polyhydroxyaromatic compounds, but inhibits that induced by the Fenton reaction. Arch Biochem Biophys 1997; 342: 13–21
69.
Pacelli R, Krishna MC, Wink DA, Mitchell JB: Nitric oxide protects DNA from hydrogen peroxide-induced double strand cleavage. Proc Am Assoc Cancer Res 1994; 35: 540
70.
Wink DA, Hanbauer I, Krishna MC, DeGraff W, Gamson J, Mitchell JB: Nitric oxide protects against cellular damage and cytotoxicity from reactive oxygen species. Proc Natl Acad Sci U S A 1993; 90: 9813–7
71.
Li LM, Kilbourn RG, Adams J, Fidler IJ: Role of nitric oxide in lysis of tumor cells by cytokine-activated endothelial cells. Cancer Res 1991; 51: 2531–5
72.
Yim CY, McGregor JR, Kwon OD, Bastian NR, Rees M, Mori M, Hibbs JB Jr, Samlowski WE: Nitric oxide synthesis contributes to IL-2-induced antitumor responses against intraperitoneal Meth A tumor. J Immunol 1995; 155: 4382–90
73.
Kurose I, Miura S, Fukumura D, Yonei Y, Saito H, Tada S, Suematsu M, Tsuchiya M: Nitric oxide mediates Kupffer cell-induced reduction of mitochondrial energization in hepatoma cells: A comparison with oxidative burst. Cancer Res 1993; 53: 2676–82
74.
Fukumura D, Yonei Y, Kurose I, Saito H, Ohishi T, Higuchi H, Miura S, Kato S, Kimura H, Ebinuma H, Ishi H: Role in nitric oxide in Kupffer cell-mediated hepatoma cell cytotoxicity in vitro and ex vivo. Hepatology 1996; 24: 141–9
75.
Curley SA, Roh MS, Feig B, Oyedeji C, Kleinerman ES, Klostergaard J: Mechanisms of Kupffer cell cytotoxicity in vitro against the syngeneic murine colon adenocarcinoma line MCA26. J Leukoc Biol 1993; 53: 715–21
76.
Klostergaard J, Leroux ME, Hung MC: Cellular models of macrophage tumoricidal effector mechanisms in vitro. Characterization of cytolytic responses to tumor necrosis factor and nitric oxide pathways in vitro. J Immunol 1991; 147: 2802–8
77.
Jiang H, Stewart CA, Fast DJ, Leu RW: Tumor target-derived soluble factor synergizes with IFN-gamma and IL-2 to activate macrophages for tumor necrosis factor and nitric oxide production to mediate cytotoxicity of the same target. J Immunol 1992; 149: 2137–46
78.
Leu RW, Leu NR, Shannon BJ, Fast DJ: IFN-gamma differentially modulates the susceptibility of L1210 and P815 tumor targets for macrophage-mediated cytotoxicity. Role of macrophage-target interaction coupled to nitric oxide generation, but independent of tumor necrosis factor production. J Immunol 1991; 147: 1816–22
79.
Xiao L, Eneroth PH, Qureshi GA: Nitric oxide synthase pathway may mediate human natural killer cell cytotoxicity. Scand J Immunol 1995; 42: 505–11
80.
Petit JF, Nicaise M, Lepoivre M, Guissani A, Lemaire G: Protection by glutathione against the antiproliferative effects of nitric oxide. Dependence on kinetics of NO release. Biochem Pharmacol 1996; 52: 205–12
81.
Pipili-Synetos E, Papageorgiou A, Sakkoula E, Sotiropoulou G, Fotsis T, Karakiulakis G, Maragoudakis ME: Inhibition of angiogenesis, tumour growth and metastasis by the NO-releasing vasodilators, isosorbide mononitrate and dinitrate. Br J Pharmacol 1995; 116: 1829–34
82.
Dong Z, Staroselsky AH, Qi X, Xie K, Fidler IJ: Inverse correlation between expression of inducible nitric oxide synthase activity and production of metastasis in K-1735 murine melanoma cells. Cancer Res 1994; 54: 789–93
83.
Kong L, Dunn GD, Keefer LK, Korthuis RJ: Nitric oxide reduces tumor cell adhesion to isolated rat postcapillary venules. Clin Exp Metastasis 1996; 14: 335–43
84.
Murata J, Ricciardi-Castagnoli P, Dessous L, Martin F, Juillerat-Jeanneret L: Microglial cells induce cytotoxic effects toward colon carcinoma cells: Measurement of tumor cytotoxicity with a gamma-glutamyl transpeptidase assay. Int J Cancer 1997; 70: 169–74
85.
Mordan LJ, Burnett TS, Zhang LX, Tom J, Cooney RV: Inhibitors of endogenous nitrogen oxide formation block the promotion of neoplastic transformation in C3H 10T1/2 fibroblasts. Carcinogenesis 1993; 14: 1555–9
86.
Gottke M, Chadee K: Exogenous nitric oxide stimulates mucin secretion from LS174T colonic adenocarcinoma cells. Inflamm Res 1996; 45: 209–12
87.
Rubbo H, Tarpey M, Freeman BA: Nitric oxide and reactive oxygen species in vascular injury. Biochem Soc Symp 1995; 61: 33–45
88.
Rubbo H, Parthasarathy S, Barnes S, Kirk M, Kalyanaraman B, Freeman BA: Nitric oxide inhibition of lipoxygenase-dependent liposome and low-density lipoprotein oxidation: termination of radical chain propagation reactions and formation of nitrogen-containing oxidized lipid derivatives. Arch Biochem Biophys 1995; 324: 15–25
89.
Hogg N, Kalyanaraman B, Joseph J, Struck A, Parthasarathy S: Inhibition of low-density lipoprotein oxidation by nitric oxide. Potential role in atherogenesis. FEBS Lett 1993; 334: 170–4
90.
Padmaja S, Huie RE: The reaction of nitric oxide with organic peroxyl radicals. Biochem Biophys Res Commun 1993; 195: 539–44
91.
Darley-Usmar VM, Hogg N, O'Leary VJ, Wilson MT, Moncada S: The simultaneous generation of superoxide and nitric oxide can initiate lipid peroxidation in human low density lipoprotein. Free Radic Res Commun 1992; 17: 9–20
92.
Rubbo H, Radi R, Trujillo M, Telleri R, Kalyanaraman B, Barnes S, Kirk M, Freeman BA: Nitric oxide regulation of superoxide and peroxynitrite-dependent lipid peroxidation. Formation of novel nitrogen-containing oxidized lipid derivatives. J Biol Chem 1994; 269: 26066–75
93.
Wang Y, Marsden PA: Nitric oxide synthases: Biochemical and molecular regulation. Curr Opin Nephrol Hypertens 1995; 4: 12–22
94.
Schmidt HH, Hofmann H, Schindler U, Shutenko ZS, Cunningham DD, Feelisch M: No NO from NO synthase. Proc Natl Acad Sci U S A 1996; 93: 14492–7
95.
Xia Y, Dawson VL, Dawson TM, Snyder SH, Zweier JL: Nitric oxide synthase generates superoxide and nitric oxide in arginine-depleted cells leading to peroxynitrite-mediated cellular injury. Proc Natl Acad Sci U S A 1996; 93: 6770–4
96.
Xia Y, Zweier JL: Superoxide and peroxynitrite generation from inducible nitric oxide synthase in macrophages. Proc Natl Acad Sci U S A 1997; 94: 6954–8
97.
Bredt DS, Hwang PM, Glatt CE, Lowenstein C, Reed RR, Snyder SH: Cloned and expressed nitric oxide synthase structurally resembles cytochrome P-450 reductase. Nature 1991; 351: 714–8
98.
McMillan K, Bredt DS, Hirsch DJ, Snyder SH, Clark JE, Masters BS: Cloned, expressed rat cerebellar nitric oxide synthase contains stoichiometric amounts of heme, which binds carbon monoxide. Proc Natl Acad Sci U S A 1992; 89: 11141–5
99.
Nishida K, Harrison DG, Navas JP, Fisher AA, Dockery SP, Uematsu M, Nerem RM, Alexander RW, Murphy TJ: Molecular cloning and characterization of the constitutive bovine aortic endothelial cell nitric oxide synthase. J Clin Invest 1992; 90: 2092–6
100.
Herdegen T, Brecht S, Mayer B, Leah J, Kummer W, Bravo R, Zimmermann M: Long-lasting expression of JUN and KROX transcription factors and nitric oxide synthase in intrinsic neurons of the rat brain following axotomy. J Neurosci 1993; 13: 4130–45
101.
Yoshizumi M, Perrella MA, Burnett JC Jr, Lee ME: Tumor necrosis factor downregulates an endothelial nitric oxide synthase mRNA by shortening its half-life. Circ Res 1993; 73: 205–9
102.
Ravichandran LV, Johns RA: Up-regulation of endothelial nitric oxide synthase expression by cyclic guanosine 3′,5′-monophosphate. FEBS Lett 1995; 374: 295–8
103.
Guo FH, De Raeve HR, Rice TW, Stuehr DJ, Thunnissen FB, Erzurum SC: Continuous nitric oxide synthesis by inducible nitric oxide synthase in normal human airway epithelium in vivo. Proc Natl Acad Sci U S A 1995; 92: 7809–13
104.
Sessa WC, Garcia-Cardena G, Liu J, Keh A, Pollock JS, Bradley J, Thiru S, Braverman IM, Desai KM: The Golgi association of endothelial nitric oxide synthase is necessary for the efficient synthesis of nitric oxide. J Biol Chem 1995; 270: 17641–4
105.
Michel T, Feron O: Nitric oxide synthases: Which, where, how, and why? J Clin Invest 1997; 100: 2146–52
106.
MacMicking J, Xie QW, Nathan C: Nitric oxide and macrophage function. Annu Rev Immunol 1997; 15: 323–50
107.
Cannon RO: Role of nitric oxide in cardiovascular disease: Focus on the endothelium. Clin Chem 1998; 44: 1809–19
108.
Miyasaka N, Hirata Y: Nitric oxide and inflammatory arthritides. Life Sci 1997; 61: 2073–81
109.
James SL: Role of nitric oxide in parasitic infections. Microbiol Rev 1995; 59: 533–47
110.
Samdani AF, Dawson TM, Dawson VL: Nitric oxide synthase in models of focal ischemia. Stroke 1997; 28: 1283–8
111.
Fukumura D, Jain RK: Role of nitric oxide in angiogenesis and microcirculation in tumors. Cancer Metastasis Rev 1998; 17: 77–89
112.
Xie K, Fidler IJ: Therapy of cancer metastasis by activation of the inducible nitric oxide synthase. Cancer Metastasis Rev 1998; 17: 55–75
113.
Albina JE, Reichner JS: Role of nitric oxide in mediation of macrophage cytotoxicity and apoptosis. Cancer Metastasis Rev 1998; 17: 39–53
114.
Christopherson KS, Bredt DS: Nitric oxide in excitable tissues: Physiological roles and disease. J Clin Invest 1997; 100: 2424–9
115.
Nathan C: Inducible nitric oxide synthase: What difference does it make? J Clin Invest 1997; 100: 2417–23
116.
Zweier JL, Wang P, Samouilov A, Kuppusamy P: Enzyme-independent formation of nitric oxide in biological tissues. Nat Med 1995; 1: 804–9
117.
Bredt DS, Hwang PM, Snyder SH: Localization of nitric oxide synthase indicating a neural role for nitric oxide. Nature 1990; 347: 768–70
118.
Schuman EM, Madison DV: Nitric oxide and synaptic function. Annu Rev Neurosci 1994; 17: 153–83
119.
Balligand JL, Cannon PJ: Nitric oxide synthases and cardiac muscle. Autocrine and paracrine influences. Arterioscler Thromb Vasc Biol 1997; 17: 1846–58
120.
Goldstein IM, Ostwald P, Roth S: Nitric oxide: A review of its role in retinal function and disease. Vision Res 1996; 36: 2979–94
121.
Vallance P, Moncada S: Role of endogenous nitric oxide in septic shock. New Horiz 1993; 1: 77–86
122.
Huang PL, Huang Z, Mashimo H, Bloch KD, Moskowitz MA, Bevan JA, Fishman MC: Hypertension in mice lacking the gene for endothelial nitric oxide synthase. Nature 1995; 377: 239–42
123.
Steudel W, Ichinose F, Huang PL, Hurford WE, Jones RC, Bevan JA, Fishman MC, Zapol WM: Pulmonary vasoconstriction and hypertension in mice with targeted disruption of the endothelial nitric oxide synthase (NOS 3) gene. Circ Res 1997; 81: 34–41
124.
Halbower AC, Tuder RM, Franklin WA, Pollock JS, Forstermann U, Abman SH: Maturation-related changes in endothelial nitric oxide synthase immunolocalization in developing ovine lung. Am J Physiol 1994; 267: L585–91
125.
Xue C, Reynolds PR, Johns RA: Developmental expression of NOS isoforms in fetal rat lung: Implications for transitional circulation and pulmonary angiogenesis. Am J Physiol 1996; 270: L88–100
126.
Corson MA, James NL, Latta SE, Nerem RM, Berk BC, Harrison DG: Phosphorylation of endothelial nitric oxide synthase in response to fluid shear stress. Circ Res 1996; 79: 984–91
127.
Nadaud S, Philippe M, Arnal JF, Michel JB, Soubrier F: Sustained increase in aortic endothelial nitric oxide synthase expression in vivo in a model of chronic high blood flow. Circ Res 1996; 79: 857–63
128.
Giaid A, Saleh D: Reduced expression of endothelial nitric oxide synthase in the lungs of patients with pulmonary hypertension. N Engl J Med 1995; 333: 214–21
129.
Steudel W, Scherrer-Crosbie M, Bloch KD, Weimann J, Huang PL, Jones RC, Picard MH, Zapol WM: Sustained pulmonary hypertension and right ventricular hypertrophy after chronic hypoxia in mice with congenital deficiency of nitric oxide synthase 3. J Clin Invest 1998; 101: 2468–77
130.
Ravichandran LV, Johns RA, Rengasamy A: Direct and reversible inhibition of endothelial nitric oxide synthase by nitric oxide. Am J Physiol 1995; 268: H2216–23
131.
DeRubertis FR, Craven PA: Calcium-independent modulation of cyclic GMP and activation of guanylate cyclase by nitrosamines. Science 1976; 193: 897–9
132.
Gruetter CA, Barry BK, McNamara DB, Gruetter DY, Kadowitz PJ, Ignarro L: Relaxation of bovine coronary artery and activation of coronary arterial guanylate cyclase by nitric oxide, nitroprusside and a carcinogenic nitrosoamine. J Cyclic Nucleotide Res 1979; 5: 211–24
133.
Ignarro LJ, Degnan JN, Baricos WH, Kadowitz PJ, Wolin MS: Activation of purified guanylate cyclase by nitric oxide requires heme. Comparison of heme-deficient, heme-reconstituted and heme- containing forms of soluble enzyme from bovine lung. Biochim Biophys Acta 1982; 718: 49–59
134.
McDonald LJ, Murad F: Nitric oxide and cyclic GMP signaling. Proc Soc Exp Biol Med 1996; 211: 1–6
135.
Hobbs AJ: Soluble guanylate cyclase: The forgotten sibling. Trends Pharmacol Sci 1997; 18: 484–91
136.
McMahon TJ, Hood JS, Bellan JA, Kadowitz PJ: N omega-nitro-L-arginine methyl ester selectively inhibits pulmonary vasodilator responses to acetylcholine and bradykinin. J Appl Physiol 1991; 71: 2026–31
137.
McMahon TJ, Ignarro LJ, Kadowitz PJ: Influence of Zaprinast on vascular tone and vasodilator responses in the cat pulmonary vascular bed. J Appl Physiol 1993; 74: 1704–11
138.
Pison U, Lopez FA, Heidelmeyer CF, Rossaint R, Falke KJ: Inhaled nitric oxide reverses hypoxic pulmonary vasoconstriction without impairing gas exchange. J Appl Physiol 1993; 74: 1287–92
139.
Frostell CG, Blomqvist H, Hedenstierna G, Lundberg J, Zapol WM: Inhaled nitric oxide selectively reverses human hypoxic pulmonary vasoconstriction without causing systemic vasodilation. A NESTHESIOLOGY 1993; 78: 427–35
140.
Scherrer U, Vollenweider L, Delabays A, Savcic M, Eichenberger U, Kleger GR, Fikrle A, Ballmer PE, Nicod P, Bartsch P: Inhaled nitric oxide for high-altitude pulmonary edema. N Engl J Med 1996; 334: 624–9
141.
Rimar S, Gillis CN: Selective pulmonary vasodilation by inhaled nitric oxide is due to hemoglobin inactivation. Circulation 1993; 88: 2884–7
142.
Dellinger RP, Zimmerman JL, Taylor RW, Straube RC, Hauser DL, Criner GJ, Davis KJ, Hyers TM, Papadakos P: Effects of inhaled nitric oxide in patients with acute respiratory distress syndrome: Results of a randomized phase II trial. Crit Care Med 1998; 26: 15–23
143.
Gerlach H, Pappert D, Lewandowski K, Rossaint R, Falke KJ: Long-term inhalation with evaluated low doses of nitric oxide for selective improvement of oxygenation in patients with adult respiratory distress syndrome. Intensive Care Med 1993; 19: 443–9
144.
Rossaint R, Falke KJ, Lopez F, Slama K, Pison U, Zapol WM: Inhaled nitric oxide for the adult respiratory distress syndrome. N Engl J Med 1993; 328: 399–405
145.
Rossaint R, Gerlach H, Schmidt-Ruhnke H, Pappert D, Lewandowski K, Steudel W, Falke K: Efficacy of inhaled nitric oxide in patients with severe ARDS. Chest 1995; 107: 1107–15
146.
Bigatello LM, Hurford WE, Kacmarek RM, Roberts JD Jr, Zapol WM: Prolonged inhalation of low concentrations of nitric oxide in patients with severe adult respiratory distress syndrome. Effects on pulmonary hemodynamics and oxygenation. A NESTHESIOLOGY 1994; 80: 761–70
147.
Manktelow C, Bigatello LM, Hess D, Hurford WE: Physiologic determinants of the response to inhaled nitric oxide in patients with acute respiratory distress syndrome. A NESTHESIOLOGY 1997; 87: 297–307
148.
Yoshimura K, Tod ML, Pier KG, Rubin LJ: Role of venoconstriction in thromboxane-induced pulmonary hypertension and edema in lambs. J Appl Physiol 1989; 66: 929–35
149.
Yoshimura K, Tod ML, Pier KG, Rubin LJ: Effects of a thromboxane A2analogue and prostacyclin on lung fluid balance in newborn lambs. Circ Res 1989; 65: 1409–16
150.
Lindeborg DM, Kavanagh BP, Van MK, Pearl RG: Inhaled nitric oxide does not alter the longitudinal distribution of pulmonary vascular resistance. J Appl Physiol 1995; 78: 341–8
151.
Shirai M, Shimouchi A, Kawaguchi AT, Sunagawa K, Ninomiya I: Inhaled nitric oxide: Diameter response patterns in feline small pulmonary arteries and veins. Am J Physiol 1996; 270: H974–80
152.
Nunn JF: Distribution of pulmoanry ventilation and perfusion, Nunn's Applied Respiratory Physiology. Edited by Nunn JF. Oxford, Butterworth-Heinemann, 1993, pp 156–97
153.
Putensen C, Rasanen J, Downs JB: Effect of endogenous and inhaled nitric oxide on the ventilation-perfusion relationships in oleic-acid lung injury. Am J Respir Crit Care Med 1994; 150: 330–6
154.
Putensen C, Rasanen J, Lopez FA, Downs JB: Continuous positive airway pressure modulates effect of inhaled nitric oxide on the ventilation-perfusion distributions in canine lung injury. Chest 1994; 106: 1563–9
155.
Hopkins SR, Johnson EC, Richardson RS, Wagner H, De RM, Wagner PD: Effects of inhaled nitric oxide on gas exchange in lungs with shunt or poorly ventilated areas. Am J Respir Crit Care Med 1997; 156: 484–91
156.
Belvisi MG, Stretton CD, Miura M, Verleden GM, Tadjkarimi S, Yacoub MH, Barnes PJ: Inhibitory NANC nerves in human tracheal smooth muscle: A quest for the neurotransmitter. J Appl Physiol 1992; 73: 2505–10
157.
Belvisi MG, Ward JK, Mitchell JA, Barnes PJ: Nitric oxide as a neurotransmitter in human airways. Arch Int Pharmacodyn Ther 1995; 329: 97–110
158.
Dey RD, Mayer B, Said SI: Colocalization of vasoactive intestinal peptide and nitric oxide synthase in neurons of the ferret trachea. Neuroscience 1993; 54: 839–43
159.
Watkins DN, Peroni DJ, Basclain KA, Garlepp MJ, Thompson PJ: Expression and activity of nitric oxide synthases in human airway epithelium. Am J Respir Cell Mol Biol 1997; 16: 629–39
160.
Dupuy PM, Shore SA, Drazen JM, Frostell C, Hill WA, Zapol WM: Bronchodilator action of inhaled nitric oxide in guinea pigs. J Clin Invest 1992; 90: 421–8
161.
Högman M, Wei SZ, Frostell C, Arnberg H, Hedenstierna G: Effects of inhaled nitric oxide on methacholine-induced bronchoconstriction: A concentration response study in rabbits. Eur Respir J 1994; 7: 698–702
162.
Högman M, Frostell C, Arnberg H, Hedenstierna G: Inhalation of nitric oxide modulates methacholine-induced bronchoconstriction in the rabbit. Eur Respir J 1993; 6: 177–80
163.
Brown RH, Zerhouni EA, Hirshman CA: Reversal of bronchoconstriction by inhaled nitric oxide. Histamine versus methacholine. Am J Respir Crit Care Med 1994; 150: 233–7
164.
Lindeman KS, Aryana A, Hirshman CA: Direct effects of inhaled nitric oxide on canine peripheral airways. J Appl Physiol 1995; 78: 1898–903
165.
Gwyn DR, Lindeman KS, Hirshman CA: Inhaled nitric oxide attenuates bronchoconstriction in canine peripheral airways. Am J Respir Crit Care Med 1996; 153: 604–9
166.
Putensen C, Rasanen J, Lopez FA: Improvement in VA/Q distributions during inhalation of nitric oxide in pigs with methacholine-induced bronchoconstriction. Am J Respir Crit Care Med 1995; 151: 116–22
167.
Albertini M, Clement MG: Inhaled nitric oxide reverses PAF-dependent bronchoconstriction in the pig. Prostaglandins Leukot Essent Fatty Acids 1995; 52: 373–80
168.
Kacmarek RM, Ripple R, Cockrill BA, Bloch KJ, Zapol WM, Johnson DC: Inhaled nitric oxide. A bronchodilator in mild asthmatics with methacholine-induced bronchospasm. Am J Respir Crit Care Med 1996; 153: 128–35
169.
Högman M, Frostell CG, Hedenstrom H, Hedenstierna G: Inhalation of nitric oxide modulates adult human bronchial tone. Am Rev Respir Dis 1993; 148: 1474–8
170.
Haddad IY, Zhu S, Crow J, Barefield E, Gadilhe T, Matalon S: Inhibition of alveolar type II cell ATP and surfactant synthesis by nitric oxide. Am J Physiol 1996; 270: L898–906
171.
Matalon S, DeMarco V, Haddad IY, Myles C, Skimming JW, Schurch S, Cheng S, Cassin S: Inhaled nitric oxide injures the pulmonary surfactant system of lambs in vivo. Am J Physiol 1996; 270: L273–80
172.
Ayad O, Wong HR: Nitric oxide decreases surfactant protein A gene expression in H441 cells. Crit Care Med 1998; 26: 1277–82
173.
Zhu GF, Sun B, Niu SF, Cai YY, Lin K, Lindwall R, Robertson B: Combined surfactant therapy and inhaled nitric oxide in rabbits with oleic acid-induced acute respiratory distress syndrome. Am J Respir Crit Care Med 1998; 158: 437–43
174.
Young JD, Sear JW, Valvini EM: Kinetics of methaemoglobin and serum nitrogen oxide production during inhalation of nitric oxide in volunteers. Br J Anaesth 1996; 76: 652–6
175.
Yoshida K, Kasama K: Biotransformation of nitric oxide. Environ Health Perspect 1987; 73: 201–5
176.
Yoshida K, Kasama K, Kitabatake M, Imai M: Biotransformation of nitric oxide, nitrite and nitrate. Int Arch Occup Environ Health 1983; 52: 103–15
177.
Roberts JD Jr, Chen TY, Kawai N, Wain J, Dupuy P, Shimouchi A, Bloch K, Polaner D, Zapol WM: Inhaled nitric oxide reverses pulmonary vasoconstriction in the hypoxic and acidotic newborn lamb. Circ Res 1993; 72: 246–54
178.
Kinsella JP, McQueston JA, Rosenberg AA, Abman SH: Hemodynamic effects of exogenous nitric oxide in ovine transitional pulmonary circulation. Am J Physiol 1992; 263: H875–80
179.
Zayek M, Cleveland D, Morin FC: Treatment of persistent pulmonary hypertension in the newborn lamb by inhaled nitric oxide. J Pediatr 1993; 122: 743–50
180.
Zayek M, Wild L, Roberts JD, Morin FC III: Effect of nitric oxide on the survival rate and incidence of lung injury in newborn lambs with persistent pulmonary hypertension. J Pediatr 1993; 123: 947–52
181.
Kinsella JP, Ivy DD, Abman SH: Inhaled nitric oxide improves gas exchange and lowers pulmonary vascular resistance in severe experimental hyaline membrane disease. Pediatr Res 1994; 36: 402–8
182.
Kinsella JP, Ivy DD, Abman SH: Ontogeny of NO activity and response to inhaled NO in the developing ovine pulmonary circulation. Am J Physiol 1994; 267: H1955–61
183.
Fratacci MD, Frostell CG, Chen TY, Wain JC Jr, Robinson DR, Zapol WM: Inhaled nitric oxide. A selective pulmonary vasodilator of heparin-protamine vasoconstriction in sheep. A NESTHESIOLOGY 1991; 75: 990–9
184.
Shah NS, Nakayama DK, Jacob TD, Nishio I, Imai T, Billiar TR, Exler R, Yousem SA, Motoyama EK, Peitzman AB: Efficacy of inhaled nitric oxide in a porcine model of adult respiratory distress syndrome. Arch Surg 1994; 129: 158–64
185.
Shah NS, Nakayama DK, Jacob TD, Nishio I, Imai T, Billiar TR, Exler R, Yousem SA, Motoyama EK, Peitzman AB: Efficacy of inhaled nitric oxide in oleic acid-induced acute lung injury. Crit Care Med 1997; 25: 153–8
186.
Rovira I, Chen TY, Winkler M, Kawai N, Bloch KD, Zapol WM: Effects of inhaled nitric oxide on pulmonary hemodynamics and gas exchange in an ovine model of ARDS. J Appl Physiol 1994; 76: 345–55
187.
Weitzberg E, Rudehill A, Lundberg JM: Nitric oxide inhalation attenuates pulmonary hypertension and improves gas exchange in endotoxin shock. Eur J Pharmacol 1993; 233: 85–94
188.
Ogura H, Cioffi WG, Offner PJ, Jordan BS, Johnson AA, Pruitt BA Jr: Effect of inhaled nitric oxide on pulmonary function after sepsis in a swine model. Surgery 1994; 116: 313–21
189.
Dahm P, Blomquist S, Martensson L, Thorne J, Zoucas E: Circulatory and ventilatory effects of intermittent nitric oxide inhalation during porcine endotoxemia. J Trauma 1994; 37: 769–77
190.
Ogura H, Cioffi WG Jr, Jordan BS, Okerberg CV, Johnson AA, Mason AD Jr, Pruitt BA Jr: The effect of inhaled nitric oxide on smoke inhalation injury in an ovine model. J Trauma 1994; 37: 294–301
191.
Gadek JE: Adverse effects of neutrophils on the lung. Am J Med 1992; 92: 27S–31S
192.
Swank DW, Moore SB: Roles of the neutrophil and other mediators in adult respiratory distress syndrome. Mayo Clin Proc 1989; 64: 1118–32
193.
Strieter RM, Kunkel SL: Acute lung injury: The role of cytokines in the elicitation of neutrophils. J Investig Med 1994; 42: 640–51
194.
Wortel CH, Doerschuk CM: Neutrophils and neutrophil-endothelial cell adhesion in adult respiratory distress syndrome. New Horiz 1993; 1: 631–7
195.
Jones R, Zapol WM, Reid L: Oxygen toxicity and restructuring of pulmonary arteries—A morphometric study. The response to 4 weeks’ exposure to hyperoxia and return to breathing air. Am J Pathol 1985; 121: 212–23
196.
Jones R, Adler C, Farber F: Lung vascular cell proliferation in hyperoxic pulmonary hypertension and on return to air:[3H]Thymidine pulse-labeling of intimal, medial, and adventitial cells in microvessels and at the hilum. Am Rev Respir Dis 1989; 140: 1471–7
197.
Wink DA, Cook JA, Pacelli R, Liebmann J, Krishna MC, Mitchell JB: Nitric oxide (NO) protects against cellular damage by reactive oxygen species. Toxicol Lett 1995; 82–83, 221–6
198.
Poss WB, Timmons OD, Farrukh IS, Hoidal JR, Michael JR: Inhaled nitric oxide prevents the increase in pulmonary vascular permeability caused by hydrogen peroxide. J Appl Physiol 1995; 79: 886–91
199.
Kavanagh BP, Mouchawar A, Goldsmith J, Pearl RG: Effects of inhaled NO and inhibition of endogenous NO synthesis in oxidant-induced acute lung injury. J Appl Physiol 1994; 76: 1324–9
200.
Garat C, Jayr C, Eddahibi S, Laffon M, Meignan M, Adnot S: Effects of inhaled nitric oxide or inhibition of endogenous nitric oxide formation on hyperoxic lung injury. Am J Respir Crit Care Med 1997; 155: 1957–64
201.
McElroy MC, Wiener-Kronish JP, Miyazaki H, Sawa T, Modelska K, Dobbs LG, Pittet JF: Nitric oxide attenuates lung endothelial injury caused by sublethal hyperoxia in rats. Am J Physiol 1997; 272: L631–8
202.
Narula P, Xu J, Kazzaz JA, Robbins CG, Davis JM, Horowitz S: Synergistic cytotoxicity from nitric oxide and hyperoxia in cultured lung cells. Am J Physiol 1998; 274: L411–6
203.
Gadek JE: Adverse effects of neutrophils on the lung. Am J Med 1992; 92: 27S–31S
204.
Strieter RM, Kunkel SL: Acute lung injury: The role of cytokines in the elicitation of neutrophils. J Investig Med 1994; 42: 640–51
205.
Fullerton DA, Eisenach JH, McIntyre RC Jr, Friese RS, Sheridan BC, Roe GB, Agrafojo J, Banerjee A, Harken AH: Inhaled nitric oxide prevents pulmonary endothelial dysfunction after mesenteric ischemia-reperfusion. Am J Physiol 1996; 271: L326–31
206.
Malmros C, Blomquist S, Dahm P, Martensson L, Thorne J: Nitric oxide inhalation decreases pulmonary platelet and neutrophil sequestration during extracorporeal circulation in the pig. Crit Care Med 1996; 24: 845–9
207.
Guidot DM, Repine MJ, Hybertson BM, Repine JE: Inhaled nitric oxide prevents neutrophil-mediated, oxygen radical-dependent leak in isolated rat lungs. Am J Physiol 1995; 269: L2–5
208.
Guidot DM, Hybertson BM, Kitlowski RP, Repine JE: Inhaled NO prevents IL-1-induced neutrophil accumulation and associated acute edema in isolated rat lungs. Am J Physiol 1996; 271: L225–9
209.
Kinsella JP, Parker TA, Galan H, Sheridan BC, Halbower AC, Abman SH: Effects of inhaled nitric oxide on pulmonary edema and lung neutrophil accumulation in severe experimental hyaline membrane disease. Pediatr Res 1997; 41: 457–63
210.
Bloomfield GL, Holloway S, Ridings PC, Fisher BJ, Blocher CR, Sholley M, Bunch T, Sugerman HJ, Fowler AA: Pretreatment with inhaled nitric oxide inhibits neutrophil migration and oxidative activity resulting in attenuated sepsis-induced acute lung injury. Crit Care Med 1997; 25: 584–93
211.
Weinberger B, Fakhrzadeh L, Heck DE, Laskin JD, Gardner CR, Laskin DL: Inhaled nitric oxide primes lung macrophages to produce reactive oxygen and nitrogen intermediates. Am J Respir Crit Care Med 1998; 158: 931–8
212.
Kermarrec N, Chollet-Martin S, Beloucif S, Faivre V, Gougerot-Pocidalo MA, Payen DM: Alveolar neutrophil oxidative burst and β2integrin expression in experimental acute pulmonary inflammation are not modified by inhaled nitric oxide. Shock 1998; 10: 129–34
213.
Biffl WL, Moore EE, Moore FA, Barnett C: Nitric oxide reduces endothelial expression of intercellular adhesion molecule (ICAM)-1. J Surg Res 1996; 63: 328–32
214.
Martelletti P, Stirparo G, Morrone S, Rinaldi C, Giacovazzo M: Inhibition of intercellular adhesion molecule-1 (ICAM-1), soluble ICAM-1 and interleukin-4 by nitric oxide expression in migraine patients. J Mol Med 1997; 75: 448–53
215.
Kanwar S, Kubes P: Nitric oxide is an antiadhesive molecule for leukocytes. New Horiz 1995; 3: 93–104
216.
Lefer AM, Lefer DJ: The role of nitric oxide and cell adhesion molecules on the microcirculation in ischaemia-reperfusion. Cardiovasc Res 1996; 32: 743–51
217.
DeCaterina R, Libby P, Peng HB, Thannickal VJ, Rajavashisth TB, Gimbrone MAJ, Shin WS, Liao JK: Nitric oxide decreases cytokine-induced endothelial activation. Nitric oxide selectively reduces endothelial expression of adhesion molecules and proinflammatory cytokines. J Clin Invest 1995; 96: 60–8
218.
Clancy RM, Leszczynska-Piziak J, Abramson SB: Nitric oxide, an endothelial cell relaxation factor, inhibits neutrophil superoxide anion production via a direct action on the NADPH oxidase. J Clin Invest 1992; 90: 1116–21
219.
Gaboury J, Woodman RC, Granger DN, Reinhardt P, Kubes P: Nitric oxide prevents leukocyte adherence: Role of superoxide. Am J Physiol 1993; 265: H862–7
220.
Fortenberry JD, Owens ML, Brown MR, Atkinson D, Brown LA: Exogenous nitric oxide enhances neutrophil cell death and DNA fragmentation. Am J Respir Cell Mol Biol 1998; 18: 421–8
221.
Meyrick B, Reid L: The effect of continued hypoxia on rat pulmonary arterial circulation. An ultrastructural study. Lab Invest 1978; 38: 188–200
222.
Rabinovitch M, Fisher K, Gamble W, Reid L, Treves S: Thallium-201: quantitation of right ventricular hypertrophy in chronically hypoxic rats. Radiology 1979; 130: 223–5
223.
Kouyoumdjian C, Adnot S, Levame M, Eddahibi S, Bousbaa H, Raffestin B: Continuous inhalation of nitric oxide protects against development of pulmonary hypertension in chronically hypoxic rats. J Clin Invest 1994; 94: 578–84
224.
Roos CM, Frank DU, Xue C, Johns RA, Rich GF: Chronic inhaled nitric oxide: Effects on pulmonary vascular endothelial function and pathology in rats. J Appl Physiol 1996; 80: 252–60
225.
Roberts JD Jr, Roberts CT, Jones RC, Zapol WM, Bloch KD: Continuous nitric oxide inhalation reduces pulmonary arterial structural changes, right ventricular hypertrophy, and growth retardation in the hypoxic newborn rat. Circ Res 1995; 76: 215–22
226.
Cornwell TL, Arnold E, Boerth NJ, Lincoln TM: Inhibition of smooth muscle cell growth by nitric oxide and activation of cAMP-dependent protein kinase by cGMP. Annu Rev Immunol 1994; 267: C1405–13
227.
Gries A, Bottiger BW, Dorsam J, Bauer H, Weimann J, Bode C, Martin E, Motsch J: Inhaled nitric oxide inhibits platelet aggregation after pulmonary embolism in pigs. A NESTHESIOLOGY 1997; 86: 387–93
228.
Bacha EA, Herve P, Murakami S, Chapelier A, Mazmanian GM, de Montpreville V, Detruit H, Libert JM, Dartevelle P: Lasting beneficial effect of short-term inhaled nitric oxide on graft function after lung transplantation. J Thorac Cardiovasc Surg 1996; 112: 590–8
229.
Barbotin-Larrieu F, Mazmanian M, Baudet B, Detruit H, Chapelier A, Libert JM, Dartevelle P, Herve P: Prevention of ischemia-reperfusion lung injury by inhaled nitric oxide in neonatal piglets. J Appl Physiol 1996; 80: 782–8
230.
Bhabra MS, Hopkinson DN, Shaw TE, Hooper TL: Attenuation of lung graft reperfusion injury by a nitric oxide donor. J Thorac Cardiovasc Surg 1997; 113: 327–33
231.
Bhabra MS, Hopkinson DN, Shaw TE, Hooper TL: Low-dose nitric oxide inhalation during initial reperfusion enhances rat lung graft function. Ann Thorac Surg 1997; 63: 339–44
232.
Chetham PM, Sefton WD, Bridges JP, Stevens T, McMurtry IF: Inhaled nitric oxide pretreatment but not posttreatment attenuates ischemia-reperfusion-induced pulmonary microvascular leak. A NESTHESIOLOGY 1997; 86: 895–902
233.
Grover FL, Fullerton DA, Zamora MR, Mills C, Ackerman B, Badesch D, Brown JM, Campbell DN, Chetham P, Dhaliwal A, Diercks M, Kinnard T, Niejadlik K, Ochs M: The past, present, and future of lung transplantation. Am J Surg 1997; 173: 523–33
234.
Davis RDJ, Pasque MK: Pulmonary transplantation. Ann Surg 1995; 221: 14–28
235.
Egan TM, Lambert CJJ, Reddick R, Ulicny KSJ, Keagy BA, Wilcox BR: A strategy to increase the donor pool: Use of cadaver lungs for transplantation. Ann Thorac Surg 1991; 52: 1113–20
236.
Steen S, Ingemansson R, Budrikis A, Bolys R, Roscher R, Sjoberg T: Successful transplantation of lungs topically cooled in the non-heart-beating donor for 6 hours. Ann Thorac Surg 1997; 63: 345–51
237.
Bacha EA, Sellak H, Murakami S, Mazmanian GM, Detruit H, de Montpreville V, Chapelier AR, Libert JM, Dartevelle PG, Herve P: Inhaled nitric oxide attenuates reperfusion injury in non- heartbeating-donor lung transplantation. Transplantation 1997; 63: 1380–6
238.
Murakami S, Bacha EA, Herve P, Detruit H, Chapelier AR, Dartevelle PG, Mazmanian GM: Prevention of reperfusion injury by inhaled nitric oxide in lungs harvested from non-heart-beating donors. Ann Thorac Surg 1996; 62: 1632–8
239.
Högman M, Frostell C, Arnberg H, Sandhagen B, Hedenstierna G: Prolonged bleeding time during nitric oxide inhalation in the rabbit. Acta Physiol Scand 1994; 151: 125–9
240.
Lee JS, Adrie C, Jacob HJ, Roberts JD Jr, Zapol WM, Bloch KD: Chronic inhalation of nitric oxide inhibits neointimal formation after balloon-induced arterial injury. Circ Res 1996; 78: 337–42
241.
Steinberg MH: Review: Sickle cell disease: Present and future treatment. Am J Med Sci 1996; 312: 166–74
242.
Steinberg MH: Determinants of fetal hemoglobin response to hydroxyurea. Semin Hematol 1997; 34: 8–14
243.
Charache S: Mechanism of action of hydroxyurea in the management of sickle cell anemia in adults. Semin Hematol 1997; 34: 15–21
244.
Garel MC, Domenget C, Caburi-Martin J, Prehu C, Galacteros F, Beuzard Y: Covalent binding of glutathione to hemoglobin: I. Inhibition of hemoglobin S polymerization. J Biol Chem 1986; 261: 14704–9
245.
Schechter AN: NO therapy? J Clin Invest 1997; 100: 955–6
246.
Roberts JD Jr: Inhaled nitric oxide for treatment of pulmonary artery hypertension in the newborn and infant. Crit Care Med 1993; 21: S374–6
247.
Roberts JD, Polaner DM, Lang P, Zapol WM: Inhaled nitric oxide in persistent pulmonary hypertension of the newborn. Lancet 1992; 340: 818–9
248.
Kinsella JP, Neish SR, Shaffer E, Abman SH: Low-dose inhalation nitric oxide in persistent pulmonary hypertension of the newborn. Lancet 1992; 340: 819–20
249.
Neonatal Inhaled Nitric Oxide Study Group: Inhaled nitric oxide and hypoxic respiratory failure in infants with congenital diaphragmatic hernia. Pediatrics 1997; 99: 838–45
250.
Hoffman GM, Ross GA, Day SE, Rice TB, Nelin LD: Inhaled nitric oxide reduces the utilization of extracorporeal membrane oxygenation in persistent pulmonary hypertension of the newborn. Crit Care Med 1997; 25: 352–9
251.
Rosenberg AA, Kennaugh JM, Moreland SG, Fashaw LM, Hale KA, Torielli FM, Abman SH, Kinsella JP: Longitudinal follow-up of a cohort of newborn infants treated with inhaled nitric oxide for persistent pulmonary hypertension. J Pediatr 1997; 131: 70–5
252.
Abman SH, Kinsella JP, Schaffer MS, Wilkening RB: Inhaled nitric oxide in the management of a premature newborn with severe respiratory distress and pulmonary hypertension. Pediatrics 1993; 92: 606–9
253.
Peliowski A, Finer NN, Etches PC, Tierney AJ, Ryan CA: Inhaled nitric oxide for premature infants after prolonged rupture of the membranes. J Pediatr 1995; 126: 450–3
254.
Skimming JW, Bender KA, Hutchison AA, Drummond WH: Nitric oxide inhalation in infants with respiratory distress syndrome. J Pediatr 1997; 130: 225–30
255.
Rossaint R, Slama K, Steudel W, Gerlach H, Pappert D, Veit S, Falke K: Effects of inhaled nitric oxide on right ventricular function in severe acute respiratory distress syndrome. Intensive Care Med 1995; 21: 197–203
256.
Abman SH, Griebel JL, Parker DK, Schmidt JM, Swanton D, Kinsella JP: Acute effects of inhaled nitric oxide in children with severe hypoxemic respiratory failure. J Pediatr 1994; 124: 881–8
257.
Puybasset L, Stewart T, Rouby JJ, Cluzel P, Mourgeon E, Belin MF, Arthaud M, Landault C, Viars P: Inhaled nitric oxide reverses the increase in pulmonary vascular resistance induced by permissive hypercapnia in patients with acute respiratory distress syndrome. A NESTHESIOLOGY 1994; 80: 1254–67
258.
Benzing A, Geiger K: Inhaled nitric oxide lowers pulmonary capillary pressure and changes longitudinal distribution of pulmonary vascular resistance in patients with acute lung injury. Acta Anaesthesiol Scand 1994; 38: 640–5
259.
Benzing A, Bräutigam P, Geiger K, Loop T, Beyer U, Moser E: Inhaled nitric oxide reduces pulmonary transvascular albumin flux in patients with acute lung injury. A NESTHESIOLOGY 1995; 83: 1153–61
260.
Gerlach H, Rossaint R, Pappert D, Falke KJ: Time-course and dose-response of nitric oxide inhalation for systemic oxygenation and pulmonary hypertension in patients with adult respiratory distress syndrome. Eur J Clin Invest 1993; 23: 499–502
261.
Michael JR, Barton RG, Saffle JR, Mone M, Markewitz BA, Hillier K, Elstad MR, Campell EJ, Troyer BE, Whatley RE, Liou TG, Samuelson WM, Carveth HJ, Hinson DM, Morris SE, Davis BL, Day RW: Inhaled nitric oxide versus conventional therapy. Effects on oxygenation in ARDS. Am J Respir Crit Care Med 1998; 157: 1372–80
262.
Troncy E, Collet JP, Shapiro S, Guimond JG, Blair L, Ducruet T, Francoeur M, Charbonneau M, Blaise G: Inhaled nitric oxide in acute respiratory distress syndrome. Am J Respir Crit Care Med 1998; 157: 1483–8
263.
Montgomery AB, Stager MA, Carrico CJ, Hudson LD: Causes of mortality in patients with the adult respiratory distress syndrome. Am Rev Respir Dis 1985; 132: 485–9
264.
Suchyta MR, Clemmer TP, Elliott CG, Orme JFJ, Weaver LK: The adult respiratory distress syndrome. A report of survival and modifying factors. Chest 1992; 101: 1074–9
265.
Gerlach M, Keh D, Gerlach H: Inhaled nitric oxide for the acute respiratory distress syndrome. Respir Care 1999; (in press)
266.
Meyrick B, Reid L: Pulmonary hypertension. Anatomic and physiologic correlates. Clin Chest Med 1983; 4: 199–217
267.
Kneussl MP, Lang IM, Brenot FP: Medical management of primary pulmonary hypertension. Eur Respir J 1996; 9: 2401–9
268.
Sitbon O, Brenot F, Denjean A, Bergeron A, Parent F, Azarian R, Herve P, Raffestin B, Simonneau G: Inhaled nitric oxide as a screening vasodilator agent in primary pulmonary hypertension. A dose-response study and comparison with prostacyclin. Am J Respir Crit Care Med 1995; 151: 384–9
269.
Robbins IM, Christman BW, Newman JH, Matlock R, Loyd JE: A survey of diagnostic practices and the use of epoprostenol in patients with primary pulmonary hypertension. Chest 1998; 114: 1269–75
270.
Adatia I, Perry S, Landzberg M, Moore P, Thompson JE, Wessel DL: Inhaled nitric oxide and hemodynamic evaluation of patients with pulmonary hypertension before transplantation. J Am Coll Cardiol 1995; 25: 1656–64
271.
Channick RN, Newhart JW, Johnson FW, Williams PJ, Auger WR, Fedullo PF, Moser KM: Pulsed delivery of inhaled nitric oxide to patients with primary pulmonary hypertension: An ambulatory delivery system and initial clinical tests. Chest 1996; 109: 1545–9
272.
Goldman AP, Rees PG, Macrae DJ: Is it time to consider domiciliary nitric oxide? Lancet 1995; 345: 199–200
273.
Sanna A, Kurtansky A, Veriter C, Stanescu D: Bronchodilator effect of inhaled nitric oxide in healthy men. Am J Respir Crit Care Med 1994; 150: 1702–4
274.
Pfeffer KD, Ellison G, Robertson D, Day RW: The effect of inhaled nitric oxide in pediatric asthma. Am J Respir Crit Care Med 1996; 153: 747–51
275.
Katayama Y, Higenbottam TW, Diaz de Atauri MJ, Cremona G, Akamine S, Barbera JA, Rodriguez-Roisin R: Inhaled nitric oxide and arterial oxygen tension in patients with chronic obstructive pulmonary disease and severe pulmonary hypertension. Thorax 1997; 52: 120–4
276.
Barbera JA, Roger N, Roca J, Rovira I, Higenbottam TW, Rodriguez-Roisin R: Worsening of pulmonary gas exchange with nitric oxide inhalation in chronic obstructive pulmonary disease. Lancet 1996; 347: 436–40
277.
Yoshida M, Taguchi O, Gabazza EC, Kobayashi T, Yamakami T, Kobayashi H, Maruyama K, Shima T: Combined inhalation of nitric oxide and oxygen in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 1997; 155: 526–9
278.
Adatia I, Lillehei C, Arnold JH, Thompson JE, Palazzo R, Fackler JC, Wessel DL: Inhaled nitric oxide in the treatment of postoperative graft dysfunction after lung transplantation. Ann Thorac Surg 1994; 57: 1311–8
279.
Date H, Triantafillou AN, Trulock EP, Pohl MS, Cooper JD, Patterson GA: Inhaled nitric oxide reduces human lung allograft dysfunction. J Thorac Cardiovasc Surg 1996; 111: 913–9
280.
Haworth SG: Pulmonary vascular disease in different types of congenital heart disease. Implications for interpretation of lung biopsy findings in early childhood. Br Heart J 1984; 52: 557–71
281.
Roberts JD Jr, Lang P, Bigatello LM, Vlahakes GJ, Zapol WM: Inhaled nitric oxide in congenital heart disease. Circulation 1993; 87: 447–53
282.
Allman KG, Young JD, Carapiet D, Stevens JE, Ostman-Smith I, Archer LN: Effects of oxygen and nitric oxide in oxygen on pulmonary arterial pressures of children with congenital cardiac defects. Pediatr Cardiol 1996; 17: 246–50
283.
Berner M, Beghetti M, Spahr-Schopfer I, Oberhansli I, Friedli B: Inhaled nitric oxide to test the vasodilator capacity of the pulmonary vascular bed in children with long-standing pulmonary hypertension and congenital heart disease. Am J Cardiol 1996; 77: 532–5
284.
Roze JC, Storme L, Zupan V, Morville P, Dinh-Xuan AT, Mercier JC: Echocardiographic investigation of inhaled nitric oxide in newborn babies with severe hypoxaemia. Lancet 1994; 344: 303–5
285.
Wessel DL, Adatia I, Giglia TM, Thompson JE, Kulik TJ: Use of inhaled nitric oxide and acetylcholine in the evaluation of pulmonary hypertension and endothelial function after cardiopulmonary bypass. Circulation 1993; 88: 2128–38
286.
Journois D, Pouard P, Mauriat P, Malhere T, Vouhe P, Safran D: Inhaled nitric oxide as a therapy for pulmonary hypertension after operations for congenital heart defects. J Thorac Cardiovasc Surg 1994; 107: 1129–35
287.
Miller OI, Celermajer DS, Deanfield JE, Macrae DJ: Very-low-dose inhaled nitric oxide: A selective pulmonary vasodilator after operations for congenital heart disease. J Thorac Cardiovasc Surg 1994; 108: 487–94
288.
Curran RD, Mavroudis C, Backer CL, Sautel M, Zales VR, Wessel DL: Inhaled nitric oxide for children with congenital heart disease and pulmonary hypertension. Ann Thorac Surg 1995; 60: 1765–71
289.
Matsui J, Yahagi N, Kumon K, Hayashi H, Watanabe Y, Haruna M, Tanigami H, Yagihara T, Takamoto S, Kamiya T: Effects of inhaled nitric oxide on postoperative pulmonary circulation in patients with congenital heart disease. Artif Organs 1997; 21: 17–20
290.
Shimpo H, Mitani Y, Tanaka J, Mizumoto T, Onoda K, Tani K, Yuasa H, Yada I, Maruyama K: Inhaled low-dose nitric oxide for postoperative care in patients with congenital heart defects. Artif Organs 1997; 21: 10–3
291.
Goldman AP, Delius RE, Deanfield JE, Miller OI, de Leval MR, Sigston PE, Macrae DJ: Pharmacological control of pulmonary blood flow with inhaled nitric oxide after the fenestrated Fontan operation. Circulation 1996; 94: S44–8
292.
Goldman AP, Delius RE, Deanfield JE, de Leval MR, Sigston PE, Macrae DJ: Nitric oxide might reduce the need for extracorporeal support in children with critical postoperative pulmonary hypertension. Ann Thorac Surg 1996; 62: 750–5
293.
Fullerton DA, Jones SD, Jaggers J, Piedalue F, Grover FL, McIntyre RC Jr: Effective control of pulmonary vascular resistance with inhaled nitric oxide after cardiac operation. J Thorac Cardiovasc Surg 1996; 111: 753–62
294.
Auler Junior JO, Carmona MJ, Bocchi EA, Bacal F, Fiorelli AI, Stolf NA, Jatene AD: Low doses of inhaled nitric oxide in heart transplant recipients. J Heart Lung Transplant 1996; 15: 443–50
295.
Fullerton DA, Jaggers J, Piedalue F, Grover FL, McIntyre RC Jr: Effective control of refractory pulmonary hypertension after cardiac operations. J Thorac Cardiovasc Surg 1997; 113: 363–8
296.
Krafft P, Fridrich P, Fitzgerald RD, Koc D, Steltzer H: Effectiveness of nitric oxide inhalation in septic ARDS. Chest 1996; 109: 486–93
297.
Holzmann A, Bloch KD, Sanchez LS, Filippov G, Zapol WM: Hyporesponsiveness to inhaled nitric oxide in isolated, perfused lungs from endotoxin-challenged rats. Am J Physiol 1996; 271: L981–6
298.
Bigatello LM, Hess D, Bloch KD, Ritz R, Hurford WE: Effect of inhaled nitric oxide on plasma cyclic GMP in ARDS patients (abstract). Am J Respir Crit Care Med 1998; 157: A683
299.
Munzel T, Kurz S, Rajagopalan S, Thoenes M, Berrington WR, Thompson JA, Freeman BA, Harrison DG: Hydralazine prevents nitroglycerin tolerance by inhibiting activation of a membrane-bound NADH oxidase. A new action for an old drug. J Clin Invest 1996; 98: 1465–70
300.
Munzel T, Sayegh H, Freeman BA, Tarpey MM, Harrison DG: Evidence for enhanced vascular superoxide anion production in nitrate tolerance. A novel mechanism underlying tolerance and cross- tolerance. J Clin Invest 1995; 95: 187–94
301.
Weimann J, Bauer H, Bigatello L, Bloch KD, Martin E, Zapol WM: ABO blood group and inhaled nitric oxide in acute respiratory distress syndrome. Lancet 1998; 351: 1786–7
302.
Ichinose F, Adrie C, Hurford WE, Zapol WM: Prolonged pulmonary vasodilator action of inhaled nitric oxide by Zaprinast in awake lambs. J Appl Physiol 1995; 78: 1288–95
303.
Ziegler JW, Ivy DD, Fox JJ, Kinsella JP, Clarke WR, Abman SH: Dipyridamole, a cGMP phosphodiesterase inhibitor, causes pulmonary vasodilation in the ovine fetus. Am J Physiol 1995; 269: H473–9
304.
Ziegler JW, Ivy DD, Fox JJ, Kinsella JP, Clarke WR, Abman SH: Dipyridamole potentiates pulmonary vasodilation induced by acetylcholine and nitric oxide in the ovine fetus. Am J Respir Crit Care Med 1998; 157: 1104–10
305.
Laubie M, Schmitt H: Long-lasting hyperventilation induced by almitrine: Evidence for a specific effect on carotid and thoracic chemoreceptors. Eur J Pharmacol 1980; 61: 125–36
306.
Gillart T, Bazin JE, Cosserant B, Guelon D, Aigouy L, Mansoor O, Schoeffler P: Combined nitric oxide inhalation, prone positioning and almitrine infusion improve oxygenation in severe ARDS. Can J Anaesth 1998; 45: 402–9
307.
Wysocki M, Delclaux C, Roupie E, Langeron O, Liu N, Herman B, Lemaire F, Brochard L: Additive effect on gas exchange of inhaled nitric oxide and intravenous almitrine bismesylate in the adult respiratory distress syndrome. Intensive Care Med 1994; 20: 254–9
308.
Zobel G, Urlesberger B, Dacar D, Rodl S, Reiterer F, Friehs I: Partial liquid ventilation combined with inhaled nitric oxide in acute respiratory failure with pulmonary hypertension in piglets. Pediatr Res 1997; 41: 172–7
309.
Houmes RJ, Hartog A, Verbrugge SJ, Bohm S, Lachmann B: Combining partial liquid ventilation with nitric oxide to improve gas exchange in acute lung injury. Intensive Care Med 1997; 23: 163–9
310.
Uchida T, Nakazawa K, Yokoyama K, Makita K, Amaha K: The combination of partial liquid ventilation and inhaled nitric oxide in the severe oleic acid lung injury model. Chest 1998; 113: 1658–66
311.
Semigran MJ, Cockrill BA, Kacmarek R, Thompson BT, Zapol WM, Dec GW, Fifer MA: Hemodynamic effects of inhaled nitric oxide in heart failure. J Am Coll Cardiol 1994; 24: 982–8
312.
Loh E, Stamler JS, Hare JM, Loscalzo J, Colucci WS: Cardiovascular effects of inhaled nitric oxide in patients with left ventricular dysfunction. Circulation 1994; 90: 2780–5
313.
Hayward CS, Rogers P, Keogh AM, Kelly R, Spratt PM, Macdonald PS: Inhaled nitric oxide in cardiac failure: Vascular versus ventricular effects. J Cardiovasc Pharmacol 1996; 27: 80–5
314.
Bocchi EA, Bacal F, Auler Junior JO, Carmone MJ, Bellotti G, Pileggi F: Inhaled nitric oxide leading to pulmonary edema in stable severe heart failure. Am J Cardiol 1994; 74: 70–2
315.
Lavoie A, Hall JB, Olson DM, Wylam ME: Life-threatening effects of discontinuing inhaled nitric oxide in severe respiratory failure. Am J Respir Crit Care Med 1996; 153: 1985–7
316.
Ma XL, Lopez BL, Christopher TA, Birenbaum DS, Vinten-Johansen J: Exogenous NO inhibits basal NO release from vascular endothelium in vitro and in vivo. Am J Physiol 1996; 271: H2045–51
317.
Chen LY, Mehta JL: Downregulation of nitric oxide synthase activity in human platelets by nitroglycerin and authentic nitric oxide. J Investig Med 1997; 45: 69–74
318.
Sheehy AM, Burson MA, Black SM: Nitric oxide exposure inhibits endothelial NOS activity but not gene expression: A role for superoxide. Am J Physiol 1998; 274: L833–41
319.
Frank DU, Horstman DJ, Morris GN, Johns RA, Rich GF: Regulation of the endogenous NO pathway by prolonged inhaled NO in rats. J Appl Physiol 1998; 85: 1070–8
320.
Ivy DD, Kinsella JP, Ziegler JW, Abman SH: Dipyridamole attenuates rebound pulmonary hypertension after inhaled nitric oxide withdrawal in postoperative congenital heart disease. J Thorac Cardiovasc Surg 1998; 115: 875–82
321.
Samama CM, Diaby M, Fellahi JL, Mdhafar A, Eyraud D, Arock M, Guillosson JJ, Coriat P, Rouby JJ: Inhibition of platelet aggregation by inhaled nitric oxide in patients with acute respiratory distress syndrome. A NESTHESIOLOGY 1995; 83: 56–65
322.
George TN, Johnson KJ, Bates JN, Segar JL: The effect of inhaled nitric oxide therapy on bleeding time and platelet aggregation in neonates. J Pediatr 1998; 132: 731–4
323.
Greenbaum R, Bay J, Hargreaves MD, Kain ML, Kelman GR, Nunn JF, Prys-Roberts C, Siebold K: Effects of higher oxides of nitrogen on the anaethetized dog. Br J Anaesth 1967; 39: 393–404
324.
Wessel DL, Adatia I, Thompson JE, Hickey PR: Delivery and monitoring of inhaled nitric oxide in patients with pulmonary hypertension. Crit Care Med 1994; 22: 930–8
325.
Imanaka H, Hess D, Kirmse M, Bigatello LM, Kacmarek RM, Steudel W, Hurford WE: Inaccuracies of nitric oxide delivery systems during adult mechanical ventilation. A NESTHESIOLOGY 1997; 86: 676–88
326.
Hallman M, Bry K, Turbow R, Waffarn F, Lappalainen U: Pulmonary toxicity associated with nitric oxide in term infants with severe respiratory failure. J Pediatr 1998; 132: 827–9
327.
Luhr O, Aardal S, Nathorst-Westfelt U, Berggren L, Johansson LA, Wahlin L, Frostell C: Pulmonary function in adult survivors of severe acute lung injury treated with inhaled nitric oxide. Acta Anaesthesiol Scand 1998; 42: 391–8
328.
Walmrath D, Schneider T, Schermuly R, Olschewski H, Grimminger F, Seeger W: Direct comparison of inhaled nitric oxide and aerosolized prostacyclin in acute respiratory distress syndrome. Am J Respir Crit Care Med 1996; 153: 991–6
329.
Zwissler B, Kemming G, Habler O, Kleen M, Merkel M, Haller M, Briegel J, Welte M, Peter K: Inhaled prostacyclin (PGI2) versus inhaled nitric oxide in adult respiratory distress syndrome. Am J Respir Crit Care Med 1996; 154: 1671–7
330.
Pappert D, Busch T, Gerlach H, Lewandowski K, Radermacher P, Rossaint R: Aerosolized prostacyclin versus inhaled nitric oxide in children with severe acute respiratory distress syndrome. A NESTHESIOLOGY 1995; 82: 1507–11
331.
Adrie C, Ichinose F, Holzmann A, Keefer L, Hurford WE, Zapol WM: Pulmonary vasodilation by nitric oxide gas and prodrug aerosols in acute pulmonary hypertension. J Appl Physiol 1998; 84: 435–41
332.
Brilli RJ, Krafte-Jacobs B, Smith DJ, Passerini D, Moore L, Ballard ET: Aerosolization of novel nitric oxide donors selectively reduce pulmonary hypertension. Crit Care Med 1998; 26: 1390–6
333.
Brilli RJ, Krafte-Jacobs B, Smith DJ, Roselle D, Passerini D, Vromen A, Moore L, Szabo C, Salzman AL: Intratracheal instillation of a novel NO/nucleophile adduct selectively reduces pulmonary hypertension. J Appl Physiol 1997; 83: 1968–75
334.
Adrie C, Hirani WM, Holzmann A, Keefer L, Zapol WM, Hurford WE: Selective pulmonary vasodilation by intravenous infusion of an ultrashort half-life nucleophile/nitric oxide adduct. A NESTHESIOLOGY 1998; 88: 190–5
335.
Ichinose F, Adrie C, Hurford WE, Bloch KD, Zapol WM: Selective pulmonary vasodilation induced by aerosolized zaprinast. A NESTHESIOLOGY 1998; 88: 410–6