Nitric oxide in gastrointestinal health and disease

Nitric oxide in gastrointestinal health and disease

GASTROENTEROLOGY 2004;126:903–913 SPECIAL REPORTS AND REVIEWS Nitric Oxide in Gastrointestinal Health and Disease VIJAY SHAH,*,‡ GREG LYFORD,§ GREG G...

149KB Sizes 0 Downloads 100 Views

GASTROENTEROLOGY 2004;126:903–913

SPECIAL REPORTS AND REVIEWS Nitric Oxide in Gastrointestinal Health and Disease VIJAY SHAH,*,‡ GREG LYFORD,§ GREG GORES,‡ and GIANRICO FARRUGIA§ *GI Research Unit, §Enteric Neuroscience Program, ‡Advanced Liver Disease Study Group, Mayo Clinic and Foundation, Rochester, Minnesota

Nitric oxide is an intracellular and intercellular messenger with important functions in a number of physiologic and pathobiologic processes within gastroenterology and hepatology, including gastrointestinal tract motility, mucosal function, inflammatory responses, gastrointestinal malignancy, and blood flow regulation. Since the broad review of this topic in GASTROENTEROLOGY more than 10 years ago, a number of advances have been made in the area of NO biology and its relevance to the gastrointestinal system.1 The aim of this review is to focus on our expanded understanding of the role NO plays in human gastrointestinal and hepatic physiology and disease processes by drawing on data from relevant in vitro and animal models as well as observational human studies.

itric oxide (NO) is a free radical signaling molecule generated by a family of P450-like enzymes, termed “NO synthases” (NOS), which generate this molecule through a series of regulated electron transfer steps. Three independent genes encoding neuronal,2 endothelial,3 and inducible NOS4 (nNOS, eNOS, and iNOS, respectively) have been cloned, each with the capacity to generate NO through both complimentary and distinct regulatory mechanisms. Though probably short-lived in biological systems, the lipophilic nature of NO allows it to diffuse quickly, thereby initiating intercellular and intracellular signals.5 The most characterized downstream NO signaling pathway relates to the soluble guanylate cyclase pathway, through which NO was shown to be identical to the previously postulated endothelial relaxing factor.5 Via this pathway, NO binds to the guanylate cyclase heme moiety thereby stimulating the enzyme to generate cGMP, which in turn activates protein kinase G (PKG), with downstream phosphorylation cascades leading to effector functions (Figure 1).6 However, in pathophysiologic conditions and probably during certain normal physiologic conditions, NOmediated signaling may also be independent of guanylate cyclase activation. For example, NO directly regulates

N

the function of ion channels, enzymes, and a number of other proteins.7 This regulation appears to occur in part through the nitrosylation of cysteine thiols in target proteins.8 NO may also contribute to cell damage in inflammatory conditions via nitration reactions.9 It has an unmatched electron and as a free radical can scavenge the more reactive superoxide, thereby generating peroxynitrite, which in turn may be responsible for nitration of protein tyrosine residues, mitochondrial energy depletion, and induction of DNA strand breaks.9 It is important to note that these cyclic guanosine monophosphate (cGMP)-dependent and independent actions, though attributed to NO, may occur in varying degrees through alternative nitrogen species and chemistry pathways and are therefore still an area of active investigation.9 These NO signaling pathways contribute to a number of physiologic functions in the gastrointestinal (GI) system including motility and vascular regulation, as well as the disease processes inherent to these functions. While the influence of NO on GI motility is largely controlled by nNOS, NO effects on vascular function in the GI system occurs largely under the purview of eNOS. Additionally, iNOS derived NO production contributes to cellular inflammation and the carcinogenic process. The stimulatory and inhibitory influence of NO on these broad processes of motility, vascular function, and inflammation are outlined below.

Gastrointestinal Motility Biology of nNOS The most prominent role of nNOS-derived NO in GI motility is as an inhibitory nonadrenergic noncholinergic (NANC)10,11 smooth muscle cell relaxant via Abbreviations used in this paper: COX, cyclooxygenase; GMP, guanosine monophosphate; IRI, ischemia–reperfusion injury; NANC, nonadrenergic noncholinergic; NO, nitric oxide; NOS, nitric oxide synthase; PGK, protein kinase G. © 2004 by the American Gastroenterological Association 0016-5085/04/$30.00 doi:10.1053/j.gastro.2003.11.046

904

SHAH ET AL.

GASTROENTEROLOGY Vol. 126, No. 3

VIP as upstream as well as downstream of nNOS derived-NO generation, thus the interrelationship of NO and VIP remains under investigation.20,21 The diatomic gas and alternative guanylate cyclase binding partner, carbon monoxide, is proposed as a cotransmitter of nNOS-derived NO,17 and crosstalk between the NO and carbon monoxide signaling pathways occurs at multiple levels including transcriptional and posttranslational.22,23 The role of nNOS-derived NO in esophageal, gastric, and intestinal motility and motor disorders is described below. Esophageal Motility and Motor Disorders Figure 1. Nitric oxide signaling and targets for potential therapeutic intervention. The free radical gas, NO, is generated by the NOS isoforms, which convert L-arginine to L-citrulline and NO through a series of enzymatic steps that involve electron transfer and hydroxylation. The primary target of NO is soluble guanylate cyclase. NO binds and activates guanylate cyclase, thereby enhancing the conversion of GTP to cGMP. cGMP acts on its kinase, protein kinase G, thereby enabling a number of downstream physiologic actions including vasodilation, NANC, and inhibition of platelet aggregation. NO interactions with superoxide anion (O2⫺), result in the generation of the oxidative molecule, peroxynitrite (ONOO⫺). NO interactions with protein thiols also appear to regulate a number of target enzymes and proteins (not shown). Sites of potential therapeutic interventions by which to activate or inhibit NO signaling pathways are shown. Potentiation of NO signaling (depicted in squares) may be achieved by providing excess arginine substrate, enhancing signaling pathways that activate NOS function, NO donors, antioxidants, or phosphodiesterase (PDE) inhibitors. Inhibition of NO signaling (depicted in circles) may be achieved by competitive and allosteric inhibitors of NOS activity and inhibitors of guanylate cyclase (GC) function.

nNOS-derived activation of smooth muscle cell guanylate cyclase. Nerves throughout the length of the luminal GI tract express nNOS with highest levels in the pyloric sphincter,12 particularly the soma and processes of myenteric nerves.13 Although initial reports of nNOS were specific to neurons,13 evidence is emerging that nNOS is widely distributed. Six splice variants of nNOS have been described in the luminal GI tract14; the functional significance and differential distribution of these splice variants within the GI tract remains to be determined. Other transmitter molecules such as vasoactive intestinal peptide (VIP),15 adenosine triphosphate,16 and carbon monoxide17 also function in conjunction with NO as NANC inhibitory signals. For example, VIP colocalizes with NOS and appears to have similar inhibitory functions on smooth muscle in the gut. However, VIP possesses relaxing effects in the gastric fundus that is retained in nNOS⫺/⫺, eNOS⫺/⫺, and iNOS⫺/⫺ mice as well as cGMP kinase 1⫺/⫺ mice, suggesting parallel and redundant pathways.18,19 Other studies have targeted

Data from human studies indicate that NO inhibits esophageal smooth muscle function. In normal human subjects, systemic administration of a recombinant human hemoglobin that scavenges NO promotes increased velocity of peristaltic contractions, simultaneous contractions, spontaneous contractions associated with chest pain, and diminished lower esophageal sphincter (LES) relaxation.24 Pharmacologic blockade of NO synthesis reduces the latency between swallows, contraction in the distal esophagus, increases basal LES pressure, increases peristaltic wave pressure, and decreases the number of transient LES relaxations.25 These and other studies suggest that deficient NO may cause esophageal dysmotility. A role for NO in achalasia is suggested by the absence of NOS immunoreactivity and enzymatic activity in LES neurons of patients with achalasia.26 A causative role for deficient NO in achalasia is supported by the phenotype of nNOS⫺/⫺ mice, which exhibit elevated baseline LES pressures and reduced swallow-induced relaxation of the LES27 in contradistinction to eNOS⫺/⫺ mice, which display relaxation responses similar to control animals.28 Therapy for achalasia using NO donors in humans, however, provides only minimal success,29 perhaps because of the inability to provide selective and adequate NO delivery to the target site. Potentiation of the NO– cGMP pathway in human esophageal motor disease has also been attempted using sildenafil, the phosphodiesterase type-5 inhibitor, which decreases swallow-induced contractions in the distal esophagus.30 Although patients with achalasia reported no benefit, chronic administration was associated with symptomatic improvement in some individuals with nutcracker esophagus, esophageal spasm, and hypertensive LES.30 Thus, although NO is critical for NANC-inhibitory signaling and sphincter relaxation in esophagus, more studies will be required to establish the efficacy of NO– cGMP pathway activation as a therapeutic target for achalasia.

March 2004

Gastric Motility and Motor Disorders NO promotes gastric accommodation and emptying. In humans, pharmacologic inhibitors of NOS increase frequency of gastric contractions, decrease gastric emptying time, and decrease fundic volume both basally and after meals,31 while NO donors slow gastric emptying and improve accommodation of the proximal stomach.32 NO has also been shown to promote fundic accommodation in animal studies.33 These studies have resulted in preliminary studies on nitrate therapy for functional dyspepsia,34,35 a condition that evidences defective fundic accommodation in a subset of patients. Data to date suggest that NO donors are able to effectively relax the stomach, but the efficacy in functional dyspepsia remains uncertain because symptoms in this condition may not be solely linked to increased tone or inadequate relaxation. A therapeutic role for NO supplementation has also been suggested in diabetic gastroenteropathy. In murine models of diabetes, NANC relaxation and NOS activity are decreased in gastric muscle preparations.36 Interestingly, insulin administration restores NOS expression and improves gastric emptying, while the phosphodiesterase inhibitor sildenafil corrects gastric motor abnormalities in these models, the latter perhaps by decreasing pyloric spasm.36 In diabetic human studies, nitrates improve postprandial gastric accommodation although postprandial symptoms are not improved.37 Hypertrophic pyloric stenosis is also associated with a decrease in nNOS expression within the hypertrophied circular smooth muscle sphincter, while NOS protein remains in the adjacent longitudinal muscle layer, suggesting a specific defect in the sphincter. The neuronal defect in circular smooth muscle may not be limited to nNOS, but may also involve widespread disruption of neuronal elements38 and interstitial cells of Cajal (ICC) networks.39 As a corollary, nNOS⫺/⫺ and PKG⫺/⫺ mice develop marked gastric dilation and a thickened pylorus at a young age,40,41 although these animals do not entirely recapitulate the phenotype of infantile hypertrophic pyloric stenosis. These studies argue for the importance of the NO– cGMP pathway in pyloric sphincter relaxation and suggest that NO donors and phosphodiesterase inhibitors may be an attractive area for future studies for a variety of gastroparetic syndromes in humans. Intestinal Motility and Motor Disorders Studies from animal models have shown a role for NO as an inhibitor of small bowel motility. In humans, NOS blockade results in an increase in fasting motor

NITRIC OXIDE AND GI SYSTEM

905

activity.42 As a corollary, postoperative ileus may be a result of increased intestinal NO production because urinary nitrate levels are raised postoperatively,43 perhaps in response to surgical manipulation of the intestine. Indeed, iNOS⫺/⫺ mice exhibit less phagocyte infiltration than controls, and are protected from the reduction in contractility that occurs in response to surgical manipulation.44 Furthermore, NOS inhibitors, when used in combination with adrenergic agents, normalize intestinal transit time in animal models of the postoperative gut.45 However, the link between NO generation and postoperative ileus in humans is not as well established at this juncture.43 Deficient NO generation has also been linked to other small and large intestinal transit disorders including diabetic enteropathy and slow-transit constipation. For example, loss of NOS expression has been described in a patient with long-standing diabetes, together with loss of immunoreactivity for VIP and a decrease in the volume of ICC.46 Based on these and other studies, the role of NO– cGMP supplementation in the treatment of diabetic GI symptoms is currently under investigation. Although the role of NO in colonic motility in humans is not well studied, some studies show reduced NOS expression in slow-transit constipation. However, the selective contribution of NO deficiency to the pathogenic basis of these disorders remains unestablished as other cellular defects are present as well.47– 49 Immunohistochemical12 and electrophysiologic50 evidence, together with the use of topical NO donors show a clear role for NO in relaxation of the anal sphincter. Topical application of glyceryl trinitrate produces a decrement in the resting pressure of the anal sphincter.51 Topical NO donors are also effective in treating chronic anal fissure because they provide relief from fissurerelated pain and promote healing of the fissure itself.52,53 This effect may come through 2 potential mechanisms, decreased anal sphincter tone and increased regional blood flow through vasodilation. Based on these studies, NO donors may also be useful in the treatment of anismus, which is characterized by elevated sphincter tone and disordered defecation.

Vascular Function in the Gastroesophageal System Biology of eNOS eNOS is the key NOS isoform responsible for NO-regulated vasodilation in the GI system. Thus it follows that in the GI system, eNOS expression is most prominent in endothelial cells lining vascular channels,

906

SHAH ET AL.

throughout the gut, liver, and pancreas, including arterial, venous, and microcirculatory vessels.54 eNOS may also be expressed in gut smooth muscle cells and ICC.55,56 eNOS-derived NO is produced under basal conditions and in response to a variety of stimuli. For example, a number of hormonal, paracrine, and mechanical factors stimulate increases in eNOS-derived NO production from existing eNOS protein, such as shear stress, insulin, bradykinin, and others.57 Integral to this process is the calcium-dependent activation of calmodulin which, in conjunction with additional molecular steps including phosphorylation, regulatory protein interactions, and aminoterminal fatty acid modifications, act in concert to finely tune NO production.58 Furthermore, although “constitutive,” a number of transcriptional and posttranscriptional mechanisms are also important to the sum generation of NO via eNOS.58 eNOS regulates a number of cellular and physiologic functions within the GI and hepatic systems including vasodilation, inhibition of leukocyte, platelet and mast cell adhesion, and protection of mucosal barrier function. Based on these physiologic functions, regional changes in eNOS-derived NO generation may contribute to the pathogenesis of a number of digestive diseases including portal hypertension, ischemia-reperfusion, and gastric mucosal injury, as described below. Gastrointestinal Blood Flow Regulation and Portal Hypertension NO plays a key role in vascular perfusion and regulation by promoting vasodilation via signaling to smooth muscle cell cGMP and PKG.6 The majority of blood flow to the GI system enters from the mesenteric vasculature, and flow regulation at the level of the mesenteric arterioles is a key determinant of total and regional intestinal blood flow.59,60 Additionally, much of the mesenteric inflow is directed to the GI mucosa to facilitate absorption and secretion. The role of NO in these processes is evidenced by the demonstration that inhibitors of NOS decrease splanchnic blood flow by inhibiting NOS at the mesenteric arteriolar level.60 In addition to increasing splanchnic inflow, eNOSderived NO generated from liver endothelial cells increases flow within the sinusoidal channels of the liver itself.57,61,62 This concept is supported by studies showing the expression and regulation of eNOS within hepatic vascular channels57,63 increase in hepatic pressure in response to NOS inhibitors61 and inhibitory influence of NOS isoform overexpression on hepatic vasoconstrictive responses.64,65 NO has also been implicated prominently in hepatic microcirculatory and splanchnic vascular dysfunction,

GASTROENTEROLOGY Vol. 126, No. 3

Figure 2. The role of NO and NOS isoforms in pathogenesis and complications of portal hypertension. (Left) The vascular contributions to the pathogenesis of portal hypertension include an increase in intrahepatic resistance and an increase in splanchnic inflow. While the former is mediated through a deficiency in eNOS-derived NO generation in the liver, the latter is mediated by an increase in NO generation in the systemic circulation, particularly at the mesenteric arteriole. Most studies implicate eNOS as the relevant NOS isoform in this process, although iNOS has been implicated as well. (Right) Complications of portal hypertension in which NO has been implicated include hepatopulmonary syndrome, hepatic encephalopathy, hepatorenal syndrome, and cirrhotic cardiomyopathy. Each of these complications has been associated with excess NO generation. However, the relevant isoform responsible for excess NO generation in these complications varies, and the therapeutic benefit of NOS inhibition is not yet established.

particularly portal hypertension (Figure 2).62,63,66,67 Although mechanical factors contribute to much of the increased resistance within the liver in portal hypertension, there is clearly a vasculogenic component to the development and perpetuation of this syndrome as well. The vascular component of portal hypertension includes an increase in splanchnic blood flow, as well as an increase in intrahepatic vascular resistance.68 Dysregulation of the NO system appears to play a key role in both these processes. For example, increased NO generation mediates the increased splanchnic blood flow and vasodilation most likely through increased activation of the eNOS isoform,69 –72 although both iNOS and nNOS isoforms have been implicated in this process as well.73,74 The mechanism of activation of eNOS in this process seems to involve both mechanical and humoral factors that signal NOS activation by pathways involving increased intracellular calcium, AKT, molecular chaperones, and others.71,75–77 However, eNOS and NO may not be indispensable in this process as some, though not all, studies in eNOS⫺/⫺ mice have shown that the hyperdynamic circulation and portal hypertension develop even in the absence of eNOS.78,79 In contradistinction to the activation of eNOS in the mesenteric endothelial cells, eNOS function is impaired in the hepatic endothelial cells, thereby contributing to increased intrahepatic resistance and portal hypertension (Figure 2).63,67,80 Additionally, contractile cells in the liver demonstrate impaired ability to dilate in response to NO.81 The signaling events and primary stimuli that

March 2004

cause impaired NOS activation in the hepatic sinusoids may involve the inhibitory NOS protein, caveolin,67,80 and/or impaired AKT activation of eNOS.82 Therapeutic approaches aimed at manipulating the NO pathway in portal hypertension and its components have been an area of investigation in both experimental systems and in humans and have met with varying success. Inhibition of NOS signaling pathways in the mesenteric vasculature has been attempted, with the goal of attenuating the hyperdynamic circulatory state. However, initial studies aimed at systemic NOS inhibition to limit portal hypertension and its other complications in humans have not been promising,83 except perhaps in the hepatopulmonary syndrome, in which case an inhibitor of guanylate cyclase, methylene blue, has been shown to reduce the excess pulmonary vasodilation that characterizes this condition.84 Attempts to pharmacologically augment the NOS system in liver to correct hepatic vasoconstriction have been largely impaired by the inability to selectively deliver NO donor agents to the desired site of action.85 However, some potential directions for therapy of portal hypertension and its complications through potentiation of NOS function, both pharmacologically or molecularly, have been investigated in experimental models or in preliminary form in humans.64,65,82,86 – 88

Mesenteric Ischemia–Reperfusion Injury NO inhibits the adhesion and activation of specific hematopoietic cells, thereby acting as a mitigating force against vascular stasis and ensuing ischemic tissue injury. Cells most prominently involved in this process and inhibited by NO include leukocytes and platelets. In the case of leukocyte function, NO inhibits leukocyte adhesion to the vascular endothelium, in part through inhibiting the ability of leukocytes to roll along the vascular channels, a prerequisite step for adhesion.89 The inhibitory influence of NO on leukocyte– endothelium interactions appears to be mediated through adhesion molecule interactions, particularly involving P-selectin and beta integrins (CD11/CD18).90 Additionally, studies using pharmacologic inhibitors implicate signaling pathways involving phospholipase A2, PAF, and leukotriene B4 in this process.91 The inhibitory actions of NO on platelet activation, adhesion, and aggregation are also well established.92 NO-mediated inhibition of platelet aggregation is mediated in large part by cGMP-PI3 kinase-dependent mechanisms, although other pathways independent of cGMP and involving peroxy-

NITRIC OXIDE AND GI SYSTEM

907

nitrite-dependent protein nitration have also been proposed.93,94 The inhibitory influence of NO on leukocyte and platelet function has significant import on mesenteric ischemia–reperfusion injury (IRI) because platelet aggregation and interactions of leukocytes with the vascular endothelium contribute to this syndrome.95,96 Experimental studies show that NO production is reduced in mesenteric IRI, inhibition of NOS function mimics the effects of IRI on the gut, and NO donor compounds protect against IRI, perhaps in part by inhibiting the upregulation of P-selectin.95 Owing to the beneficial effects of NO supplementation on experimental IRI, this condition is another potential target of NO donor compounds. Mucosal Permeability and Injury Although the influence of NO on mucosal permeability and protection of barrier function is controversial, experiments using pharmacologic inhibitors of NOS indicate that inhibition of basal NO production enhances epithelial mucosal permeability, thus implicating constitutive NO production in the protection of GI mucosal barrier function.97 The mechanism of this NO protective effect may be through enhanced mucosal blood flow and a stabilizing influence on mast cell function, although direct effects on epithelial cell function may also be important.97,98 However, excess NO production associated with inflammatory states is also characterized by increased epithelial permeability and a loss of mucosal barrier function. Thus, the level of NO generation, relevant isoform-generating NO, redox status of the epithelial cells, and other specifics relating to the cellular milieu may determine the sum effect of NO on mucosal permeability and protection. The potentially protective influence of NO on mucosal barrier function has been extrapolated to a possible beneficial role of NO donor agents in specific disease processes and toxicities associated with impaired mucosal barrier function, such as ulcer disease, nonsteroidal antiinflammatory drug (NSAID) toxicity, and alcohol. For example, clinical studies have shown that coadministration of NO donor agents with NSAIDs may protect from NSAID-induced ulcers, and combination agents in which the NSAID or aspirin is linked to a NO-releasing moiety may result in less mucosal injury, as compared with the use of traditional cyclooxygenase (COX) inhibitor agents, without impairing their beneficial effects, and may even enhance mucosal tissue repair.99 –101 Thus, a compelling target of NO donor compounds relates to protection of gastric mucosal integrity.

908

SHAH ET AL.

Inflammation and Carcinogenesis Biology of iNOS iNOS is the NOS isoform most broadly implicated in the processes of inflammation and carcinogenesis in the GI system. iNOS expression can be detected within almost any GI cell type, given the appropriate stimulatory milieu, though most notably in inflammatory cells. Some reports also suggest constitutive expression in the epithelia of the normal human colon102 and myenteric neurons.55 iNOS is regulated predominantly at the transcriptional level through mechanisms dependent on NF-␬B.103 However, recent studies are highlighting the importance of additional transcription factors and response elements in the regulation of iNOS gene transcription as well.104 The enzymatic activity of iNOS is not altered by increased calcium transients, owing to its high affinity for calmodulin, which facilitates binding even in the absence of stimulus dependent increases in intracellular calcium. However, putative posttranslational mechanisms of iNOS regulation have also been described.105 iNOS-derived NO generation occurs at orders of magnitude greater than that of other isoforms. However, it has been postulated that other reactive oxygen species may be contemporaneously generated by iNOS, which may contribute to differential molecular targets and cellular influences of iNOS-mediated NO generation. In the GI system, iNOS-derived NO generation has been importantly implicated in epithelial cell injury/apoptosis, host immune defense, and perpetuation of inflammatory responses. The specific functional roles of iNOS-mediated NO on these processes is described later. Colitis and Colon Cancer Although the role of NO in Crohn’s disease is unclear, a role for NO has been proposed in the pathogenesis of ulcerative colitis.106 In human ulcerative colitis, mucosal iNOS expression and activity, serum nitrate levels, and luminal NO generation are increased.106 –109 Levels of nitrotyrosine (a marker of nitrosative stress) are also increased, supporting a pathogenic effect of NO in this condition.107 In animal models of colitis, colonic NO generation is also increased in association with development of colitis, although blockade of NO production in these models has not consistently ameliorated measures of colitis.110,111 Furthermore, induction of colitis in the iNOS⫺/⫺ suggests that iNOS may actually be important in recovery from experimental colitis.112 Other colitides such as collagenous colitis are also associated with increased NO generation,107 although in collagenous colitis, nitrotyrosine staining is not elevated,

GASTROENTEROLOGY Vol. 126, No. 3

perhaps corresponding to the generally lower degree of inflammation seen in this condition. In addition to its potential role in the pathogenesis of colitides, increased NO generation may also contribute to the reduced colonic motility associated with intestinal inflammation, such as toxic megacolon,113,114 as well as the secretory diarrhea associated with these colitides.107 Elevated iNOS activity has been shown in colon adenomas, colon cancer, and metastases,115 and it is thought that NO may contribute to the progression of adenoma to carcinoma by damaging DNA, increasing gene expression of COX-2, or generating posttranslation modifications via nitrosylation of proteins. Indeed, it has been suggested that the increased cancer incidence in chronic ulcerative colitis may be related to excess NO generation.116 An area of mounting experimental interest is the putative role of NOS inhibitors in colon cancer chemoprotection, paralleling the role that COX-2 inhibitors appear to play in this process. Using an azoxymethaneinduced colon cancer model in rats, Rao et al.117 showed a reduction in the development of aberrant crypts, a precursor to colon cancer in animals treated with the preferential iNOS inhibitors, SC-51 and aminoguanidine, similar to that observed with the COX inhibitor, sulindac, and the specific COX-2 inhibitor, celecoxib. Further studies in this area appear warranted. Hepatotoxicity NO has been implicated in the processes of apoptosis and hepatotoxicity as well as hepatoprotection, depending on the specific experimental conditions and nature of the models.66,118 The models reflect a diverse array of diseases, including IRI, toxin-mediated injury, and others.66,119 –121 For example, the influence of pharmacologic supplementation of NO in these models is often difficult to interpret because models that use NO donors in isolated cell systems likely do not specifically duplicate either eNOS or iNOS function. Likewise, in vivo studies using broad NOS inhibitors that block both iNOS and eNOS function lead to complex effects, making interpretation of isoform selectivity difficult. For example, the effects of inhibiting eNOS and altering blood flow may be deleterious, whereas the effects of iNOS suppression may be beneficial. In addition, the redox context of the injurious model may also influence results. Models with superoxide generation may favor peroxynitrite formation, whereas disease states with little oxidative stress may allow NO generation without peroxynitrite generation. Peroxynitrite promotes apoptosis in some experimental conditions via inducing mitochondrial dysfunction with cytochrome c release, which in

March 2004

turn stimulates caspase activation and apoptosis.66 In contrast, NO can nitrosylate multiple caspases inhibiting their function and blocking apoptosis.122 In addition, NO may also inhibit apoptosis by activating PKG, though the mechanism by which this kinase blocks apoptosis remains obscure. Owing to the limitations in interpreting NOS isoform specificity through pharmacologic studies, in vivo models using isoform-specific NOS gene deletion are of particular utility. In this regard, iNOS has been implicated as a cause of immune-mediated liver injury in the murine concanavalin A model of T-cell–associated liver injury and also in acetaminophen hepatoxicity.123–126 However, iNOS has also been shown to be important in liver regeneration and recovery from liver injury.127 Thus, the role of NO in liver injury will also depend on the type of injury occurring and the importance of liver regeneration in recovering from the injury. These issues, and others, may account for the apparent disparities in the literature relating to the influence of iNOS-derived NO on cell injury. Hepatic Carcinogenesis Chronic injury and inflammation related to NO may also predispose to the promotion and development of neoplasia. Indeed, there is experimental evidence implicating NO in the development of cholangiocarcinoma and hepatocellular carcinoma. The procarcinogenic influence of NO may be related in part to the aforementioned influence of NO toward inhibiting apoptosis and thereby promoting dysregulated cell proliferation. However, other mechanisms may be relevant as well, including DNA and protein damage. For example, NO and downstream reactive nitrogen species can directly damage DNA by promoting base pair mutation, strand break, and cross-linking.128 Indeed, this event has been observed in the DNA of the key proliferative checkpoint protein, p53.128 NO may also regulate key proteins involved in carcinogenesis through posttranslational modifications including nitration and nitrosylation. For example, cysteine thiol S-nitrosylation and subsequent inhibition of the DNA repair enzyme hOgg1, has been shown in cholangiocarcinoma cell lines and implicated in the transformation process.129 Nitrosylation of caspases can also occur resulting in apoptosis, thereby contributing to the carcinogenic process.130 Lastly, NO may promote tumor growth through the promotion of tumor angiogenesis. Indeed, multiple steps required for tumor angiogenesis are promoted by NO, either downstream of growth factors or directly, most notably endothelial cell proliferation and vascular permeability.131 Importantly, these mechanisms of carcinogenesis may be relevant to

NITRIC OXIDE AND GI SYSTEM

909

the development of other GI epithelial malignancies as well.

Summary NO is implicated in multiple important processes in gastrohepatic biology and diseases, and the pathogenic role of NO in these processes remains an area of active investigation. Interestingly, some disease conditions are associated with excess NO generation; other disease conditions, such as achalasia, are associated with deficient NO generation, while in some syndromes, such as portal hypertension and liver injury, both deficiencies and excesses of NO generation appear to contribute, with the sum influence of NO being based on spatial factors and temporal kinetics. These paradoxical observations highlight the complexities of NO-based signaling in GI health and disease processes. Significant interest has now been focused toward modulating the NO system as a therapeutic option in several arenas, including motility disorders of the GI tract, GI cancer, vascular disorders, and modulation of inflammation and injury. Thus, with advances in NO targeting, delivery, and isoform selectivity, it is likely we will see more human trials using organ-selective NO supplementation and isoform-specific NOS inhibitors targeting a wide variety of diseases in the GI system.

References 1. Stark ME, Szurszewski JH. Role of nitric oxide in gastrointestinal and hepatic function and disease. Gastroenterology 1992;103: 1928 –1949. 2. 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 –718. 3. Sessa WC, Harrison JK, Barber CM, Zeng D, Durieux ME, D’Angelo DD, Lynch KR, Peach MJ. Molecular cloning and expression of a cDNA encoding endothelial cell nitric oxide synthase. J Biol Chem 1992;267:15274 –15276. 4. Xie QW, Cho HJ, Calaycay J, Mumford RA, Swiderek KM, Lee TD, Ding A, Troso T, Nathan C. Cloning and characterization of inducible nitric oxide synthase from mouse macrophages. Science 1992;256:225–228. 5. Palmer RM, Ferrige AG, Moncada S. Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nature 1987;327:524 –526. 6. Denninger J, Marletta M. Guanylate cyclase and the NO/cGMP signaling pathway. Biochim Biophys Acta 1999;1411:334 –350. 7. Stamler J, Lamas S, Fang F. Nitrosylation: the prototypic redoxbased signaling mechanism. Cell 2001;106:675– 683. 8. Jaffrey S, Erdjument-Bromage H, Ferris C, Tempst P, Snyder S. Protein S-nitrosylation: a physiological signal for neuronal nitric oxide. Nat Cell Biol 2001;3:193–197. 9. Beckman J, Koppenol W. Nitric oxide, superoxide, and peroxynitrite: the good, the bad, and the ugly. Am J Physiol 1996;271: C1424 –C1437. 10. Boeckxstaens GE, Pelckmans PA, Jordaens FH, Van Maercke

910

11.

12.

13.

14.

15.

16.

17.

18.

19.

20.

21.

22.

23.

24.

25.

26.

27.

28.

29.

SHAH ET AL.

YM, Herman AG. Nonadrenergic noncholinergic mechanisms in the ileocolonic junction. Nature 1990;345:346 –347. Stark ME, Bauer AJ, Sarr MG, Szurszewski JH. Nitric oxide mediates inhibitory nerve input in human and canine jejunum. Gastroenterology 1993;104:398 – 409. Chakder S, Bandyopadhyay A, Rattan S. Neuronal NOS gene expression in gastrointestinal myenteric neurons and smooth muscle cells. Am J Physiol 1997;273:C1868 –C1875. Bredt DS, Hwang PM, Snyder SH. Localization of nitric oxide synthase indicating a neural role for nitric oxide. Nature 1990; 347:768 –770. Saur D, Paehge H, Schusdziarra V, Allescher HD. Distinct expression of splice variants of neuronal nitric oxide synthase in the human gastrointestinal tract. Gastroenterology 2000;118: 849 – 858. Grider JR, Cable MB, Said SI, Makhlouf GM. Vasoactive intestinal peptide as a neural mediator of gastric relaxation. Am J Physiol 1985;248:G73–G78. Xue L, Farrugia G, Szurszewski JH. Effect of exogenous ATP on canine jejunal smooth muscle. Am J Physiol 2000;278:G725– G733. Xue L, Farrugia G, Miller SM, Ferris CD, Snyder SH, Szurszewski JH. Carbon monoxide and nitric oxide as coneurotransmitters in the enteric nervous system: evidence from genomic deletion of biosynthetic enzymes. Proc Natl Acad Sci U S A 2000;97:1851– 1855. Dick JM, Van Molle W, Brouckaert P, Lefebvre RA. Relaxation by vasoactive intestinal polypeptide in the gastric fundus of nitric oxide synthase-deficient mice. J Physiol 2002;538:133–143. Pfeifer A, Klatt P, Massberg S, Ny L, Sausbier M, Hirneiss C, Wang GX, Korth M, Aszodi A, Andersson KE, Krombach F, Mayerhofer A, Ruth P, Fassler R, Hofmann F. Defective smooth muscle regulation in cGMP kinase I-deficient mice. EMBO J 1998;17:3045–3051. Allescher HD, Kurjak M, Huber A, Trudrung P, Schusdziarra V. Regulation of VIP release from rat enteric nerve terminals: evidence for a stimulatory effect of NO. Am J Physiol 1996;271: G568 –G574. Mashimo H, He XD, Huang PL, Fishman MC, Goyal RK. Neuronal constitutive nitric oxide synthase is involved in murine enteric inhibitory neurotransmission. J Clin Invest 1996;98:8 –13. Thorup C, Jones CL, Gross SS, Moore LC, Goligorsky MS. Carbon monoxide induces vasodilation and nitric oxide release but suppresses endothelial NOS. Am J Physiol 1999;277:F882– 889. Bolotina VM, Najibi S, Palacino JJ, Pagano PJ, Cohen RA. Nitric oxide directly activates calcium-dependent potassium channels in vascular smooth muscle. Nature 1994;368:850 – 853. Murray JA, Ledlow A, Launspach J, Evans D, Loveday M, Conklin JL. The effects of recombinant human hemoglobin on esophageal motor functions in humans. Gastroenterology 1995;109: 1241–1248. Konturek JW, Thor P, Lukaszyk A, Gabryelewicz A, Konturek SJ, Domschke W. Endogenous nitric oxide in the control of esophageal motility in humans. J Physiol Pharmacol 1997;48:201– 209. Mearin F, Mourelle M, Guarner F, Salas A, Riveros-Moreno V, Moncada S, Malagelada JR. Patients with achalasia lack nitric oxide synthase in the gastro-oesophageal junction. Eur J Clin Invest 1993;23:724 –728. Sivarao DV, Mashimo HL, Thatte HS, Goyal RK. Lower esophageal sphincter is achalasic in nNOS(⫺/⫺) and hypotensive in W/W(v) mutant mice. Gastroenterology 2001;121:34 – 42. Kim CD, Goyal R, Mashimo H. Neuronal NOS provides nitrergic inhibitory neurotransmitter in mouse lower esophageal sphincter. Am J Physiol 1999;277:G280 –G284. Gelfond M, Rozen P, Gilat T. Isosorbide dinitrate and nifedipine

GASTROENTEROLOGY Vol. 126, No. 3

30.

31.

32.

33.

34.

35.

36.

37.

38.

39.

40.

41.

42.

43.

44.

45.

46.

47.

48.

treatment of achalasia: a clinical, manometric and radionuclide evaluation. Gastroenterology 1982;83:963–969. Eherer A, Schwetz I, Hammer H, Petnehazy T, Scheidl S, Weber K, Krejs G. Effect of sildenafil on oesophageal motor function in healthy subjects and patients with oesophageal motor disorders. Gut 2002;50:758 –764. Tack J, Demedts I, Meulemans A, Schuurkes J, Janssens J. Role of nitric oxide in the gastric accommodation reflex and in meal induced satiety in humans. Gut 2002;51:219 –224. Konturek JW, Thor P, Domschke W. Effects of nitric oxide on antral motility and gastric emptying in humans. Eur J Gastroenterol Hepatol 1995;7:97–102. Coulie B, Tack J, Sifrim D, Andrioli A, Janssens J. Role of nitric oxide in fasting gastric fundus tone and in 5-HT1 receptormediated relaxation of gastric fundus. Am J Physiol 1999;276: G373–G377. Kuiken SD, Vergeer M, Heisterkamp SH, Tytgat GN, Boeckxstaens GE. Role of nitric oxide in gastric motor and sensory functions in healthy subjects. Gut 2002;51:212–218. Gilja OH, Hausken T, Bang CJ, Berstad A. Effect of glyceryl trinitrate on gastric accommodation and symptoms in functional dyspepsia. Dig Dis Sci 1997;42:2124 –2131. Watkins CC, Sawa A, Jaffery S, Blackshaw S, Barrow RK, Snyder SH, Ferris CD. Insulin restores neuronal nitric oxide synthase expression and function that is lost in diabetic gastropathy. J Clin Invest 2000;106:373–384. Undeland KA, Hausken T, Gilja OH, Aanderud S, Berstad A. Gastric meal accommodation and symptoms in diabetes. A placebo-controlled study of glyceryl trinitrate. Eur J Gastroenterol Hepatol 1998;10:677– 681. Langer JC, Berezin I, Daniel EE. Hypertrophic pyloric stenosis: ultrastructural abnormalities of enteric nerves and the interstitial cells of Cajal. J Pediatr Surg 1995;30:1535–1543. Vanderwinden JM, Liu H, De Laet MH, Vanderhaeghen JJ. Study of the interstitial cells of Cajal in infantile hypertrophic pyloric stenosis [erratum appears in Gastroenterology 1996;111(5): 1403]. Gastroenterology 1996;111:279 –288. Huang PL, Dawson TM, Bredt DS, Snyder SH, Fishman MC. Targeted disruption of the neuronal nitric oxide synthase gene. Cell 1993;75:1273–1286. Mashimo H, Kjellin A, Goyal RK. Gastric stasis in neuronal nitric oxide synthase-deficient knockout mice. Gastroenterology 2000;119:766 –773. Russo A, Fraser R, Adachi K, Horowitz M, Boeckxstaens G. Evidence that nitric oxide mechanisms regulate small intestinal motility in humans. Gut 1999;44:72–76. Moojen TM, Van Gulik TM, Hoek FJ, Gouma DJ, Tytgat GN, Boeckxstaens GE. Possible role of nitric oxide in postoperative ileus: a comparative study. Neurogastroenterol Motil 1999;11: 403– 408. Kalff JC, Schraut WH, Billiar TR, Simmons RL, Bauer AJ. Role of inducible nitric oxide synthase in postoperative intestinal smooth muscle dysfunction in rodents. Gastroenterology 2000; 118:316 –327. De Winter BY, Boeckxstaens GE, De Man JG, Moreels TG, Herman AG, Pelckmans PA. Effect of adrenergic and nitrergic blockade on experimental ileus in rats. Br J Pharmacol 1997; 120:464 – 468. He CL, Soffer EE, Ferris CD, Walsh RM, Szurszewski JH, Farrugia G. Loss of interstitial cells of cajal and inhibitory innervation in insulin-dependent diabetes. Gastroenterology 2001;121:427– 434. He CL, Burgart L, Wang L, Pemberton J, Young-Fadok T, Szurszewski J, Farrugia G. Decreased interstitial cell of cajal volume in patients with slow-transit constipation. Gastroenterology 2000;118:14 –21. Lyford G, He CL, Soffer EE, Hull TL, Strong SA, Senagore AJ,

March 2004

49.

50.

51.

52.

53.

54.

55.

56.

57.

58.

59.

60.

61.

62.

63.

64.

65.

66. 67.

Burgart L, Young-Fadok T, Szurszewski J, Farrugia G. Pan-colonic decrease in interstitial cells of Cajal in patients with slow transit constipation. Gut 2002;51:496 –501. Tomita R, Fujisaki S, Ikeda T, Fukuzawa M. Role of nitric oxide in the colon of patients with slow-transit constipation. Dis Colon Rectum 2002;45:593– 600. O’Kelly T, Brading A, Mortensen N. Nerve mediated relaxation of the human internal anal sphincter: the role of nitric oxide. Gut 1993;34:689 – 693. Loder PB, Kamm MA, Nicholls RJ, Phillips RK. “Reversible chemical sphincterotomy” by local application of glyceryl trinitrate. Br J Surg 1994;81:1386 –1389. Lund J, Scholefield J. A randomised, prospective, double-blind, placebo-controlled trial of glyceryl trinitrate ointment in treatment of anal fissure. Lancet 1997;349:11–14. Carapeti EA, Kamm MA, McDonald PJ, Chadwick SJ, Melville D, Phillips RK. Randomised controlled trial shows that glyceryl trinitrate heals anal fissures, higher doses are not more effective, and there is a high recurrence rate. Gut 1999;44:727– 730. Fischer H, Becker J, Boknik P, Huber V, Luss H, Neumann J, Schmitz W, Domschke W, Stachura J, Konturek J. Expression of constitutive nitric oxide synthase in rat and human gastrointestinal tract. Biochim Biophys Acta 1999;1450:414 – 422. Vannucchi M-G, Corsani L, Bani D, Faussone-Pellegrini M-S. Myenteric neurons and interstitial cells of Cajal of mouse colon express several nitric oxide synthase isoforms. Neurosci Lett 2002;326:191–195. Teng B, Murthy KS, Kuemmerle JF, Grider JR, Sase K, Michel T, Makhlouf GM. Expression of endothelial nitric oxide synthase in human and rabbit gastrointestinal smooth muscle cells. Am J Physiol 1998;275:G342–G351. Shah V, Haddad F, Garcia-Cardena G, Frangos J, Mennone A, Groszmann R, Sessa W. Liver sinusoidal endothelial cells are responsible for nitric oxide modulation of hepatic resistance. J Clin Invest 1997;100:2923–2930. Papapetropoulos A, Rudic R, Sessa W. Molecular control of nitric oxide synthases in the cardiovascular system. Cardiovasc Res 1999;43:509 –520. Shah V, Kamath P, de Groen P. Physiology of the splanchnic circulation. In: Topol E, Lanzer F, eds. Theory and practice of vascular diseases. Germany: Springer Verlag, 2002:1688 – 1694. Kusayama T, Yamazaki J, Nagao T. Flow dependence of nitric oxide-mediated pressure change in rat mesenteric beds with different tonus. Eur J Pharmacol 1996;312:301–307. Mittal MK, Gupta TK, Lee F-Y, Sieber CC, Groszmann RJ. Nitric oxide modulates hepatic vascular tone in normal rat liver. Am J Physiol 1994;267:G416 –G422. Pannen B, Bauer M, Noldge-Schomburg G, Zhang J, Robotham J, Clemens M, Geiger K. Regulation of hepatic blood flow during resuscitation from hemorrhagic shock: role of NO and endothelins. Am J Physiol 1997;272:H2736 –H2745. Rockey DC, Chung JJ. Reduced nitric oxide production by endothelial cells in cirrhotic rat liver: endothelial dysfunction in portal hypertension. Gastroenterology 1998;114:344 –351. Yu Q, Shao R, Qian H, George S, Rockey D. Gene transfer of nNOS to cirrhotic rat liver ameliorates portal hypertension. J Clin Invest 2000;105:741–748. Shah V, Chen A, Cao S, Hendrickson H, Weiler D, Smith L, Yao J, Katusic Z. Gene transfer of recombinant endothelial nitric oxide synthase to liver in vivo and in vitro. Am J Physiol 2000; 279:G1023–G1030. Clemens M. Nitric oxide in liver injury. Hepatology 1999;30:1–5. Shah V, Toruner M, Haddad F, Cadelina G, Papapetropoulos A, Sessa W, Groszmann R. Impaired endothelial nitric oxide synthase activity associated with enhanced caveolin binding in

NITRIC OXIDE AND GI SYSTEM

68. 69.

70.

71.

72.

73.

74.

75.

76.

77.

78.

79.

80.

81.

82.

83.

84.

911

experimental liver cirrhosis. Gastroenterology 1999;117:1222– 1228. Shah V. Cellular and molecular basis of portal hypertension. Clin Liver Dis 2001;5:629 – 644. Cahill P, Foster C, Redmond E, Gingalewski C, Wu Y, Sitzmann J. Enhanced nitric oxide synthase activity in portal hypertensive rabbits. Hepatology 1995;22:598 – 606. Sieber CC, Groszmann RJ. In vitro hyporeactivity to methoxamine in portal hypertensive rats: reversal by nitric oxide blockade. Am J Physiol 1992;262:G996 –G1001. Shah V, Wiest R, Garcia-Cardena G, Cadelina G, Groszmann R, Sessa W. Hsp90 regulation of endothelial nitric oxide synthase contributes to vascular control in portal hypertension. Am J Physiol 1999;277:G463–G468. Martin P, Xu D, Niederberger M, Weigert A, Tsai P, St. John J, Gines P, Schrier R. Upregulation of endothelial constitutive NOS: a major role in the increased NO production in cirrhotic rats. Am J Physiol 1996;270:F494 –F499. Morales-Ruiz M, Jimenez W, Perez-Sala D, Ros J, Leivas A, Lamas S, Rivera F, Arroyo V. Increased nitric oxide synthase expression in arterial vessels of cirrhotic rats with ascites. Hepatology 1996;24:1481–1486. Xu L, Carter E, Ohara M, Martin P, Rogachev B, Morris K, Cadnapaphornchai M, Knotek M, Schrier R. Neuronal nitric oxide synthase and systemic vasodilation in rats with cirrhosis. Am J Physiol 2000;279:F1110 –F1115. Ros J, Claria J, To-Figueras J, Planaguma A, Cejudo-Martin P, Fernandez-Varo G, Martin-Ruiz R, Arroyo V, Rivera F, Rodes J, Jimenez W. Endogenous cannabinoids: a new system involved in the homeostasis of arterial pressure in experimental cirrhosis in the rat. Gastroenterology 2002;122:85–93. Batkai S, Jarai Z, Wagner J, Goparaju S, Varga K, Liu J, Wang L, Mirshahi F, Khanolkar A, Makriyannis A, Urbaschek R, Garcia N Jr, Sanyal A, Kunos G. Endocannabinoids acting at vascular CB1 receptors mediate the vasodilated state in advanced liver cirrhosis. Nature Med 2001;7:827– 832. Pateron D, Tazi K, Sogni P, Heller J, Chagneau C, Poirel O, Philippe M, Moreau R, Lebrec D. Role of aortic nitric oxide synthase 3 (eNOS) in the systemic vasodilation of portal hypertension. Gastroenterology 2000;119:196 –200. Iwakiri Y, Cadeline G, Sessa W, Groszmann R. Mice with targeted deletion of eNOS develop hyperdynamic circulation associated with portal hypertension. Am J Physiol 2002;283: G1074 –G1081. Theodorakis N, Wang Y, Skill N, Metz M, Cahill P, Redmond E, Sitzmann J. The role of nitric oxide synthase isoforms in extrahepatic portal hypertension: studies in gene knock-out mice. Gastroenterology 2003;124:1500 –1508. Shah V, Hendrickson H, Cao S, Yao J, Katusic Z. Regulation of hepatic endothelial nitric oxide synthase by caveolin and calmodulin after bile duct ligation in rats. Am J Physiol 2001;280: G1209 –G1216. Dudenhoefer A, Loureiro-Silva M, Cadelina G, Gupta T, Groszmann R. Bioactivation of nitroglycerin and vasomotor response to nitric oxide are impaired in cirrhotic rat livers. Hepatology 2002;36:381–385. Morales-Ruiz M, Cejudo-Martin P, Fernandez-Varo G, Tugues S, Ros J, Angeli P, Rivera F, Arroyo V, Rodes J, Sessa W, Jimenez W. Transduction of the liver with activated Akt normalizes portal pressure in cirrhotic rats. Gastroenterology 2003;125:522–531. Forrest E, Jones A, Dillon J, Walker J, Hayes P. The effect of nitric oxide synthase inhibition on portal pressure and azygos blood flow in patients with cirrhosis. J Hepatol 1995;23:254 – 258. Schenk P, Madl C, Rezaie-Majd S, Lehr S, Muller C. Methylene blue improves the hepatopulmonary syndrome. Ann Intern Med 2000;133:701–706.

912

SHAH ET AL.

85. Angelico M, Carli L, Piat C, Gentile S, Capocaccia L. Effects of isosorbide-5-mononitrate compared with propranolol on first bleeding and long-term survival in cirrhosis. Gastroenterology 1997;113:1632–1639. 86. Bosch-Marce M, Morales-Ruiz M, Jimenez W, Bordas N, Sole M, Ros J, Deulofeu R, Arroyo V, Rivera F, Rodes J. Increased renal expression of nitric oxide synthase type III in cirrhotic rats with ascites. Hepatology 1998;27:1191–1199. 87. Failli P, DeFranco RMS, Caligiuri A, Gentilini A, Romanelli RG, Marra F, Batignani G, Guerra CT, Laffi G, Gentilini P, Pinzani M. Nitrovasodilators inhibit platelet-derived growth factor-induced proliferation and migration of activated human hepatic stellate cells. Gastroenterology 2000;119:479 – 492. 88. Fiorucci S, Antonelli E, Morelli O, Mencarelli A, Alessandro C, Mello T, Palazzetti B, Tallet D, del Soldato P, Morelli A. NCX1000, a NO releasing derivative of ursodeoxycholic acid, selectively delivers NO to the liver and protects against development of portal hypertension. Proc Natl Acad Sci U S A 2001;98:8897– 8902. 89. Kubes P, Suzuki M, Granger D. Nitric oxide: an endogenous modulator of leukocyte adhesion. Proc Natl Acad Sci U S A 1991;88:4651– 4655. 90. Kanwar S, Kubes P. Nitric oxide is an antiadhesive molecule for leukocytes. New Horizons 1995;3:93–104. 91. Arndt H, Russell J, Kurose I, Kubes P, Granger D. Mediators of leukocyte adhesion in rat mesenteric venules elicited by inhibition of nitric oxide synthesis. Gastroenterology 1993;105:675– 680. 92. Loscalzo J. Nitric oxide insufficiency, platelet activation and arterial thrombosis. Circ Res 2001;88:756 –762. 93. Low S, Sabetkar M, Bruckdorfer K, Naseem K. The role of protein nitration in the inhibition of platelet activation by peroxynitrite. FEBS Lett 2002;511:59 – 64. 94. Pigazzi A, Heydrick S, Folli F, Benoit S, Michelson A, Loscalzo J. Nitric oxide inhibits thrombin receptor-activating peptide-induced phosphoinositide 3-kinase activity in human platelets. J Biol Chem 1999;274:14368 –14375. 95. Kurose I, Wolf R, Grisham M, Granger D. Modulation of ischemia/reperfusion-induced microvascular dysfunction by nitric oxide. Circ Res 1994;74:376 –382. 96. Panes J, Granger D. Leukocyte-endothelial cell interactions: molecular mechanisms and implications in gastrointestinal disease. Gastroenterology 1998;114:1066 –1090. 97. Kanwar S, Wallace J, Befus D, Kubes P. Nitric oxide synthesis inhibition increases epithelial permeability via mast cells. Am J Physiol 1994;266:G222–G229. 98. Alican I, Kubes P. A critical role for nitric oxide in intestinal barrier function and dysfunction. Am J Physiol 1996;270:G225– G237. 99. Lanas A, Bajador E, Serrano P, Fuentes J, Carreno S, Guardia J, Sanz M, Montoro M, Sainz R. Nitrovasodilators, low-dose aspirin, other nonsteroidal antiinflammatory drugs, and the risk of upper gastrointestinal bleeding. N Engl J Med 2000;343:834 – 839. 100. Fiorucci S, Antonelli E, Santucci L, Morelli O, Miglietti M, Federici B, Mannucci R, del Soldato P, Morelli A. Gastrointestinal safety of nitric oxide-derived aspirin is related to inhibition of ICE-like cysteine proteases in rats. Gastroenterology 1999; 116:1089 –1106. 101. Wallace J, Reuter B, Cicala C, McKnight W, Grisham M, Cirino G. Novel nonsteroidal anti-inflammatory drug derivatives with markedly reduced ulcerogenic properties in the rat. Gastroenterology 1994;107:173–179. 102. Roberts PJ, Riley GP, Morgan K, Miller R, Hunter JO, Middleton SJ. The physiological expression of inducible nitric oxide synthase (iNOS) in the human colon. J Clin Pathol 2001;54:293– 297.

GASTROENTEROLOGY Vol. 126, No. 3

103. Taylor B, de Vera M, Ganster R, Wang Q, Shapiro R, Morris SJ, Billiar T, Geller D. Multiple NF-kappaB enhancer elements regulate cytokine induction of the human inducible nitric oxide synthase gene. J Biol Chem 1998;273:15148 –15156. 104. Feng X, Guo Z, Nourbakhsh M, Hauser H, Ganster R, Shao L, Geller D. Identification of a negative response element in the human inducible nitric-oxide synthase (hiNOS) promoter: the role of NF-kappa B-repressing factor (NRF) in basal repression of the hiNOS gene. Proc Natl Acad Sci U S A 2002;99:14212– 14217. 105. Ratovitski E, Bao C, Quick R, McMillan A, Kozlovsky C, Lowenstein C. An inducible nitric-oxide synthase (NOS)-associated protein inhibits NOS dimerization and activity. J Biol Chem 1999;274:30250 –30257. 106. Boughton-Smith NK, Evans SM, Hawkey CJ, Cole AT, Balsitis M, Whittle BJ, Moncada S. Nitric oxide synthase activity in ulcerative colitis and Crohn’s disease. Lancet 1993;342:338 –340. 107. Perner A, Andresen L, Normark M, Fischer-Hansen B, Sorensen S, Eugen-Olsen J, Rask-Madsen J. Expression of nitric oxide synthases and effects of L-arginine and L-NMMA on nitric oxide production and fluid transport in collagenous colitis. Gut 2001; 49:387–394. 108. Middleton SJ, Shorthouse M, Hunter JO. Increased nitric oxide synthesis in ulcerative colitis. Lancet 1993;341:465– 466. 109. Lundberg J, Hellstrom P, Lundberg J, Alving K. Greatly increased luminal nitric oxide in ulcerative colitis. Lancet 1994;344: 1673–1674. 110. Hogaboam CM, Jacobson K, Collins SM, Blennerhassett MG. The selective beneficial effects of nitric oxide inhibition in experimental colitis. Am J Physiol 1995;268:G673–G684. 111. Armstrong AM, Campbell GR, Gannon C, Kirk SJ, Gardiner KR. Oral administration of inducible nitric oxide synthase inhibitors reduces nitric oxide synthesis but has no effect on the severity of experimental colitis. Scand J Gastroenterol 2000;35:832– 838. 112. McCafferty D, Mudgett J, Swain M, Kubes P. Inducible nitric oxide synthase plays a critical role in resolving intestinal inflammation. Gastroenterology 1997;112:1022–1027. 113. Coulie B, Camilleri M, Bharucha AE, Sandborn WJ, Burton D. Colonic motility in chronic ulcerative proctosigmoiditis and the effects of nicotine on colonic motility in patients and healthy subjects. Aliment Pharmacol Ther 2001;15:653– 663. 114. Mourelle M, Casellas F, Guarner F, Salas A, Riveros-Moreno V, Moncada S, Malagelada JR. Induction of nitric oxide synthase in colonic smooth muscle from patients with toxic megacolon. Gastroenterology 1995;109:1497–1502. 115. Ambs S, Bennett W, Merriam W, Ogunfusika M, Oser S, Harrington A, Shields P, Felley-Bosco E, Hussain S, Harris C. Relationship between p53 mutations and inducible nitric oxide synthase expression in human colorectal cancer. J Natl Cancer Inst 1999;91:86 – 88. 116. Landino LM, Crews BC, Timmons MD, Morrow JD, Marnett LJ. Peroxynitrite, the coupling product of nitric oxide and superoxide, activates prostaglandin biosynthesis. Proc Natl Acad Sci U S A 1996;93:15069 –15074. 117. Rao CV, Indranie C, Simi B, Manning PT, Connor JR, Reddy BS. Chemopreventive properties of a selective inducible nitric oxide synthase inhibitor in colon carcinogenesis, administered alone or in combination with celecoxib, a selective cyclooxygenase-2 inhibitor. Cancer Res 2002;62:165–170. 118. Li J, Billiar T. Nitric oxide. IV. Determinants of nitric oxide protection and toxicity in liver. Am J Physiol 1999;276:G1069 – G1073. 119. Nanji A, Greenberg S, Tahan S, Fogt F, Loscalzo J, Sadrzadeh S, Xie J, Stamler J. Nitric oxide production in experimental alcoholic liver disease in the rat: role in protection from injury. Gastroenterology 1995;109:899 –907.

March 2004

120. Harada H, Pavlick K, Hines I, Lefer D, Hofman J, Bharwani S, Wolfe R, Grisham M. Sexual dimorphism in reduced-size liver ischemia and reperfusion injury in mice: role of endothelial cell nitric oxide synthase. Proc Natl Acad Sci U S A 2003;100:739 – 744. 121. Hatano E, Bennett B, Manning A, Qian T, Lemasters J, Brenner D. NF-kappaB stimulates inducible nitric oxide synthase to protect mouse hepatocytes from TNF-alpha- and Fas-mediated apoptosis. Gastroenterology 2001;120:1251–1262. 122. Kim YM, Talanian RV, Billiar TR. Nitric oxide inhibits apoptosis by preventing increases in caspase-3-like activity via two distinct mechanisms. J Biol Chem 1997;272:31138 –31148. 123. Koerber K, Sass G, Kiemer AK, Vollmar AM, Tiegs G. In vivo regulation of inducible no synthase in immune-mediated liver injury in mice. Hepatology 2002;36:1061–1069. 124. Sass G, Koerber K, Bang R, Guehring H, Tiegs G. Inducible nitric oxide synthase is critical for immune-mediated liver injury in mice. J Clin Invest 2001;107:439 – 447. 125. Jaeschke H, Gores GJ, Cederbaum AI, Hinson JA, Pessayre D, Lemasters JJ. Mechanisms of hepatotoxicity. Toxicol Sci 2002; 65:166 –176. 126. Bourdi M, Masubuchi Y, Reilly TP, Amouzadeh HR, Martin JL, George JW, Shah AG, Pohl LR. Protection against acetamino-

NITRIC OXIDE AND GI SYSTEM

127.

128.

129.

130.

131.

913

phen-induced liver injury and lethality by interleukin 10: role of inducible nitric oxide synthase. Hepatology 2002;35:289 –298. Rai RM, Lee FY, Rosen A, Yang SQ, Lin HZ, Koteish A, Liew FY, Zaragoza C, Lowenstein C, Diehl AM. Impaired liver regeneration in inducible nitric oxide synthase-deficient mice. Proc Natl Acad Sci U S A 1998;95:13829 –13834. Jaiswal M, LaRusso NF, Gores GJ. Nitric oxide in gastrointestinal epithelial cell carcinogenesis: linking inflammation to oncogenesis. Am J Physiol 2001;281:G626 –G634. Jaiswal M, LaRusso NF, Nishioka N, Nakabeppu Y, Gores GJ. Human Ogg1, a protein involved in the repair of 8-oxoguanine, is inhibited by nitric oxide. Cancer Res 2001;61:6388 – 6393. Torok NJ, Higuchi H, Bronk S, Gores GJ. Nitric oxide inhibits apoptosis downstream of cytochrome C release by nitrosylating caspase 9. Cancer Res 2002;62:1648 –1653. Garcia-Cardena G, Folkman J. Is there a role for nitric oxide in tumor angiogenesis? J Natl Cancer Inst 1998;90:560 –561.

Received April 10, 2003. Accepted July 24, 2003. Address requests for reprints to: Vijay Shah, M.D., GI Research Unit, Alfred 2-435, Mayo Clinic Rochester, 200 First Street SW, Rochester, Minnesota 55905. e-mail: [email protected]; fax: (507) 255-6318.