Nitric oxide: An overview*

12
Nitric Oxide: An Overview David A. Rodeberg, MD, Mark S. Chaet, MD, Robert C. Bass, MD, Marc S. Arkovitz, MD, Victor F. Garcia, MD, Cincinnati, Ohio Nitric oxide (NO), a paracrine-acting gas enzy- matically synthesized from L-arginine, is a unique biologic mediator that has been impli- cated in a myriad of physiologic and pathophys- iologic states. It is an important regulator of vas- cular tone and may be the mediator of the hemodynamic changes involved in sepsis and cirrhosis. In addition, there is increasing evi- dence that NO is involved in coagulation, im- mune function, inhibitory innervation of the gas- trointestinal tract, protection of gastrointestinal mucosa, and the hepatotoxicity of cirrhosis. It has already been speculated that NO may repre- sent a point of control or intervention in a num- ber of disease states. The purpose of this paper is to provide the surgeon with a broad overview of the scientific and clinical aspects of this im- portant molecule. Am J Surg. 1995;170:292-303. S ince the initial identification of nitric oxide (NO) as a biologically active molecule, a tremendous number of basic science and clinical investigations about NO have entered the literature. To underscore this point, NO was named "Molecule of the Year" by Science (1992). Nitric oxide should not be confused with the anesthetic nitrous ox- ide (N20). This point was made clear by Sir Humphrey Davy, who nearly succumbed after inhaling a large amount of NO. 1 Given the broad range of functions attributed to NO, we sought to review the NO literature that would be of value to surgeons. Many of the activities and mechanisms of action attributed to NO have been elucidated using ani- mal models. For the purpose of this manuscript, these results have been generalized and combined to provide a clearer and more comprehensive picture of how NO probably func- tions in humans. Nitric oxide, a gaseous, paramagnetic, free radical, is produced from the amino acid L-arginine (L-Arg) in nu- merous mammalian celt types (Figure 1). Nitric oxide possesses a half-life of 3 to 5 seconds and is rapidly neu- From the Department of Surgery (DAR, RCB), University of Cincinnati Medical Center, and the Department of Pediatric Surgery (MSC, MSA, VFG), Childrens Hospital Medical Center, Cincinnati, Ohio. Supported by Shriners of North America Research Fellowship and Surgical Infection Society Research Fellowship. Requests for reprints should be addressed to David A. Rodeberg, MD, University of Cincinnati Medical Center, Department of Surgery, 231 Bethesda Avenue, Cincinnati, Ohio 45267. Manuscript submitted April 15, 1994 and accepted in revised form November 15, 1994. tralized by hemoglobin, methylene blue, and superoxide anion. The enzyme involved in NO production, NO syn- thase (NOS), converts the terminal guanidine group of t-Arg to NO with a m-hydroxyl-L-arginine intermediate compound. 2'3 The reaction requires nicotinamide adenine dinucleotide phosphate (NADPH) as a cofactor and uses molecular oxygen to oxidize the substrate to the final re- action products, NO and L-citrulline.4 Two forms of NOS have been identified, each with unique basal activity lev- els and cofactor requirements. 4 The isoenzyme found in neuronal tissue and vascular endothelium is constitutively expressed and requires calcium/calmodulin as cofactors. The NOS initially identified in the macrophage is in- ducible, calcium independent, and produces NO in quan- tities several magnitudes greater than the constitutive form. Both forms ofNOS are competitively inhibited by L-Arg analogues such as L-N°-monomethyl-arginine (L- NMMA), NG-nitro-L-arginine-methyl ester (L-NAME), and NQnitro-L-arginine (L-NNA). 2,5 These compounds have been important for investigations of the biologic function of NO, as well as for creating an avenue for clin- ical investigation of NO blockade. Initial investigations concentrated upon NO's involve- ment in cellular communication and vascular smooth-mus- cle relaxation. Further studies have revealed the ubiquitous nature of NO and its effects upon many other biological sys- tems. Presented here is a review of the currently identified effects and mechanisms of mammalian NO production. THE VASCULAR SYSTEM Background The vasodilatory property of acetylcholine has long been appreciated. It wasn't until 1980, however, that Furchgott and Zawadzki6 demonstrated that the endothelium was re- quired for acetylcholine-induced dilation. The acetylcholine molecule acts on a receptor present on the endothelial cell, resulting in the release of a diffusible substance, termed en- dothelium-derived relaxing factor (EDRF), causing the re- laxation of adjacent smooth-muscle cells. Early investiga- tions demonstrated that EDRF release is induced by many stimuli, including acetylcholine, bradykinin, histamine, ade- nine nucleotides, substance P, 5-hydroxytryptamine, en- dothelial stretch, shear stress, and hypoxia.7 EDRF induces smooth-muscle relaxation by causing an increase of cyclic guanosine monophosphate (cGMP) within the smooth-mus- cle cell.s'11 Following a separate line of investigation, Murad 12 demonstrated that the effects of exogenously ad- ministered nitrates (azide, hydroxylamine, sodium nitrite, ni- troglycerin, sodium nitroprusside, and nitrosamines) were mediated by the radical NO. Ignarro13demonstrated that en- dothelial cells were not required for exogenous-nitrate-me- diated vasodilation. The nitrovasodilators were known to act 292 THE AMERICAN JOURNAL OF SURGERY ® VOLUME 170 SEPTEMBER 1995

Transcript of Nitric oxide: An overview*

Nitric Oxide: An Overview David A. Rodeberg, MD, Mark S. Chaet, MD, Robert C. Bass, MD,

Marc S. Arkovitz, MD, Victor F. Garcia, MD, Cincinnati, Ohio

Nitric oxide (NO), a paracrine-acting gas enzy- matically synthesized from L-arginine, is a unique biologic mediator that has been impli- cated in a myriad of physiologic and pathophys- iologic states. It is an important regulator of vas- cular tone and may be the mediator of the hemodynamic changes involved in sepsis and cirrhosis. In addition, there is increasing evi- dence that NO is involved in coagulation, im- mune function, inhibitory innervation of the gas- trointestinal tract, protection of gastrointestinal mucosa, and the hepatotoxicity of cirrhosis. It has already been speculated that NO may repre- sent a point of control or intervention in a num- ber of disease states. The purpose of this paper is to provide the surgeon with a broad overview of the scientific and clinical aspects of this im- portant molecule. Am J Surg. 1995;170:292-303.

S ince the initial identification of nitric oxide (NO) as a biologically active molecule, a tremendous number of basic science and clinical investigations about NO

have entered the literature. To underscore this point, NO was named "Molecule of the Year" by Science (1992). Nitric oxide should not be confused with the anesthetic nitrous ox- ide (N20). This point was made clear by Sir Humphrey Davy, who nearly succumbed after inhaling a large amount of NO. 1 Given the broad range of functions attributed to NO, we sought to review the NO literature that would be of value to surgeons. Many of the activities and mechanisms of action attributed to NO have been elucidated using ani- mal models. For the purpose of this manuscript, these results have been generalized and combined to provide a clearer and more comprehensive picture of how NO probably func- tions in humans.

Nitric oxide, a gaseous, paramagnetic, free radical, is produced from the amino acid L-arginine (L-Arg) in nu- merous mammalian celt types (Figure 1). Nitric oxide possesses a half-life of 3 to 5 seconds and is rapidly neu-

From the Department of Surgery (DAR, RCB), University of Cincinnati Medical Center, and the Department of Pediatric Surgery (MSC, MSA, VFG), Childrens Hospital Medical Center, Cincinnati, Ohio.

Supported by Shriners of North America Research Fellowship and Surgical Infection Society Research Fellowship.

Requests for reprints should be addressed to David A. Rodeberg, MD, University of Cincinnati Medical Center, Department of Surgery, 231 Bethesda Avenue, Cincinnati, Ohio 45267.

Manuscript submitted April 15, 1994 and accepted in revised form November 15, 1994.

tralized by hemoglobin, methylene blue, and superoxide anion. The enzyme involved in NO production, NO syn- thase (NOS), converts the terminal guanidine group of t-Arg to NO with a m-hydroxyl-L-arginine intermediate compound. 2'3 The reaction requires nicotinamide adenine dinucleotide phosphate (NADPH) as a cofactor and uses molecular oxygen to oxidize the substrate to the final re- action products, NO and L-citrulline. 4 Two forms of NOS have been identified, each with unique basal activity lev- els and cofactor requirements. 4 The isoenzyme found in neuronal tissue and vascular endothelium is constitutively expressed and requires calcium/calmodulin as cofactors. The NOS initially identified in the macrophage is in- ducible, calcium independent, and produces NO in quan- tities several magnitudes greater than the constitutive form. Both forms ofNOS are competitively inhibited by L-Arg analogues such as L-N°-monomethyl-arginine (L- NMMA), NG-nitro-L-arginine-methyl ester (L-NAME), and NQnitro-L-arginine (L-NNA). 2,5 These compounds have been important for investigations of the biologic function of NO, as well as for creating an avenue for clin- ical investigation of NO blockade.

Initial investigations concentrated upon NO's involve- ment in cellular communication and vascular smooth-mus- cle relaxation. Further studies have revealed the ubiquitous nature of NO and its effects upon many other biological sys- tems. Presented here is a review of the currently identified effects and mechanisms of mammalian NO production.

T H E V A S C U L A R SYSTEM Background

The vasodilatory property of acetylcholine has long been appreciated. It wasn't until 1980, however, that Furchgott and Zawadzki 6 demonstrated that the endothelium was re- quired for acetylcholine-induced dilation. The acetylcholine molecule acts on a receptor present on the endothelial cell, resulting in the release of a diffusible substance, termed en- dothelium-derived relaxing factor (EDRF), causing the re- laxation of adjacent smooth-muscle cells. Early investiga- tions demonstrated that EDRF release is induced by many stimuli, including acetylcholine, bradykinin, histamine, ade- nine nucleotides, substance P, 5-hydroxytryptamine, en- dothelial stretch, shear stress, and hypoxia. 7 EDRF induces smooth-muscle relaxation by causing an increase of cyclic guanosine monophosphate (cGMP) within the smooth-mus- cle cell. s'11 Following a separate line of investigation, Murad 12 demonstrated that the effects of exogenously ad- ministered nitrates (azide, hydroxylamine, sodium nitrite, ni- troglycerin, sodium nitroprusside, and nitrosamines) were mediated by the radical NO. Ignarro 13 demonstrated that en- dothelial cells were not required for exogenous-nitrate-me- diated vasodilation. The nitrovasodilators were known to act

292 THE AMERICAN JOURNAL OF SURGERY ® VOLUME 170 SEPTEMBER 1995

through stimulation of soluble guanylate cyclase leading to the formation of cGMP, similar to EDRE s'9

In 1987, both Palmer and Ignarro independently proposed that EDRF and NO were the same molecule. 13'14 In a model using porcine endothelium cultured on microcarrier beads, they demonsn'ated many similarities between these two com- pounds. Both EDRF and NO were inhibited by superoxide anion, methylene blue, and hemoglobin, and were enhanced by superoxide dismutase. Both molecules possessed a half- life of only 3 to 5 seconds. Each caused an increase in in- tracellular cGMP levels in smooth-muscle cells by activat- ing a soluble guanylate cyclase enzyme. Simultaneous EDRF bioassay and NO chemoluminescence assays illustrated that the amount of NO released was sufficient to account for all of EDRF's activity. 14

The release of NO from endothelial cells is primarily me- diated by guanosine triphosphate (GTP)-associated proteins (G proteins), which link the receptors to NOS) 5 Different receptors stimulate different G proteins; with some being pertussis-toxin sensitive (serotonin) and others, toxin in- sensitive (bfadykinin). There is also the possibility that the pertussis-toxin-insensitive mechanisms ,nay not involve G proteins at all. The release of NO is dependent on pulsatile flow as well as shear stress, and is enhanced by other va- sodilatory agents. ]6a7 Nitric oxide acts in a typical paracrine fashion. Diffusion into the vascular lumen affects platelets, while abluminal diffusion affects the surrounding smooth muscle. 7 It is possible that NO itself is not the diffusing agent but enters the extracellular domain as an S-nitrosylthiol in- termediate compound that is formed in physiologic condi- tions when NO interacts with sulflaydryl groups. This hy- pothesis has been supported by the observation by Ignarro et al is that the activity of both exogenous and endogenous nitrates requires the presence of sulfhydryl groups. Further work demonstrated that EDRF has a longer half-life than

I OVERVIEW OF NITRIC OXIDE/RODEBERG ET AL

02

~ ' NO + L-citrulline L-arginine Nitric Oxide Synthase

nitrite (NO2-)

nitrate (NOa-)

Figure 1. Synthesis of nitric oxide (NO) in mammalian cells.

pure NO, which is more consistent with the need for an S- nitrosylthiol intermediate. 19'2° In addition, vascular Smooth- muscle cells and platelets respond to both S-nitrosylthiols and NO similarly. It has been postulated that, in the blood vessels of the central nervous system, NO is not the effec- tor molecule at all, but that the responses are solely depen- dent on the S-nitrosylthiols. 21 However, the mo, re widely ac- cepted opinion is that NO actually causes the smobth-muscle relaxation, and that the S-nitrosylthiols represent only in- termediate compounds or reaction products. 22

The mechanism by which NO causes the relaxation of vas- cular smooth muscle was initially proposed after the observa- tion that exogenous nitrates and NO caused an increase in cGMP, but not cyclic adenosine monophosphate (cAMP). 23 The cGMP-mediated relaxation of smooth muscle appears to occur by two mechanisms. First NO was observed to decrease intracellular calcium levels and increase the permeability of the potassium channels, leading to a hyperp01arization of the plasma membrane that prevents subsequent contracn0n . . 24 Second, cGMP activates protein kinases, which eventually lead to the dephosphorylation of the myosin light chains, pre- venting the myosin/actin interaction. 7

platelet aggregation

I 0 2 -

EDCF I

T B A 2

platelet disaggregation

EDRF I I ] ]

P G H 2 e n d o t h e l i n N O PGI 2

1 1

cGMP cAMP K* channel

contraction dilation

I endotheliUm

vascular smooth muscle

Figure 2. Interactions between vascular endothelium, smooth muscle, and platelets. Nitric oxide (NO) increases the permeability of the potassium channels, leading to a hyperpolarization of the plasma membrane that prevents subsequent contraction. NO and prostacyclin (PGI2) inhibit platelet aggregation and platelet or monocyte adhesion to the endothelium, and stimulate platelet dis - aggregation. PGH 2 = prostaglandin H2; TBA 2 = thromboxane; 02- = superoxide anion; EDRF = endothelium-derived relaxing fac- tor; EDCF = endothelium-derived contracting factors; K + = potassium ion; cGMP = cyclic guanosine monophosphate; cAMP = cyclic adenosine monophosphate.

THE AMERICAN JOURNAL OF SURGERY ® VOLUME 170 SEPTEMBER 1995 29~

OVERVIEW OF NITRIC OXIDE/RODEBERG ET AL 1

• Vascular Tone Normal vascular tone is determined bythe interactions of

endothelium-derived vasodilators (ie, NO, prostacyclin [PGI2]) and vasoconstrictors (ie, endothelin) (Figure 2). Under baseline physiologic conditions, this balance is in fa- vor of vasodilation. 25"27 NO and PGI 2 inhibit smooth-muscle contraction and proliferation, platelet aggregation, and platelet or monocyte adhesion to the endothelium, and both stimulate platelet disaggregation. 2~,2s,29 These compounds op- erate synergistically in their vasodilatory and platelet-antiag- gregatory actions, but act through different second-messen- ger mechanisms; PGI 2 through cAMP, and NO through cGME Despite PGI2's significantly longer half-life, it appears that NO production is the major determinant of resting vas- cular tone. 3°'33 Using L-Arg analogues, researchers have demonstrated that the vasodilatory response to NO occurs predominantly in the arterial system, suggesting that arteries have a higher basal release of NO and are more dependent on NO for vascular tone regulation when compared to veins. Since veins have less NO release, they have a compensatory increased level of soluble guany!ate cyclase. This Would ex- plain the clinical observation that the venom capacitance vessels are more susceptible to the vasodilatory action of ex- ogenous nitrovasodilators, which bypass NOS and directly stimulate the soluble guanylate cyclase. The development of tolerance to exogenously administered nitrovasodilators ap- pears to be due to impaired metabolism to NO and desensi- tization of soluble guanylate cyclase to NO. 1s'34"36

Disease States Nitric oxide is an important physiologic regulator of blood

flow and perfusion pressure to isolated vascular beds in re- sponse to appropriate physiologic stimuli. For example, the renal, cardiac, pulmonary, and muscular vasculatums depend upon NO for basal regulation. 374: These observations have not only led to the improved understanding of vasomotor regulation, but also to many theories about various organ. based disease processes. Several investigations have pre- sented evidence for the involvement of dysfunctional en- dothelium in hypertension, 42,43 atherosclerosis, 44 and vasospasm. 45

Decreased levels of activity of NO have been implicated as a mechanism of primary or essential hypertension. 46"49 The degree of endothelial dysfunction parallels the degree of hy. pertension. The inability of vessels to vasodilate appears to be related to decreased NO release and not to the reduced ability of smooth-muscle cells to respond to NO. 5°'5: This may be due to decreased t-Arg availability 52-54 or decreased G-protein function in atherosclerotic vessels secondary to the presence of Oxidized low-density lipoprotein (LDL). 55 A model has been proposed by Flavahan, 56 in which the en- dothelial dysfunction occurs in distinct phases. Early in the disease when there are low concentrations of oxidized LDL, the dysfunction would be due to impairment of the G-pro- tein--dependent pathways. Later, as more oxidized LDL ac- cumulates, other signal transduction processes in the en- dothelial cell become dysfunctional, eventually inhibiting the release of NO by a nonspeeific action, such as decreased L-Arg availability, decreased sensitivity of the vascular smooth muscle to NO, and/or an increased degradation of

NO. Some studies suggest that a deficiency in NO may even exacerbate atherogenesis. This may occur since NO inhibits both the release of platelet granules containing mitogens and vascular smooth-muscle mitogenesis. 57'5s • Other pathologic conditions include NO inactivation by advanced glycosylation products found within endothelial and smooth-muscle cells of a diabetic animal model. 59"61 In addition, the hypotension of septic shock is dependent on NO and will be discussed in further detail in the immuno- logic section. Similarly, the vascular hyporeactivity to va- sopressor agents such as norepinephrine, seen with hemo- dynamic decompensation after prolonged hemorrhagic shock, is mediated by NO. 62 The vasospasm seen after sub- arachnoid hemorrhage is probably due to inactivation of NO by extravascular hemoglobin. 63 As more is learned about the mechanisms behind these abnormalities in NO metabolism, therapies based on NOS blockade or NO ad- ministration should become useful. Studies are currently un- der way looking at the therapeutic use of inhaled NO to improve the pulmonary hypertension and ventilation/per- fusion mismatch that occurs during respiratory distress syn- drome, and L-Arg analogue inhibitors are being used in models of septic shock.

Coagulation Nitric oxide can act in a paracrine fashion, and as such has

effects on both Sides of the endothelial cell. Within the lu- men of the vessel, NO interacts with platelets, inhibiting both adhesion to endothelial cells and aggregation. 64,65 In 1986, Azuraa et a166 demonstrated that EDRF inhibited arachidonic-acid-induced platelet aggregation. Similarly, exogenous nitrovasodilators and NO were found to increase platelet intracellular cGMP, leading to a subsequent inhibi- tion of aggregation. 67 Endothelial-dependent inhibition of platelet aggregation is controlled by two separate mecha- nisms. Endothelial-derived PGI 2 inhibits platelet aggrega- tion by increasing intracellular cAMP, whereas NO works by increasing cGMP. 68 The two molecules act synergistically, but NO appears to be the major reguiat0rymolecule, since platelet adhesion to collagen and endothelial matrix can be completeiy blocked by NO, but only partially prevented by PGI2 .69"71 Nitric oxide also inhibits platelet and leukocyte adhesion to endothelial cells, as well as mediating platelet plug disaggregation. 26'28'29'7° Furthermore, nitrovasodilators have been shown to prolong the activity of tissue pla> minogen activator, thereby increasing thrombolytic activity and preventing vessel occlusion. 72

Platelets also release NO, which acts in an autocrine fash- ion to prevent aggregationY The platelets are stimulated to release NO by compounds such as bradykinin. 69 The platelet NOS enzyme is, like the endothelial cells, a constitutively ex- pressed NADPH- and calcium/calmodulin-dependent en- zyme. Platelet NOS activity is dependent on L-Arg, inhibited by L-Arg competitive analogues, and potentiated by cGMP phosphodiesterase inhibitors, similar to endothelial NOS.

T H E I M M U N E SYSTEM Background

Nitric oxide ha s emerged as a central participant in the im- mune response. The investigation of NO as an immunore-

294 THE AMERICAN JOURNAL OF SURGERY ® VOLUME 170 SEPTEMBER 1995

I OVERVIEW oF NITRIC OXIDF-JRODEBERG ET AL

Figure 3. Proposed mechanism of macrophage cytotOxic effects. Macro- phages stimulated with interferon-gamma (IFN-7), lipopolysaccharide (LPS), and tu- mor necrosis factor (TNF) produce an ih- ducible form of nitric oxide (NO) syn- thetase, which in turn produces NO from L-arginine (L-ARG) in amounts several magnitudes greater than does the consti- tutive form found in neuronal and vascu- lar tissue: Macrophage-derived NO causes an increase in target cell intracel- lular cyclic guanosine monophosphate (cGMP) levels and a decrease in DNA synthesis. It is thought that NO binds to the iron-sulfhydryl (Fe-S) prosthetic groups of aconitase and complexes I and II of the mitochondrial electron transport chain, disrupting Oxidative metabolism and causing target cell death.

IFN-~/

LPS

f L . . . . . . ! T N F A AA

TUMOR/BACTERIA

active Compound began with the observation that high lev- els of urinary nitrates were recovered from a patient with in- fectious diarrhea. 74 Later, the production of nitrates and ni- trites was detected after immunostimulation of cell cultures, laboratory animals, and human subjects. 75"77 The source of these nitrogen compounds remained unclear until Stuehr and Marietta 7s demonstrated that serum and urinary nitrates were increased in normal mice after immunostimulation, but not in C3H/HeJ mice, which have a genetic alteration ren- dering their macrophages resistant to endotoxin. The rodent macrophage cell culture model has served as an invaluable tool for the investigation of immunostimulated nitrite and nitrate production. It was through the use of this model that Marietta et a179 identified NO as the intermediate compound in the L-Arg-to-nitrite/nitrate pathway.

The enzyme responsible for NO production in the mam- malian macrophage differs from that in the endothelial cell. In contrast to the constitutively expressed enzyme found in endothelium, the macrophage NOS is inducible and not cal- cium dependent, but does require the cofact0rs NADPH and tetrahydrobiopterin for NO production, s° Immunologic stimulation increases the amount of inducible NOS by in- creasing its rate of transcription. 8~'84 Macrophages stimulated with interferon gamma (IFN-7) and !ipopolysaccharide (LPS) have NOS messenger ribonucleic acid (mRNA) pre- sent by 2 hours, and NOS protein, by 4 hours. The NOS protein may remain present for several days. s2 The inducible form of NOS produces NO in amounts several magnitudes greater than the constitutive form. Similar to the constitu- tive form, inducible NOS is inhibited by L-Arg analogues. The activity of the inducible form of NOS is preferentially inhibited by the L-Arg analog L-canavanine and by amino- guanidine, s5 The expression of the inducible form of NOS also is inhibited by glucocorticoids. 73'77'sl The mechanism through which steroids inhibit inducible NOS is unclear, since glucocorticoids affect the production of other cytokines that regulate NO production.

Although cultured macrophages can produce large amounts of NO, multiple cytokines are required to achieve

maximal induction. Laboratories have used various combi- nations of IFN- 7, tumor necrosis factor-alpha (TNF-cz), and interleukiml (IL-1), in addition to bacterial endotoxins and exotoxins, to induce macrophage NO production. IFN- 7 or LPS alone can induce noncytotoxic levels of NO. However, when cytokines work synergistically, they can induce cy- totoxic levels of NO. s<s7 Priming cell cultures with IFN- 7 prior to exposure to other agents results in enhanced NO production. The synergistic activity between IFN- 7 and mi- crobial products is mediated by TNF-cz. 87 TNF-cz acts in an autocrine fashion to amplify the synthesis and release of NO by primed macrophages; however, like IL-1, it cannot stim- ulate NO synthesis by itself, s7 Antimicrobial activity and NO production parallel TNF-cz activity. One of the diffi- culties in this field of research has been the inability to in- duce human macrophage NO production with the methods used for rodent cell systems. There is a report, however, of the involvement of NO in human macrophage-mediated killing of an avirutent strain of Mycobacterium avium, ss In addition, human NO production has been demonstrated in hepatocytes after stimulation with various combinations of Escherichia coli LPS, IL-t6, TNF-a, and IFN-7. 89 Further work in developing an inducible human macrophage model is needed to fully understand the function of NO in the hu- man immune response.

Inducible NO inhibits the viability and replication of many types of microbes. A strong correlation exists between an- timicrobial activity and production of L-Arg-derived NO by cytokine-activated cells observed during in vitro studies. 87'9° The mechanism by which macrophage NO effects its cy- totoxic activity remains unclear, although several hypothe- ses have been entertained (Figure 3). Superoxide anion and NO interact in vitro to form peroxynitrite, a precursor to two potent oxidants, hydroxide ion (OH-) and nitrite (NO2-).91,92 Although evidence for this reaction in living or- ganisms has not been demonstrated, it is possible that the cytotoxicity of NO relies upon these and other reactive ni- trite and oxygen intermediates. As previously noted, NO ac- tively binds heme groups. Some investigators postulate that

THE AMERICAN JOURNAL OF SURGERY ® VOLUME 170 SEPTEMBER 1995 295

OVERVIEW OF NITRIC OXIDE/RODEBERG ET AL I

NO binds to the iron-sul~ydryl prosthetic groups of aconi- tase and complexes I and II of the mitochondrial electron transport chain, resulting in cell death.

Nonspecific Immunity Nitric oxide production plays a role in the induction of non-

specific immunity and the antimicrobial response to a variety of extracellular parasites and some tumor cells. The impor- tance of the macrophage and various cytokines such as TNF. cz and IFNq, in response to parasitic infection has been well documented. 93,94 Further investigation has implicated NO as the final mediator of parasitic defense. Nitric-oxide-depen- dent mechanisms have been identified in the killing of intra- cellular Trypanosoma, Mycobacterium, Schistosoma, and Leishmania organisms. 94"97 The importance of macrophage NO production in tumoricidal activity has also been described us- ing in vitro models. 98'99 The precise mechanism of NO-medi- ated killing of these organisms is unknown. However, these observations suggest that macrophage NO production con- tributes significantly to nonspecific immunity.

Specific Immunity The role of NO in specific immunity has been another in-

teresting field of investigation. Human lymphocyte blasto- genic proliferation is augmented in the presence of t-Arg or exogenous NO during the synthesis phase of the cell cycle when deoxyribonucleic-acid (DNA) replication occurs, and is decreased when specific inhibitors of NOS are present, m° These observations suggest that NO production promotes DNA synthesis in immunologically activated lymphocytes. The mechanism may involve NO, since increases of cGMP have been demonstrated in proliferating lymphocytes. 1°1 Contradictory data have been reported using murine T-cell populations. In this system, T-cell proliferation is inhibited when co-cultured with NO-secreting, concanavalin-A-acti- vated macrophages3 °2 Interestingly, in the murine system, it appears that the T-helper type 2 (Th2) cell production of IL-4 inhibits macrophage NOS, while IFN-~{ released by T- helper type 1 (Thl) cells induces NOS. m3 This represents a mechanism by which Thl and Th2 cells, mediators of de- layed-type hypersensitivity, reciprocally regulate macrophage NO synthesis. The differing effects of NO on lymphocyte proliferation may be due to quantitative differences in NO production, supporting a proposed theory that low concen- trations of NO are required for lymphocyte replication, while excess amounts of NO are cytotoxic.

The involvement of NO in transplant rejection has been addressed from both diagnostic and therapeutic aspects. Langrehr and colleagues 1°4 using a rodent allograft model, demonstrated that NO production by mixed splenic lym- phocytes and sponge matrix allograft-infihrating cells inhibit T-ceU antigen-specific proliferation. Further work using FK 506, an inhibitor of T-cell IL-2 production, suggests that NO production from sponge matrix graft-infiltrating cells is de- pendent upon IL-2 production3 °5 These observations sup- port the theory that NO is involved in regulation of the re- sponse to allografts. The interaction between T-cell cytokine stimulation of macrophage NO and NO suppression of T- cell proliferation and cytolytic activity may be the mecha- nisms involved in this regulation.

In the above-described models, NO appears to contribute to the inhibition of cell-mediated rejection. In contrast to these observations, NO has been implicated as an important mediator of graft-versus-host reaction (GvHR). Significant amounts of NO are produced in the early course of GvHR and may serve as a useful marker to the onset of this process. 1°6a°7 Blockade of NO production with the L-Arg analogue L-NMMA abolishes the intestinal mucosal injury associated with GvHR in mice after intraperitoneal injec- tion of splenocytes. 1°s These findings suggest a possible ther- apeutic role for specific NO-blocking agents in the man- agement of GvHR enteropathy. Using a rat heart allograft model, researchers have identified NO as an important part of vascularized allograft rejection. 1 Further studies on NO and solid organ transplantation are currently underway in several laboratories.

Sepsis and Multisystem Organ Failure The inducible form of NO is produced by macrophages re-

siding in various tissues in response to several noxious stim- uli, including LPS. This response leads to overall NO levels that exceed the resting, nonstressed states. It is this relative overproduction of NO and the subsequent vasodilation that are thought to mediate N©'s physiologic role during sepsis and multiorgan failure. Clinical and experimental studies, however, failed to define a specific role for NO during sep- sis or multiorgan failure. 1°9q15

Studies in patients who were septic from multiple causes (excluding trauma) have shown increased serum levels of NO, measured as the ratio of NOr/NO3-. 113 All trauma pa- tients, including septic trauma patients, had NO levels that were lower than that of normal controls. The authors rea- soned that trauma patients had an inhibition of NO release secondary to hypovolemia. In this study, elevated NO lev- els were inversely related to systemic vascular resistance. Conversely, elevated NO levels correlated positively with in- creased serum endotoxi~ levels. Several subsequent clinical trials evaluated the efficacy of NOS blockade in treating the systemic effects of sepsis. 1~4,n5 Petros et al n5 demonstrated that the IV infusion of L-NMMA and L-NAME reversed the intractable hypotension and vascu!ar dilation observed in 2 septic patients. Similarly, Lorente et al u4 described a cohort of septic patients in whom an infusion of L-NMMA was as- sociated with elevated blood pressure, systemic vascular re- sistance, and peripheral vascular resistance. These changes were transient, reversible with infusion of t-Arg, and paral- leled by a concomitant decrease in cardiac index and oxy- gen delivery. The size of the groups and the nature of the studies did not allow for evaluation of the effects of L- NMMA on survival, morbidity, and mortality.

Most of the experimental research of the role of NO dur- ing sepsis takes advantage of competitive inhibition of NOS. Inhibition of NOS has been looked at in various organ sys- tems, as well as in the organism as a whole. It appears that NO mediates its effects during sepsis via its vasodilating properties.62,~16a 17 More important, however, is the fact that recent studies have shown both beneficial and harmful ef- fects of endogenous NO on the stressed organism as a whole. Kilbourn et al l°9'11° demonstrated that blocking NOS with the L-Arg analogue L-NMMA reversed hypotension induced

296 THE AMERICAN JOURNAL OF SURGERY ® VOLUME 170 SEPTEMBER 1995

OVERVIEW OF NITRIC OXIDF-JRODEBERG ET AL

by endotoxin and TNF infusion. The animals again became hypotensive when given L-Arg. Similarly, Lorente et al 112 de- scribed an increase in blood pressure, systemic vascular re- sistance, pulmonary artery pressure, and pulmonary vascular resistance when septic sheep where treated with L-NMMA. Pretreatment with L-Arg in this experiment abolished these changes. The conclusion to be drawn from these studies is that NO plays a pathologic role in the hemodynamic re- sponse to sepsis.

While these studies would tend to suggest that NOS block- ade might be beneficial to the septic organism, and thus clin- ically important, other studies have drawn the opposite con- clusion. Studies using LPS-injected rabbits concluded that L-NMMA injection prior to LPS administration had no ap- preciable effect on hypotension induced by LPS) Is Moreover, NOS blockade resulted in significantly higher mortality and coronary vasoconstriction. These effects were partially reversible with the addition of the NO donor, S- nitroso-N-acetyl penicillamine. Similarly, Klabunde and Helgren 119 showed that administration of L-NMMA in a ca- nine sepsis model did little to abrogate endotoxin-induced hypotension.

The effects of NO on various organ systems during sepsis have also been explored. Nitric oxide is produced by inducible NOS present in the Kupffer cells of the liver. When stimu- lated in an in vitro model by LPS, however, hepatocytes re- quired a combination ofTNE IL-1, OFN.?, and LPS for NO production3 2°'121 In addition, in vitro studies have found an association between NO production and inhibition of total protein synthesis in hepatocytes. 122 The conclusion from this latter study was that NO is harmful to the liver during sep- sis. In vivo studies using LPS-challenged mice have, however, found that NOS blockade with L-NMMA was associated with significant liver injury, manifested by an elevation in liver enzymes, as well as in ornithine carbamoyltransferase. 12° The article suggests that NO improves or protects hepatic blood flow and perfusion during sepsis. The significance of the in vitro inhibition of protein synthesis is somewhat less clear in light of these in vivo findings.

In vitro studies have also demonstrated both beneficial as well as negative effects of NO on myocardium. Ballingand et a1123 described an elevated production of NO by myocytes when these cells were exposed to medium conditioned with LPS-stimulated macrophages. This elevated NO production was associated with reduced myocyte contractility, which was reversed with L-NMMA. Conversely, in an experimen- tal model of feline splanchnic ischemia/reperfusion, serum myocardial depressant factor levels were reduced with the addition of sodium nitrite (NaNO2), an NO-generating molecule, n6

Studies in rats have used NOS blockade to investigate what role NO may play in the renal pathophysiology associated with hyp0volemic shock) 24 Specifically, the falls in mean ar- terial blood pressure, renal plasma flow, and glomerular fil- tration rate seen in hemorrhagic shock were reversed with the addition of L-NMMA. This sparing effect was reversed with the addition of h-Arg. The conclusion from this study was that NO plays an important role in the renal impair- ment seen during hemorrhagic shock. Similar studies in sep- tic models have not been reported.

In summary, sepsis, hypovolemia, and hypoxia each play a significant role in the pathophysiology of multiorgan failure and macrophage activation. Inducible NO from activated macrophages may be responsible for the profound loss of vas- cular tone seen in septic patients. NO production by acti- vated macrophages is complex and closely interrelated with the production of other mediators of inflammation, such as endotoxin, cytokines, prostaglandins, and interleukins. The problem with the experimental models that deal with NO synthesis is that NOS blockade is nonspecific, blocking both the inducible and constitutive forms, thus making it difficult to describe a specific role to the inducible form of NO. The role of NO during multiorgan failure most likely represents a complex interaction between the inducible and constitu- tive forms of NOS. The ability to reconcile these conflict- ing results will be determined by the ability to selectively block either the inducible or the constitutive form of NOS.

T H E G A S T R O I N T E S T I N A L SYSTEM Background

Nitric oxide has been implicated in a number of physio- logic and pathophysiologic functions in the gut. The mech- anisms by which NO is suspected of carrying out these ac- tions fall into three major categories--neurotransmitter, vasorelaxant, and paracrine mediator. There is considerable evidence that NO is one of the nonadrenergic, noncholin- ergic (NANC) neurotransmitters, functions to protect gas- tric and intestinal mucosae, mediates the hemodynamic de- rangements seen in sepsis and cirrhosis, and acts as a paracrine mediator of hepatotoxicity. 125

NANC Neurotransmission An important class of neurons in the wall of the gut, the

NANC neurons mediate relaxation of the gastrointestinal smooth muscles, which may include many of the inhibitory reflexes of the gastrointestinal tract, such as relaxation of the lower esophageal sphincter (LES) during swallowing, prox- imal stomach relaxation during filling, and descending in- hibition of small-intestine contraction due to disten- sion. 126'127 The neurotransmitter of NANC neurons has not been conclusively identified, although vasoactive intestinal peptide (VIP) and adenosine triphosphate (ATP) have been implicated. 127 However, there is now increasing evidence that NO, alone or in concert with other substances, is one of the primary neurotransmitters of the NANC neurons in many tissues.

Investigators have determined that stimulation of NANC neurons in canine ileocecal junction and rat gastric fundus causes the release of a compound with the characteristics of N O J 28'129 By using L-Arg analogues and oxyhemoglobin, it has been demonstrated that NO is involved in mediating NANC relaxation in the LES, 13° stomach, 13H33 small intes- tine, 134a35 sphincter of Oddi, 136 ileocecal junction and colon, ~37'138 and anal sphincters 139,14° in a variety of species. In human tissues, L-Arg analogues completely blocked NANC relaxation of the internal anal sphincter and par- tially blocked those in circular colonic muscles. TM These re- suits support the hypothesis that NO is the NANC neuro- transmitter for the internal anal sphincter and that NO plays a role in colonic smooth-muscle relaxation. In circular mus-

THE AMERICAN JOURNAL OF SURGERY ® VOLUME 170 SEPTEMBER 1995 297

OVERVIEW OF NITRIC OXIDE/RODEBERG ET AL I

TABLE

Criteria From the Literature for Nitric Oxide (NO) as a Neurotransmitter

Synthesis of NO in neurons 136,137

Release of a substance with characteristics of NO on stimulation of NANC neurons m,112

Application of exogenous NO mimics effects of NANC nerve stimulation130j 32

A mechanism for inactivation (NO rapidly and spontaneously breaks down) TM

Inhibition of NO synthesis with specific antagonists attenuates the effect of NANC stimulation m,~a2,1a4,~a5

NANC = nonadrenergic, noncholinergic neurotransmitters,

cle of human jejunum, exogenously administered NO grossly mimics the effects of NANC stimulation; however, there are differences in the characteristics of the electrical responses. In human jejunum, NANC stimulation elicits smooth-mus- cle-membrane hyperpolarizations known as inhibitory junc- tional potentials. Inhibitory junctional potentials are char- acterized by a rapid and Sustained component to the hyperpolarization. L-Arg analogues block the sustained com- ponent but not the initial rapid portion of the inhibitory junctional potentials. 142 These data suggest that more than one transmitter may be involved in mediating the NANC response in the human jejunum.

Nitric oxide has fulfilled most of the criteria classically used to define a neurotransmitter (Table) and may be the final NANC neurotransmitter. 143445 Several other neurotrans- mitters such as ATE y-aminobutyric acid, and VIP have been implicated in NANC neurotransmission, and the precise re- lationship of NO to other potential neurotransmitters re- mains to be defined. 14644s Nitric oxide may be co-released with other neurotransmitters, or its production may be stim- ulated by a neurotransmitter in the target muscle of some tissues. 149 tt is also possible that different NANC neuro- transmitters may function in different parts of the gut, may vary between species, or may change within a specific organ with aging and development. 126

Gastrointestinal Mucosa In the stomach, a decrease in gastric mucosal blood flow

has been identified as a prerequisite to the development of acute erosions and stress ulcers) 5°,151 Nitric oxide synthase has been detected in the gastric mucosa, and NO appears to play a role in protecting the gastric mucosa during physio- logic stress by acting as an endogenous vas0dilator and thus supporting mucosal blood f low. 152'125 Using hydrogen gas clearance to measure gastric mucosal blood flow, Pique et aP 53a54 have demonstrated that endogenous NO modulates basal tone in the gastric vasculature. In similar experiments Using L-Arg analogues to block NO production, NO was ob- served to have no effect on resting-acid output, but did make a substantial contribution to the vasodilation associated with the stimulation of acid secretion. 154 In other experiments, the use of L-NMMA alone did not result in injury to rat gas- tric mucosa. When used in conjunction with sensory neu- ropeptide depletion (capsaicin) or PGI 2 blockage (in- domethacin), however, L-NMMA resulted in a dose-dependent, acute mucosal injury. 155 Thus it appears that

NO, PGI2, and sensory neuropeptides interact to prevent gastric mucosal injury. In a rat model of ethanol-induced in- jury to gastric mucosa in ex vivo studies, topical application of NO reduced the severity of mucosal damage. 156 A brief delay in the application of NO, however, resulted in a loss of the protective effect. Since ethanol is associated with ini- tial vasoconstriction and then vasodilation of the mucosal blood vessels, MacNaughton et a1156 concluded that NO may act by blocking the initial vasoconstriction associated with ethanol. The exact mechanism of NO's protective effect is unclear, but may relate to vasodilation, inhibition of platelet aggregation in the gastric microvasculature, or a protective effect on the epithelial cells themselves.- t25 No studies have been done in humans to date.

Loss of the intestinal mucosal barrier has been implicated in the cause of bacterial translocation and multisystem or- gan failure. 157J58 Recent work in animal models suggests that NO may help protect against some of the effects of endo- toxic shock by protecting the mucosa of the small intestine. In the small intestine of the rat', pretreatment with L-Arg analogues exacerbates acute intestinal vascular damage, plasma leakage, and hemorrhage, and is associated with en- dotoxin infusion3 59 S-nitroso-N-acetyl-penicillamine, which generates NO, moderated the damage in this model. 16° In an autoperfused segment of feline small intes- tine, infusion of L-NAME resulted in a sixfold increase in mucosal permeability, as assessed by leakage of chromium 51-labeled ethylenediaminetetraacetic acid ([SICr]EDTA) without gross mucosal damage)6 l This effect was reversed by treatment with L-Arg and appeared to be independent of blood flow or neutrophil adherence. This data led Kubes TM

to propose that NO has a direct effect on the epithelial cell barrier, and its inactivation under pathophysiologic condi- tions may contribute to loss of mucosal barrier function. In addition to supporting mucosal blood flow, NO may have an antioxidant effect by binding to and neutralizing superoxide anion, thereby limiting the damage induced by neutrophils during endotoxemia/25 Although these results suggest a role for NO in maintaining a mucosal barrier, the mechanism of this effect is unclear and may be multifactorial.

Hemodynamic Disturbances During Cirrhosis and Sepsis Portal hypertension secondary to cirrhosis and septic shock

involves a similar set of systemic hemodynamic disturbances. In both situations there is increased cardiac output, increased heart rate, decreased systemic vascular resistance, decreased blood pressure, and a resistance to vasopressors) 62a63 Tl~ere is now evidence that NO may be the mediator of these he- modynamic alterations.

Patients with cirrhosis have been noted to have high plasma levels of endotoxin despite the absence of clinically apparent infection/64 This finding, coupled with recognition of the similarity between the hemodynamic alterations of endotoxemia and cirrhosis, led Vallance and Moncada 165 to hypothesize that NO was the mediator of the hemodynamic alterations of cirrhosis. Methylene blue, an antagonist of NO, has been shown to increase blood pressure in patients with hypotension due to liver failureJ 66 In a rat model of cirrhosis, L-NMMA increased blood pressure, decreased car- diac output, and increased systemic vascular resistance. 167

298 THE AMERICAN JOURNAL OF SURGERY ® VOLUME 170 SEPTEMBER 1995

Though these results are intriguing, it is by no means clear that endotoxin is the trigger for the hyperdynamic state seen in cirrhosis. It seems likely that NO has a role in the he- modynamic alterations of cirrhosis, but the exact nature of that role is yet unclear.

Hepatotoxicity Nitric oxide may be a potentially important paracrine me-

diator of function in the diseased liver. 168A69 West et aP 6s hy- pothesized that alterations in hepatocellular function may arise via interactions between hepatocytes and Kupffer cells, the specialized macrophages that line hepatic sinusoids. They demonstrated that when gentamicin-killed E coli or LPS was added to co-cultures of hepatocytes and Kupffer cells, but not hepatocytes alone, there was a marked decrease in hepatocyte protein synthesis, t6s,169 The Kupffer-cell me- diated decrease in protein synthesis was dependent on me- tabolism of L.Arg and associated with the production of ni- trates, nitrites, and t.-citrulline. 17° Kupffer cells were demonstrated to produce N O with a time course similar to that required for reduction of hepatocyte protein synthe- sis. 171'172 Finally, addition of N O or NO-generating com- pounds to hepatocytes inhibited protein synthesis in a time- dependent and reversible fashionJ 2z Total mRNA was not affected, indicating that NO acts at a translational or post- translational step.

A second source of NO that alters hepatocyte protein syn- thesis may be the hepatocyte itself. Curran et a1173 demon- strated that products from endotoxin-stimulated Kupffer cells were capable of stimulating the conversion of L-Arg to nitrites and nitrates within hepatocytes, and this was asso- ciated with a concurrent decrease in total protein synthesis. The mechanism by which N O modulates hepatocyte pro- tein synthesis remains unclear. It does not involve cell death or increases in cGME ~z2 Although N O inhibits mitochon- drial respiration, the effects last only I to 2 hours, and there- fore cannot be responsible for the 12-hour persistent inhi- bition of hepatocyte protein synthesis. 174

CONCLUSION Nitric oxide is a unique biologic mediator that has been im-

plicated in many physiologic and pathophysiologic states. It is an important regulator of vascular tone an d may be involved in sepsis, coagulation, immune function, inhibitory innerva- tion of the gastrointestinal tract, protection of gastrointesti- nal mucosa, and the hepatotoxicity of cirrhosis. Because N O may represent a point of control or intervention in a number of disease states, surgeons should be aware of the scientific and clinical aspects of this important molecule.

REFERENCES 1. Lancaster JR, Langrehr JM, Bergonia HA, et al. EPR detection of heme and nonheme iron-containing protein nitrosylation by nitric ox- ide during rejection of rat heart altograft. J Biol Chem. 1992;267: 10994-10995. 2. Pahner RMJ, Rees DD, Ashton PS, Moncada S. L-arginine is the physiologic precursor for the formation of nitric oxide in endothelium dependent relaxation. Biochem Biophys Res Commun. 1988;153: 1251-1256. 3. Leone AM, Palmer RMJ, Knowles RG, et al. Constitutive and in- ducible nitric oxide synthases incorporate molecular oxygen into both

I OVERVIEW OF NITRIC OXIDE/RODEBERG ET AL

nitric oxide and citrulline. J Biol Chem. 1991;266(35):23790-23795. 4. Moncada S, Higgs EA. Endogenous nitric oxide: physiology, pathol- ogy and clinical relevance. Eur J Clin Invest. 1991;21:361-374. 5. Bredt DS, Snyder SH. Nitric oxide, a novel neuronal messenger. Neuron. 1992;8:3 11. 6. Furchgott RF, gawadzki JV. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature. 1980;288:373-376. 7. )kngg~rd EE. Endogenous and exogenous nitrates. Acta Anaesthesiol Scand. 1992;36:7-10. 8. Miki W, Kawabe Y, Kuriyama K. Activation of cerebral guanylate cyclase by nitric oxide. Biochem Biophys Res Commun. 1977;75: 851-856. 9. Katsuki S, Arnold W, Mittal C, Murad E Stimulation of guanylate cyclase by sodium nitroprusside, nitroglycerin and nitric oxide in var- ious tissue preparations and comparison to the effects of sodium azide and hydroxylamine. J Cyclic NucIeotide Res. 1977;3:23-35. 10. Holzmann S. Endothdium-induced relaxation by acetylcholine as- sociated with larger rises in cyclic GMP in coronary arterial strips. J Cyclic Nucleotide Res. 1982;8:409-419. 11. Rapoport RM, Murad E Agonist-induced endothelium-dependent relaxation in rat thoracic aorta may be mediated through cGMP. Circ Res. 1983;52(3):352-357. 12. Murad E Cyclic guanosine monophosphate as a mediator of va- sodilation. J Clin Invest. 1986;78:1-5. 13. Ignarro LJ, Buga GM, Wood KS, et al. Endothelium-derived re- laxing factor produced and released from artery and vein is nitric ox- ide. Proc Natl Acad Sci USA. 1987;84:9265-9269. 14. Palmer RMJ, Ferrige AG, Moncada S. Nitric oxide release ac- counts for the biological activity of endothelium-derived relaxing fac- tor. Nature. 1987;327:524-526. 15. Flavahan NA, Vanhoutte PM. G proteins and endothelial re- sponses. Blood Vessels. 1990;27:218-229. 16. Pohl U, Busse R, Kuon E, Bassenge E. Pulsatile perfusion stimu- lates the release of endothelial autocoids. J Appt Cardiol. 1986;1: 215-235. 17. Rubanyi GM, Romero JC, Vahoutte PM. Flow induced release of endothelium derived relaxing factor. Am J Physiol. 1986;250: H1145-H 1149. 18. Ignarro LJ, Lippton H, Edwards JC, et al. 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 Pharma~ol Exp Ther. 1981;218:739-749. 19. Stamler JS, Simon DI, Osborne JA, et al. S-Nitrosylation of pro- teins with nitric oxide: synthesis and characterization of biologically active compounds. Proc Natl Acad Sci USA. 1992; 89:444-448. 20. Fukuto JM, Wallace GC, Hszieh R, Chaudhuri G. Chemical ox- idation of N-hydroxyguanidine compounds. Biochem Pharmacol. 1992;43:6074513. 21. Rosenblum WI. Endothelium-derived relaxing factor in brain blood vessels is not nitric oxide. Stroke. 1992;23(10):1527-1532. 22. Palmer RMJ. The discovery of nitric oxide in the vessel wall. Arch Surg. 1993;128:39(>401. 23. Keely SL, Lincoln TM, Corbin JP. Interaction of acetylcholine and epinephrine on heart cyclic AMP-dependent protein kinases. Am J Physiol. 1978;234:H432-H438. 24. Fujino A, Nakaya S, Wakatsuki T, et al. Effects of nitroglycerin on ATP-induced Ca++-mobilization, Ca++-activated K channels and contraction of cultured smooth muscle cells of porcine coronary artery. J PharmacoI Exp Ther. 1991;256:371-377. 25. Furchgott RF, Vanhoutle PM. Endothelium-derived relaxing and contracting factors. FASEB J. 1989;3:2007-2018. 26. Moncada S, Radomski MW, Palmer RM. Endothelium derived re- laxing factor: identification as nitric oxide and role in the control of vascular tone and platelet function. Biochem Pharmacol. 1988;37: 2495-2501. 27. Ignarro LJ. Biosynthesis and metabolism of endothelium-derived

THE AMERICAN JOURNAL OF SURGERY ® VOLUME 170 SEPTEMBER 1995 299

OVERVIEW OF NITRIC OXIDE/RODEBERG ET AL I

nitric oxide. Annu Rev Pharmacol Toxicol. 1990;30:535-560. 28. Gryglewski RJ, Botting RM, Vane JR. Mediators produced by the endothelium. Hypertension. 1988; 12:530-548. 29. Bath PMW, Hassel DG, Gladwin AM, et al. Nitric oxide and prostacyclin: divergence of inhibitory effects on monocyte chemotaxis and adhesion to endothelium in Vitro. Arterioscler Thromb. 1991;11: 254-260. 30. Rees DD, Pahner RM, Moncada S. Role of endothelium-denved nitric oxide in the regulation of blood pressure. Proc Natl Acad Sci USA. 1989;86:3375-3378. 31. Vallance P, Collier J, Moncada S. Effects of endothelium-derived nitric oxide on peripheral arteriolar tone in man. Lancet. 1989;ii:997-1000. 32. Gardiner SM, Compton AM, Bennett T, et al. Control of regional blood flow by endothelium-derived nitric oxide. Hypertension. 1990; 15:486-492. 33. Ekelund U, Mellander S. Role of endothelium-derived nitric ox- ide in the regulation of tonus in large-bore arterial resistance vessels, arterioles and veins in cat skeletal muscle. Acta Physiol Scand. 1990; 140:301-309. 34. Kukovetz WR, Holzmann S. Mechanism of nitrate induced va- sodilatation and tolerance. Z Kardiol. 1983;72:14-19. 35. Axellsson KL, Andersson RG. Tolerance towards nitroglycerin in- duced in vivo is correlated to a reduced cGMP response and an al- teration in cGMP turnover. Eur J Pharmacol. 1983;88:71-79. 36. Waldman SA, Rapoport RM, Ginsburg R, Murad E Desensitization to nitroglycerin in vascular smooth muscle from rat and human. Biochem Pharmacol. 1986;35:3525-3531. 37. Amezcua JL, Palmer RMJ, deSouza BM, et aL Nitric oxide syn- thesized from L-arginine regulates vascular tone in the coronary cir- culation of the rabbit. Br J Pharmacol. 1989;97:1119-1124. 38. Tolins JP, Palmer RMJ, Moncada S, et al. Role of endothelium- derived relaxing factor (EDRF) in regulation of renal hemodynamic responses. Am J Physiol. 1990;258:H655-H662. 39. Persson MG, Gustafsson LE, Wiklund NP, et al. Endogenous ni- tric oxide as a probable modulator of pulmonary circulation and hy- poxic pressor response in vivo. Acta Physiol Scand. 1990;140:449-457. 40. Vila J, Esplugues JV, Martinez-Cuesta MA, et al. NQMonomethyl- L-arginine and NO-nitro-L-arginine inhibit endothelium-dependent re- laxations in human isolated omental arteries.J Pharm Pharmacol. 1991; 43:869-870. 41. Salom MG, Lahera V, Miranda-Guardiola F, Romero JC. Blockade of pressure natriuresis induced by inhibition of renal synthesis of ni- tric oxide in dogs. Am J Physiol. 1991;31:F718-F722. 42. Panza JA, Quyumi AA, Brush JR Jr, Epstein SE. Abnormal en- dothelium-dependent vascular relaxation in patients with essential hy- pertension. NEJM. 1990;323:22-27. 43. Linder L, Kiowski W, Fritz BR, Luscher TE Indirect evidence for release of endothelium-derived relaxing factor in human forearm cir- culation in vivo: EDRF response in essential hypertension. Circulation. 1990;81:1762-1767. 44. F6rstennaml U, Mugge A, Alheid U, et al. Selective attenuation of endothelium-mediated vasodilation in atherosclerotic human coro- nary arteries. Circ Res. 1988;62:185-190. 45. Ludmer PL, Selwyn AP, Shook TL, et al. Paradoxical vasocon- striction induced by acetylcholine in atherosclerosis coronary arteries. NEJM. 1986;315:1046-1051. 46. Winquist RJ, Bunting PB, Baskin EP, Wallace AA. Decreased en- dothelium dependent relaxation in New Zealand genetic hyperten- sive rats. J Hypertens. 1984;2:541-545. 47. Liischer TF, Raij L, Vanhoutte PM. Effect of hypertension and its reversal on endothelium dependent relaxations in the rat aorta. J Hypertens. 1987;5(suppl 5):$153~S155. 48. Otsuka U, Dipiero A, Hirt E, et al. Vascular relaxation and cGMP in hypertension. Am J Physiol. 1988;254:H163-H169. 49. Snyder SH, Bredt DS. Biological roles of nitric oxide. Sci Am. 1992;266:68-77. 50. Shimokawa H, Vanhoutte PM. Impaired endothelium-dependent

relaxation to aggregating platelets and related vasoactive substances in porcine coronary arteries in hypercholesterolemia and atheroscle- rosis. Circ Res. 1989;64:900-914. 51. Minor RL, Myers PR, Guerra R, et al. Diet induced atherosclero- sis increases the release of nitrogen oxides from rabbit aorta. J Clin Invest. 1990;86:2109-2116. 52. Nakki T, Hishikawa K, Suzuki H, et al. L-arginine-induced hy- potension. Lancet. 1990;336:696. Letter. 53. Cooke JP, Andon NA, Girerd XJ, et al. Arginine restores cholin- ergic relaxation of hypercholesterolemic rabbit aorta. Circulation. 1991;83:1057-1062. 54. Tanner FC, Noll G, Boulanger CM, L6scher TF. Oxidized low den- sity lipoproteins inhibit relaxations of porcine coronary arteries: role of scavenger receptor and endothelium derived nitric oxide. Circulation. 1991;83:2012-2020. 55. Shimokawa H, Flavahan NA, Vanhoutte PM. Loss of endothelial pertussis toxin sensitive G-protein function in atherosclerotic porcine coronary arteries. Circulation. 1991;83:652-660. 56. Flavahan NA. Atherosclerosis or lipoprotein-induced endothelial dysfunction. Circulation. 1992;85:1927-1938. 57. Barrett ML, Willis AL, Vane JR. Inhibition of platelet-derived mi- togen release by nitric oxide (EDRF). Agents Actions Suppl. 1989;27: 448-491. 58. Garg UC, Hassid A. Nitric oxide-generating vasodilators and 8- bromocyclic guanosine monosulphate inhibit mitogenesis and prolif- eration of cultured rat vascular smooth muscle Cells. J Clin Invest. 1989; 83:1774-1777. 59. Durante W, Sen AK, Sunahara FA. Impairment of endothelium dependent relaxation in aortae of spontaneous diabetic rats. Br J Pharmacol. 1988;94:463-468. 60. Kappagoda T, Jayakody L, Rajotte R, et al. Endothelium depen- dent relaxation to acetydidine in the aorta of streptozorocia induced diabetic rat and the BB diabetic rat. Clin Invest Med. 1989;12: 187-193. 61. Bucala R, Tracey KJ, Cerami A. Advanced glycosylation products quench nitric oxide and mediate defective endothelium dependent vasodilation in experimental diabetes. J Clin Invest. 1991;87:432-438. 62. Thiemermann C, Szab6 C, Mitchell JA, Vane JR. Vascular hy- poreactivity to vasoconstrictor agents and hemodynamic decompen- sation in hemorrhagic shock is mediated by nitric oxide. Proc Natl Acad Sci USA. 1993;90:267-271. 63. Fisher CM, Kistler JP, Davis JM. The correlation of cerebral vasospasm and the amount of subarachnoid blood detected by com- puterized cranial tomography after aneurysm. In: Wilkins RH, ed. Cerebral Artery Spasm. London/Baltimore: Williams & Wilkins; 1981: 397-408. 64. Lam JY, Cheseboro JH, Fuster V. Platelets, vasoconstriction and nitroglycerin during arterial wall injury. A new anti-thrombotic role for an old drug. Circulation. 1988;78:712-716. 65. DeCaterino R, Gianessi D, Bernini W, et al. In vivo actions of or- ganic nitrates on platelet function in humans. Z Kardiol. 1989;78(suppl 2):56-60. 66. Azuma H, Ishikawa M, Sekizaki S. Endothelium-dependent inhi- bition of platelet aggregation. Br J PharmacoI. 1986;88:411-415. 67. Mellion BT, Ignarro LJ, Ohlstein EH, et al. Evidence for the in- hibitory role of guanosine 3-,5-monophosphate in ADP-induced hu- man platelet aggregation in the presence of nitric oxide and related vasodilators. Blood. 1981;57:946-955. 68. Alheid U, Reichwehr I, F6rstermann U. Human endothelial cells inhibit platelet aggregation by separately stimulating platelet cyclic AMP and cyclic GMP. Eur J Pharmacol. 1989;164:103-110. 69. Radomski MW, Palmer RMJ, Moncada S. The anti-aggregating properties of vascular endothelium: interactions between prostacyclin and nitric oxide. Br J Pharmacol. 1987;92:639-646. 70. Radomski MW, Palmer RMJ, Moncada S. The role of nitric ox- ide and cOMP in platelet adhesion to vascular endothelium. Biochem Biophys Res Commun. 1987;148:1482-1489. 71. MacDonald PB, Read MA, Dusting GJ. Synergistic inhibition of

308 THE AMERICAN JOURNAL OF SURGERY ® VOLUME 170 SEPTEMBER 1995

platelet aggregation by endothelium-derived relaxing factor and prostacyclin. Thromb Res. 1988;49:437-449. 72. Korbut R, Lindbury PS, Vane JR. Prolongation of fibrinolytic ac- tivity of tissue plasminogen activator by nitrovasodilators. Lancet. 1990;335:669. Letter. 73. Radomski MW, Palmer RM, Moncada S. Glucocorticoids inhibit the expression of an inducible, but not the constitutive, nitric oxide synthase in vascular endothelial cells. Proc Natl Acad Sci USA. 1990; 87:10043-10047. 74. Tannenbaum SR, Fett D, Young VR, et al. Nitrite and nitrate formed by endogenous synthesis in the human intestine. Science. 1978; 200:1487-1489. 75. Wagner DA, Young VR, Tannenbaum SR, =et al. Mammalian ni- trate biochemistry: metabolism and endogenous synthesis. In: O'Neill IK, ed. N-nitroso Compounds! Occurrence, Biolo~cal Effects and Relevance to Human Cancer: Proceedings of the VIIIth International Symposium on N.nitroso Compounds; September 5-9, 1983; Banff, Canada. New York: Oxford University Press; 1984:15. IARC Scientific Publications. 76. Wagner DA, Young VR, Tannenbaum SR. Mammalian nitrate biosynthesis: incorporation of 15NH3 into nitrate is enhanced by endotoxin treatment. Proc Natl Acad Sci USA. 1983;80:4518-4521. 77. Iyengar R, Stuehr DJ, Marietta MA. Macrophage synthesis of ni- trite, nitrate, and N-nitrosamines: precursors and role of the respira- tory burst. Proc Natl Acad Sci USA. 1987;84:6369-6373. 78. Stuehr DJ, Marietta MA. Mammalian nitrate biosynthesis: mouse macrophages produce nitrite and nitrate in response to E coli LPS. Proc Natl Acad Sci USA. 1985;82:7738-7742. 79. Marietta MA, Yoon PS, Iyengar R, et al. Macrophage oxidation of L-arginine to nitrite and nitrate: nitric oxide is an intermediate. Biochemistry. 1988;27:8706-8711. 80. Kwon NS, Nathan CF, Stuehr DJ. Reduced biopterin as a cofac- tot in the generation of nitrogen oxides by murine macrophages. J Biol Chem. 1989;264:20496-20501. 81. Lyons CR, Orloff GH, Cunningham JM. Molecular cloning and functional expression of an inducible nitric oxide synthase from a murine macrophage cell line. J Biol Chem. 1992;267:637045374. 82. Xie Q-W, Cho HJ, Calaycay J, et al. Cloning and characterization of inducible nitric oxide synthase from mouse macrophages. Science. 1992;256:225-228. 83. Lowenstein CJ, Glatt CS, Bredt DS, Synder SH. Cloned and ex- pressed macrophage nitric oxide synthase contrasts with the brain en- zyme. Proc Natl Acad Sci USA. 1992;89:6711-6715. 84. Bredt DS, Hwang PM, Glatt CE, et al. Cloned and expressed ni- tric oxide synthase structurally resembles cytochrome P-450 reductase. Nature. 1991;351:714-718. 85. McCall TB, Feelish M, Palmer RM, Moncada S. Identification of N-iminoethyl-L-ornithine as an irreversible inhibitor of nitric oxide Synthase in phagocytic cells. Br J Pharmacol. 1991;102:234-238. 86. Nathan CE Nitric oxide as a secretory product of mammalian cells. FASEB J. 1992;6:3051-3064. 87. Nathan CF, Hibbs JB Jr. Role of nitric oxide synthesis in macrophage antimicrobial activity. Curt Opin Immunol. 1991;3:65-70. 88. Denis M. Tumor necrosis factor and granulocyte macrophage- colony stimulating factor stimulate human macrophages to restrict growth of virulent Mycobacterium avium and to kill avirulent M avium: killing effector mechanism depends on the generation of reactive ni- trogen intermediates. J Leukoc Biol. 1991;49:380-387. 89. Nussler AK, Di Silvio M, Billar TR, et al. Stimulation of the ni- tric oxide synthase pathway in human hepatocytes by cytokines and endotoxin. J Exp Med. 1992;176:261-264. 90. Stuehr DJ, Griffith OW. Mammalian nitric oxide synthases. Adv Enzymol Relat Areas Mol Biol. 1992;65:287-346. 91. Beckman JS, Beckman TW, Chen J, et al. Apparent hydroxyl radical production by peroxynitrite: implications for endothelial in- jury from nitric oxide and superoxide. Proc Natl Acad Sci USA. 1990;87: 1620-1624. 92. Hogg N, Darley-Usmar VM, Wilson MT, Moncada S. Production

I OVERVIEW OF NITRIC OXIDE/RODEBERG ET AL

of hydroxyl radicals from the simultaneous generation of superoxide and nitric oxide. Biochem J. 1992;281:419-424. 93. Liew FY, Parkinson C, Millot CS, et al. Tumor necrosis factor in Leishmaniasis. Immunology. 1990;69:570-577. 94. Titus RG, Kelso A, Louis JA. Intracellular destruction of L. trop. ica by macrophages activated with macrophage activating factor/in- terferon. Clin Exp Immunol. 1984;55:157-165. 95. Liew FY, Li Y, Millott S. Tumor necrosis factor synergizes with in- terferon gamma in mediating killing of Leishmania major through the induction of nitric oxide. J Immunol. 1990;145:4306-4310. 96. Green SJ, Mellouk S, Hoffman SL, et al. Cellular mechanisms of nonspecific immunity to intracellular infection: cytokine-induced syn- thesis of toxic nitrogen oxides from L-arginine by macrophages and hepatocytes. Immunol Lett. 1990;25:15-20. 97. Munoz-Fernandez MA, Fernandez MA, Fresno M. Synergism be- tween tumor necrosis factor-alpha and interferon-gamma on macro- phage activation for the killing of intracellular Trypanosoma cruzi through a nitric oxide-dependent mechanism. EurJ lmmunol. 1992;22: 301-307. 98. Klostergaard J, Leroux ME, Hung MC. Cellular models of macro- phage tumorocidal effector mechanisms in vitro. J Immunol. 1991;147: 2802-2808. 99. Jiang H, Stewart CA, Fast DJ, Leu RW. Tumor target-derived soluble factor synergizes with interferon 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: 2137-2146. 100. Efron DT, Kirk SJ, Regan MC, et al. Nitric oxide generation from t-arginine is required for optimal human peripheral blood lymphocyte DNA synthesis. Surgery. 1991;110:327-334. 101. Hadden JW, Coffey RG, Ananthakrishnana R, Hadden EM. Cyclic nucleotides and calcium in lymphocyte regulation and activa- tion. Ann NY.Acad Sci. 1979;332:241-254. 102. Albina JE, Abate JA, Henry WL. Nitric oxide production is re- quired for murine resident peritoneal macrophages to suppress mito- gen-stimulated T cell proliferation. J Immunol. 1991;147: 144-148. 103. Liew FY, Li Y, Severn A, et al. A possible novel pathway of reg- ulation by murine T helper type 2 cells of a T helper type 1 cell ac- tivity via the modulation of induction of nitric oxide synthase on macrophages. Eur J Immunol. 1991;21:2489-2494. 104. Langrehr JM, Hoffman RA, Billar TR, et al. Nitric oxide syn- thesis in the in vivo allograft response: a possible regulatory mecha- nism. Surgery. 1991;110:335-341. 105. Langrehr JM, Muller AR, Markus PM, et al. FK 506 il~hibits ni- tric oxide production by cells infiltrating sponge matrix allografts. Transplant Proc. 1991;23:3260-3261. 106. Langrehr JM, Muller AR, Bergonia HA, et al. Detection of ni- tric oxide by electron paramagnetic resonance spectroscopy during re- jection and graft-versus-host disease after small bowel transplantation in the rat. Surgery. 1992;112:395-401. 107. Langrehr JM, Murase N, Markus P, et al. Nitric oxide production in host-versus-grafr and graft-versus-host reactions in the rat. J Clin Invest. 1992;90:679-683. t08. Garside P, Hutton AK, Severn A, et al. Nitric oxide mediates intestinal pathology in graft-versus-host disease. EurJ Immunol. 1992; 22:2141-2145. 109. Kilboum RG, Jubran A, Gross SS, et al. Reversal of endotoxin- mediated shock by NC-methyl-L-arginine, an inhibitor of nitric oxide synthesis. Biochem Biophys Res Commun. 1990;172:1132-:1138. 110. Kilboum RG, Gross SS, Jubran A, et al. NO-methyl-L-arginine inhibits tumor necrosis factor-induced hypotension: implications for the involvement of nitric oxide. Proc Natl Acad Sci USA. 1990;87: 3629-3632. 111. Westemberger U, Thanner S, Ruf HH, et al. Formation of free radicals and nitric oxide derivative of hemoglobin in rats during shock syndrome. Free Rad Res Comm. 1990;11:167-178. 112. Lorente JA, Landin L, Penes E, et al. Role of nitric oxide in the hemodynamic changes of sepsis. Crit Care Med. 1993;21:759-767.

THE AMERICAN JOURNALOF SURGERY ~ VOLUME 170 SEPTEMBER 1995 :301

OVERVIEW OF NITRIC OXIDE/RODEBERG ET AL ]

113. Ochoa JB, Udekwu AO, Billiar TR, et al. Nitrogen oxide levels in patients after trauma and during sepsis. Ann Surg. 1991;214: 621-626. 114. Lorente JA, Landin L, DePablo R, et al. L-Arginine pathway in the sepsis syndrome. Crit Care Med. 1993;21:1287-1295. 115. Petros A, Bennett D, Vallance P. Effect of nitric oxide synthase inhibitors on hypotension in patients with septic shock. Lancet. 1991; 338:1557-1558. 116. Aoki N, Johnson G, Lefer AM. Beneficial effects of two forms of NO administration in feline splanchnic artery occlusion shock. Am J Physiol. 1990;258:G275-G280. 117. Ma XL, Johnson G, Lefer AM. Mechanism of inhibition of ni- tric oxide production in a murine model of splanchnic artery occlu- sion shock. Arch In Pharmacodyn Ther. 1991;311:89-103. 118. Wright CE, Rees DD, Moncada S. Protective and pathological roles of nitric oxide in endotoxin shock. Cardiovasc Res. 1992;26: 48-57. 119. Klabunde RE, Helgren MC. Cardiovascular actions of N~-methyl-L-arginine are abolished in a canine shock model using high dose endotoxin. Res Commun Chem Pathol Pharmacol. 1992; 78:57-68. 120. Harbrecht BG, Billiar TR, Stadler J, et al. Nitric oxide serves to reduce hepatic damage during murine endotoxemia. Crit Care Med. 1992;20:1568-1574. 121. Curran RD, Billiar TR, Stuehr DJ, et al. Multiple cytokines are required to induce hepatic nitric oxide production and inhibit total protein synthesis. Ann Surg. 1990;212:462-471. 122. Curran RD, Ferrari FK, Kispert PH, et aL Nitric oxide and nitric oxide-generating compounds inhibit hepatocyte protein synthesis. FASEB J. 1991;5:2085-2092. 123. Ballingand J, Ungureanu D, Kelly RA, et al. Abnormal contrac- tile function due to induction of nitric oxide synthesis in rat cardiac myowtes follows exposure to activated macrophage-conditioned medium. J Clin Invest. 1993;91:2314-2319. 124. Lieberthal W, McGarry J, Sheils J, et al. Nitric oxide inhibition in rats improves blood pressure and renal function during hypovolemic shock. Am J Physiol. 1991;261:F868-F872. 125. Stark ME, Szurszewski JH. Role of nitric oxide in gastrointesti- nal and hepatic function and disease. Gastroenterology. 1992;103: 1928-1949. 126. Sanders KM, Ward SM. Nitric Oxide as a mediator of nona- drenergic noncholinergic neurotransmission. Am J Physiol. 1992;262: G379~G392. 127, Hoyle CHV, Bumstock G. Neuromuscular transmission in the gastrointestinal tract. In: Rauner B, Wood J, Schultz S, eds. Handbook of Physiology. The Gastrointestinal System. Motility and Circulation. Bethesda, Maryland: American Physiological Society; 1989:435-464. 128. Buk H, Boeckxstaens GE, Pelckmans PA, et al. Nitric oxide as an inhibitory non-adrenergic non-cholinergic neurotransmitter. Nature. 1990;345:346-347. 129. Boeckxstaens GE, Pelckmans PA, Bogers JJ, et al. Release of ni- tric oxide upon stimulation of nonadrenergic noncholinergic nerves in the rat gastric fundus. J Pharmacol Exp Ther. 1991;256:441-447. 130. Tottrup A, Svane D, Forman A. Nitric oxide mediating NANC inhibition in opossum lower esophageal sphincter. AmJ Physiol. 1991; 260:G385-G389. 131. Boeckxstaens GE, Pelckmans PA, Bult H, et al. Nonadrenergic noncholinergic relaxation mediated by nitric oxide in the ileocolonic junction. Eur J Pharmacol. 1990; 190:239-246. 132. Ozaki H, Blondfield DP, Hori M, et al. Spontaneous release of nitric oxide inhibits electrical, Ca > and mechanical transients in ca- nine gastric smooth muscle. J Physiol (Lord). 1992;445:231-247. 133. Desai KM, Sessa WC, Vane JR. Involvement of nitric oxide in the reflex relaxation of the stomach to accommodate food or fluid. Nature. 1991;351:477-479. t34. Toda N, Baba H, Okamura T. Role of nitric oxide in nonadren- ergic, noncholinergic nerve-mediated relaxation in dog duodenal lon- gitudinal muscle strips. Jpn J Pharmacol. 1990;53:281-284.

135. Gustafusson LE, Wiklund CU, Wiklund NP, et al. Modulation of autonomic neuroeffector transmission by nitric oxide in guinea pig ileum. Biochem Biophys Res Commun. 1990;173:106-110. 136. Kaufman HS, Shermak MA, May CA, et al. Nitric oxide inhibits resting sphincter of Oddi activity. Am J Surg. 1993;165:74-80. 137. Thombury KD, Ward SM, Dalziel HH, et al. Nitric oxide and nitrosocysteine mimic nonadrenergic, noncholinergic hyperpolariza- tion in canine proximal colon. AmJ Physiol. 1991;261:G553~;557. 138. Dalziel HH, Thombury KD, Ward SM, Sanders KM. Involvement of nitric oxide synthetic pathway in inhibitory junction potentials in canine proximal colon. Am J Physiol. 1991;260: G789-G792. 139. Rattan S, Chakder S. Role of nitric oxide as a mediator of in- ternal anal sphincter relaxation. AmJ Physiol. 1992;262:G107-G112. 140. Gillespie JS, Liu X, Martin W. The effects of L-arginine and NC-monomethyt L-arginine on the response of the rat anococcy- geus muscle to NANC nerve stimulation. Br J Pharmacol. 1989;98: 1080-1082. 141. Burleigh DE. NC-Nitro-L-arginine reduces nonadrenergic, non- cholinergic relaxations of human gut. Gastroenterology. 1992;102: 679-683. 142. Stark ME, Bauer AJ, Sarr MG, Szurszewski JH. Nitric Oxide me- diates inhibitory nerve input in human and canine jejunum. Gastroenterology. 1993;104:398-409. 143. Bredt DS, Hwang PM, Snyder SH. Localization of nitric oxide synthase indicating a neural role for nitric oxide. Nature. 1990;347: 768-770. 144. Ward SM, Xue C, Shuttleworth CW, et al. NADPH diaphorase and nitric oxide synthase colocalization in enteric neurons of canine proximal colon. Am J Physiol. t992;263:G277-G284. 145. Boeckxstaens GE, Pelckmans PA, Bult H, et al. Evidence for ni- tric oxide as mediator of non-adrenergic, non-cholinergic relaxations induced by ATP and GABA in the canine gut. Br J Pharmacol. 1991;102:434-438. 146. Burnstock G, Satchell DG, Smythe AA. A comparison of the excitatory and inhibitory effects of non-adrenergic, non-cholinergic nerve stimulation and exogenously applied ATP on a variety of smooth muscle preparations from different vertebrate species. Br J Pharmacol. 1972;46:234-242. 147. Goyal RK, Rattan S, Said S. VIP as a possible neurotmnsmitter of non-cholinergic non-adrenergic inhibitory neurons. Nature. t980; 288:378-380. 148. Grider JR, Murthy KS, Jin JG, Makhlouf GM. Stimulation of ni- tric oxide from muscle cells by VIP: prejunctional enhancement of VIP release. Am J Physiol. 1992;262:G774-G778. 149. Stark ME, Bauer AJ, Szurszewski-JH. Effect of nitric oxide on cir- cular muscle of the canine small intestine. J Physiol (Lond). 1991;444: 743-761. 150. Mersereau WM, Hinchey EJ. Effect of gastric acidity on gastric ulceration induced by hemorrhage in the rat utilizing a gastric cham- ber technique. Gastroenterology. 1973;64:1130-1135. 151. Ritchie WE Shearburn IlI EW. Influence of isoproterenol and cholestyramine on acute gastric mucosal ulcerogenesis. Gastro- enterology. 1977;73:62-65. 152. Whittle BJR, Boughton-Smith NK, Moncada S. Biosynthesis and role of the endothelium-derived vasodilator, nitric oxide, in the gas- tric mucosa. Ann N Y Acad Sci. 1992;664:126-139. 153. Pique JM, Whittle BJR, Espluges JV. The vasodilator role of en- dogenous nitric oxide in the rat gastric microcirculation. Eur J Pharmacol. 1989;174:293-296. 154. Pique JM, Espluges JV, Whittle BJR. Endogenous nitric oxide as a mediator of gastric mucosal vasodilation during acid secretion. Gastroenterology. 1992;102:168-174. 155. Whittle BJR, Lopez-Belamonte J, Moncada S. Regulation of gas- tric mucosal integrity by endogenous nitric oxide: interactions with prostanoids and sensory neuropeptides in the rat. Br J Pharmacol. 1990;99:607-611. 156. MacNaughton WK, Cirino G, Wallace JL. Endothelium-derived

302 THE AMERICAN JOURNAL OF SURGERY ® VOLUME 170 SEPTEMBER 1995

relaxing factor (nitric oxide) has protective actions in the stomach. Life Sci. 1~J89;45:1869-1876. 157~ Deitch EA. Bacterial translocation of the gut flora. J Trauma. 1990;30(suppl 12):S184~q189. 158. Meakins JL, Marshal JC. The gastrointestinal tract: the "motor" of MOE Arch Surg. 1986;121:197-201. 159. Hutcheson IR, Whittle BJR, Boughton-Smith NK. Role of ni- tric oxide in maintaining vascular integrity in endotoxin-induced acute intestinal damage in the rat. BrJ Pharmacol. 1990;101:815-820. 160. Boughton-Smith NK, Hutcheson IR, Deakin AM, et al. Protective effect of S-nitroso-N-acetyl-penicillamine in endotoxin-induce acute intestinal damage in the rat. Eur J Pharmac& 1990;191: 485-488. 161. Kubes P. Nitric oxide modulates epithelial permeability in the fe- line small intestine. Am J Physiol. 1992;262:G1138-G1142. 162. Murray JF, Dawson AM, Sherlock S. Circulatory changes in chronic liver disease. AmJ Med. 1958;32:358-367. 163. Parrillo JE, Parker MM, Natanson C. Septic shock in human. Advances in the understanding of pathogenesis, cardiovascular dys- function and therapy. Ann Intern Med. 1990;113:227-242. 164. Lumsden AB, Henderson J M, Kumer MH. Endotoxin levels mea- sured by a chromogenic assay in portal, hepatic, and peripheral blood in patients with cirrhosis. Hepatology. 1988;8:232-236. 165. Vallance P, Moncada S. Hyperdynamic circulation in cirrhosis: a role for nitric oxide? Lancet. 1991;337:776-778. 166. Midgley S, Grant IS, Haynes WG, Webb DJ. Nitric oxide in liver failure. Lancet. 1991;338:1590. Letter.

OVERVIEW OF NITRIC OXIDE/RODEBERG ET AL

167. Pizcueta P, Pique JM, Fernandez M, et al. Modulation of the hy- perdynamic circulation of cirrhotic rats by nitric oxide inhibition. Gastroenterology. 1992;103:1909-1915. 168. West MA, Keller GA, Cerra FB, Simmons RL. Killed Escherichia coli stimulates macrophage-mediated alterations in hepatocellular function during in vitro coculture: a mechanism of altered liver func~ tion in sepsis. Infect Immun. 1985;49:563-570. 169. West MA, Billiar TR, Mazuski JE, et al. Endotoxin modulation of hepatocyte secretory and cellular protein synthesis is mediated by Kupffer cells. Arch Surg. 1988;123:1400-1405. 170. Billiar TR, Curran RD, Stuehr DJ, et al. An >-Arginine-depen- dent mechanism mediates Kupffer cell inhibition of hepatocyte pro- tein synthesis in vitro. J Exp Med. 1989;169:1467-1472. 171. Billiar TR, Curran RD, Stuehr DJ, et al; Evidence that activa- tion of Kupffer cells results in production of L-arginine metabolites that release cell-associated iron and inhibit hepatocyte protein syn- thesis. Surgery. 1989;106:364-372. 172. Billiar TR, Curran RD, Ferrari FK, Williams DL. Kupffer cell: hepatocyte cocultures release nitric oxide in response to bacterial en- dotoxin. J Surg Res. 1990;48:349-353. 173. Curran RD, Billiar TR, Stuehr DJ, et al. Hepatocytes produce nitrogen oxides from L-arginine in response to inflammatory products of Kupffer cells. J Exp Med. 1989;170:1769-1774. 174. Stadler J, Billiar TR, Curran RD, et al. Effect of exogenous and endogenous nitric oxide on mitochondrial respiration of rat hepato- cytes. Am J Physiol. 1991;260:C91~C916.

THE AMERICAN JOURNAL OF SURGERY ~ VOLUME 170 SEPTEMBER 1995 303