Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Eur J Pharmacol ; 581(3): 315-23, 2008 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-18215658

RESUMEN

The mechanism of action of 5-aminosalicylic acid (5-ASA), the active therapeutic moiety of a number of clinically used anti-colitic agents, is unclear. The present study investigates whether the beneficial effects in vivo could involve induction of the heat shock protein, heme oxygenase-1 (HO-1), known to provide endogenous anti-oxidant and anti-inflammatory moieties which can modulate colonic inflammation. The effects of 5-ASA on the colonic expression and activity of HO-1 along with its effect on the inflammatory damage have been evaluated in the colitis provoked by instillation of trinitrobenzene sulphonic acid (TNBS) over 48 h in the rat. Intracolonic administration of 5-ASA (8, 25 and 75 mg/kg/day) dose-dependently reduced the TNBS-provoked macroscopic colonic inflammatory injury, myeloperoxidase (MPO) activity and TNF-alpha levels, while also dose-dependently increasing colonic heme oxygenase enzyme activity. Colonic HO-1 protein expression, determined by Western blot analysis in this colitis model, was likewise further induced by 5-ASA. Intracolonic administration of 5-ASA alone under unchallenged conditions also induced colonic HO-1 protein expression and stimulated heme oxygenase enzyme activity. Administration of zinc protoporphyrin (50 micromol/kg/day, s.c.), which prevented the increase in colonic heme oxygenase activity, abolished the anti-colitic effect of 5-ASA. These results suggest that 5-ASA may exert its colonic anti-oxidant and anti-inflammatory effects in vivo in part through the up-regulation of HO-1 enzyme expression and activity.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Colitis/tratamiento farmacológico , Hemo-Oxigenasa 1/efectos de los fármacos , Mesalamina/farmacología , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Western Blotting , Modelos Animales de Enfermedad , Hemo-Oxigenasa 1/metabolismo , Masculino , Mesalamina/administración & dosificación , Peroxidasa/efectos de los fármacos , Peroxidasa/metabolismo , Protoporfirinas/farmacología , Ratas , Ratas Wistar , Ácido Trinitrobencenosulfónico , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
2.
Eur J Pharmacol ; 561(1-3): 164-71, 2007 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-17292349

RESUMEN

Reactive oxygen species, suggested to be involved in inflammatory bowel disease, may be modulated by endogenous anti-oxidant products of heme oxygenase-1 (HO-1). In the present work, HO-1 expression in trinitrobenzene sulphonic acid (TNBS)-induced colitis in the rat and the effects of HO-1 modulation, particularly by the HO-1 inducer, heme, were further evaluated. Colitis was induced by intracolonic challenge with TNBS and assessed macroscopically and by myeloperoxidase (MPO) assay. Heme oxygenase activity was determined by measurement of bilirubin formation and HO-1 protein expression was determined by Western blotting. TNBS challenge led to an early and substantial induction of HO-1 protein expression and heme oxygenase activity in the colon that peaked after 48-72 h and declined over 10 days. Heme (30 micromol/kg/day, s.c) increased colonic HO-1 protein expression and enzyme activity and decreased colonic damage and myeloperoxidase activity. Short-term administration of cadmium chloride (2 mg/kg, s.c.), another known HO-1 inducer, also reduced the colonic injury and myeloperoxidase levels. In contrast, the HO-1 inhibitor, zinc protoporphyrin (50 micromol/kg/day, s.c) significantly increased the colonic damage and myeloperoxidase activity over 10 days, as did tin protoporphyrin (30 micromol/kg/day, s.c). These results support the proposal that induction of HO-1 provides a protective mechanism in this model under both acute and more-chronic conditions, and that its selective up-regulation could thus be of therapeutic potential in colitis.


Asunto(s)
Colitis Ulcerosa/enzimología , Inhibidores Enzimáticos/farmacología , Hemo-Oxigenasa 1/efectos de los fármacos , Hemo/farmacología , Protoporfirinas/farmacología , Animales , Bilirrubina/biosíntesis , Western Blotting , Cloruro de Cadmio/farmacología , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Modelos Animales de Enfermedad , Hemo-Oxigenasa 1/metabolismo , Masculino , Metaloporfirinas/farmacología , Peroxidasa/metabolismo , Ratas , Ratas Wistar , Regulación hacia Arriba/efectos de los fármacos
3.
Br J Pharmacol ; 147(5): 575-82, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16314851

RESUMEN

The effects of the inhibitors of glycogen synthase kinase-3beta (GSK-3beta), TDZD-8 and SB 415286, which can substantially reduce the systemic inflammation associated with endotoxic shock in vivo, have now been investigated on the acute colitis provoked by trinitrobenzene sulphonic acid (TNBS) in the rat. Administration of the GSK-3beta inhibitor TDZD-8 (0.1, 0.33 or 1.0 mg kg-1, s.c., b.i.d., for 3 days) caused a dose-dependent reduction in the colonic inflammation induced by intracolonic TNBS assessed after 3 days, both as the area of macroscopic involvement and as a score using 0-10 scale. Likewise, following administration of the GSK-3beta inhibitor SB 415286 (0.1, 0.33 or 1.0 mg kg-1, s.c., b.i.d., for 3 days), the extent and degree of the TNBS-provoked colonic inflammation was reduced. Administration of either TDZD-8 or SB 415286 reduced the fall in body weight following challenge with TNBS at each dose level studied. The increase in myeloperoxidase activity, an index of neutrophil infiltration into the TNBS-induced inflamed colon, was significantly inhibited by both TDZD-8 and SB 415286 at each dose level. The increase in the levels of the proinflammatory cytokine, TNF-alpha, in the inflamed colon was also significantly inhibited by either compound at the highest doses evaluated. The elevated levels of the transcription factor NF-kappaB subunit p65, as determined by Western blot in the nuclear extracts from the TNBS-provoked inflamed colonic tissue, were dose-dependently reduced by TDZD-8 or SB 415286 treatment. These findings demonstrate that two chemically distinct selective inhibitors of the activity of GSK-3beta reduce the inflammation and tissue injury in a rat model of acute colitis. The mechanisms underlying this anti-inflammatory action may be related to downregulation of NF-kappaB activity, involved in the generation of proinflammatory mediators.


Asunto(s)
Aminofenoles/uso terapéutico , Colitis/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Maleimidas/uso terapéutico , Tiadiazoles/uso terapéutico , Animales , Peso Corporal/efectos de los fármacos , Colon/química , Colon/efectos de los fármacos , Colon/patología , Relación Dosis-Respuesta a Droga , Glucógeno Sintasa Quinasa 3 beta , Masculino , Tamaño de los Órganos/efectos de los fármacos , Peroxidasa/análisis , Ratas , Ratas Wistar , Factor de Transcripción ReIA/análisis , Factor de Necrosis Tumoral alfa/análisis
4.
Eur J Pharmacol ; 522(1-3): 130-8, 2005 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-16213481

RESUMEN

The histamine H(4) receptor is a G-protein coupled receptor with little homology to the pro-inflammatory histamine H(1) receptor, expressed on cells of the immune system with hematopoietic lineage such as eosinophils and mast cells. The effects of the recently described highly selective histamine H(4) receptor antagonists JNJ 10191584 and JNJ 7777120 have now been investigated on the acute colitis provoked by trinitrobenzene sulphonic acid over 3 days in the rat. Treatment with JNJ 10191584 (10-100 mg/kg p.o., b.i.d.) caused a dose-dependent reduction in macroscopic damage, inhibition of the TNBS-provoked elevation of both colonic myeloperoxidase and tumour necrosis factor-alpha (TNF-alpha), and a reduction in the histologically assessed increase in mucosal and submucosal thickness and neutrophil infiltration. JNJ 7777120 (100 mg/kg p.o., b.i.d.) likewise reduced the macroscopic injury and the increases in colonic myeloperoxidase and TNF-alpha levels. These findings indicate a pro-inflammatory role for the histamine H(4) receptor in this model and suggest a novel pharmacological approach to the treatment of colitis.


Asunto(s)
Bencimidazoles/farmacología , Colitis/prevención & control , Indoles/farmacología , Piperazinas/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Enfermedad Aguda , Animales , Peso Corporal/efectos de los fármacos , Colitis/inducido químicamente , Colitis/metabolismo , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Relación Dosis-Respuesta a Droga , Masculino , Tamaño de los Órganos/efectos de los fármacos , Peroxidasa/metabolismo , Ratas , Ratas Wistar , Receptores Histamínicos , Receptores Histamínicos H4 , Ácido Trinitrobencenosulfónico , Factor de Necrosis Tumoral alfa/metabolismo
5.
Eur J Pharmacol ; 524(1-3): 169-73, 2005 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-16253235

RESUMEN

Salivary mucus and amylase have an anti-bacterial nature. Bacterial endotoxin is considered to decrease mucus secreting cell activity by nitric oxide-dependent mechanisms. In this study, the actions of endotoxin on amylase secreting cell activity have been studied. Endotoxin (Escherichia coli lipopolysaccharide; 3 mg/kg, i.v., 5 h) evoked nitric oxide synthase 2 (NOS2) induction in the rat whole parotid tissue (assessed by Western blot and the citrulline assay) and in rat isolated parotid acinar cells (assessed by Western blot and immunohistochemistry), and reduced basal and acetylcholine-stimulated amylase secretion from these isolated cells. However, N(G)-nitro-L-arginine methyl ester (0.1 mg/ml, 4 days in drinking water, yielding a dose of 25 mg/kg/day) did not affect amylase release under basal or acetylcholine-stimulated conditions, either in control acinar cells or those from endotoxin challenged rats. Thus, basal, acetylcholine-evoked or endotoxin-decreased cellular amylase secretion from rat isolated parotid acinar cells does not appear to be modulated by endogenous nitric oxide.


Asunto(s)
Amilasas/metabolismo , Lipopolisacáridos/farmacología , Óxido Nítrico/metabolismo , Glándula Parótida/efectos de los fármacos , Acetilcolina/farmacología , Animales , Calcio/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Edema/metabolismo , Edema/patología , Inhibidores Enzimáticos/farmacología , Inmunohistoquímica , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Glándula Parótida/metabolismo , Glándula Parótida/patología , Ratas , Ratas Wistar
6.
Curr Opin Pharmacol ; 4(6): 538-45, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15525540

RESUMEN

After a decade of intense pharmacological and drug development activity by the pharmaceutical industry, compounds derived from two key strategies for reducing gastrointestinal effects of non-steroidal anti-inflammatory drugs (NSAIDs) have been subjected to rigorous clinical appraisal. Despite the undoubted therapeutic and commercial success of the selective cyclooxygenase (COX)-2 inhibitors, known as the coxibs, with second-generation compounds already approved and launched, some concerns over their full gastrointestinal profile still linger, while the cardiovascular safety of this class has become a key issue. Likewise, Phase II evaluation of compounds incorporating a nitric oxide (NO)-donating moiety into standard NSAIDs (the NO-NSAIDs or CINODs) has created recent controversy over the full clinical profile of these compounds. Other approaches such as NO-COX-2 inhibitors and dual COX-lipoxygenase inhibitors are already warranting interest. It might therefore be too early to predict the eventual winning strategy for safer anti-inflammatory drugs.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Enfermedades Gastrointestinales/enzimología , Óxido Nítrico/fisiología , Prostaglandina-Endoperóxido Sintasas/efectos de los fármacos , Antiinflamatorios no Esteroideos/antagonistas & inhibidores , Antiinflamatorios no Esteroideos/uso terapéutico , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/efectos adversos , Inhibidores de la Ciclooxigenasa/uso terapéutico , Industria Farmacéutica/tendencias , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/metabolismo , Humanos , Proteínas de la Membrana , Estudios Multicéntricos como Asunto , Óxido Nítrico/uso terapéutico , Prostaglandina-Endoperóxido Sintasas/fisiología , Prostaglandina-Endoperóxido Sintasas/uso terapéutico
7.
Eur J Pharmacol ; 500(1-3): 427-39, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15464050

RESUMEN

By far the most attention has been paid to the deleterious actions of nonsteroidal anti-inflammatory drugs (NSAIDs), including isoform selective agents that inhibit cyclooxygenase (COX), on the upper gastrointestinal tract, particularly the gastric and duodenal mucosa. However, recent studies confirm a relatively high incidence of serious clinical events, especially with the more-established drugs of this class, involving the small intestine. Pathogenic factors that have been proposed from early studies in such enteropathy have included the enterohepatic circulation of the nonsteroidal anti-inflammatory drugs, inhibition of cyclooxygenase, surface epithelial changes and focal microvascular events. More recent work has concerned the role of infiltrating inflammatory cells, the relative roles of cyclooxygenase isoforms, COX-1 and COX-2 and the key involvement of inducible nitric oxide (NO) synthase and its product in combination with superoxide, peroxynitrite. In the present review, evidence for the underlying involvement of each these processes, and their sequential integration in the development of the intestinal injury and ulceration promoted by nonsteroidal anti-inflammatory drugs has been considered.


Asunto(s)
Inhibidores de la Ciclooxigenasa/efectos adversos , Enfermedades Gastrointestinales/inducido químicamente , Animales , Humanos , Úlcera/inducido químicamente
8.
Eur J Pharmacol ; 461(1): 63-71, 2003 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-12568917

RESUMEN

The role of bacteria and nitric oxide (NO), formed by the inducible isoform of NO synthase (iNOS), in a widespread systemic inflammatory microvascular response that follows indomethacin administration, has been investigated in the rat. Subcutaneous administration of indomethacin (10 mg kg(-1)) daily for 2 days produced an increase in microvascular leakage of radiolabelled albumin accompanied by expression of iNOS activity in the lung, liver, spleen and kidney, as well as in the jejunum, caecum, colon and ileum. Pretreatment with dexamethasone (1 mg kg(-1) day(-1), s.c.) reduced indomethacin-provoked microvascular leakage and the expression of iNOS activity in all the tissues studied. The widespread microvascular leakage and iNOS activity was also inhibited by pretreatment with ampicillin (200 mg kg(-1) day(-1), p.o.), metronidazole (200 mg kg(-1) day(-1), p.o.) or by polymyxin B (15 mg kg(-1) day(-1), s.c.). Administration of the highly selective iNOS inhibitor GW 273629 (3-[[2-(ethanimidoylamino)ethyl]sulphonyl]-L-alanine; five doses of 5 mg kg(-1), s.c. over 48 h) substantially inhibited the microvascular leakage in the affected organs. Such findings suggest the involvement of indigenous gut bacteria, lipopolysaccharide and iNOS expression following indomethacin-induced enteropathy in this widespread systemic inflammatory microvascular response.


Asunto(s)
Antiinflamatorios no Esteroideos , Bacterias/patogenicidad , Indometacina , Inflamación/metabolismo , Inflamación/microbiología , Óxido Nítrico Sintasa/metabolismo , Ampicilina/farmacología , Animales , Antibacterianos/farmacología , Antiinflamatorios/farmacología , Permeabilidad Capilar , Dexametasona/farmacología , Modelos Animales de Enfermedad , Inflamación/inducido químicamente , Intestinos/efectos de los fármacos , Intestinos/microbiología , Intestinos/patología , Masculino , Metronidazol/farmacología , Microcirculación , Insuficiencia Multiorgánica/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II , Especificidad de Órganos , Polimixina B/farmacología , Ratas , Ratas Wistar , Albúmina Sérica Radioyodada , Sulfonas/farmacología
9.
Fundam Clin Pharmacol ; 17(3): 301-13, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12803569

RESUMEN

Non-steroidal anti-inflammatory drugs (NSAIDs) causes extensive damage to the gastrointestinal (GI) tract. The underlying mechanisms of gastric injury include topical irritant actions that disrupt the epithelial barrier, as well as the inhibition of cyclo-oxygenase (COX), which is predominantly the COX-1 isoform in the mucosa. This damage can be attenuated by antisecretory agents or by mucosal protective agents such as the synthetic prostanoids or nitric oxide (NO) donors. Compounds designed to attenuate topical irritancy, or have protective agents incorporated, such as NO-containing NSAIDs, the CINODs (cyclo-oxygenase-inhibiting NO-donating drugs) show reduced mucosal injury. NSAIDs also cause injury in the small intestine, which appears to result from initial COX inhibition, with subsequent translocation of indigenous bacteria, induction of NO synthase and production of the cytotoxic moiety, peroxynitrite. The COX-2 selective agents, the coxibs, which inhibit prostanoid biosynthesis at inflammatory sites, but not the endogenous protective prostanoids in the gut formed by COX-1, have proved so far to be a successful therapeutic approach to reducing NSAIDs GI damage. The clinical outcome of the use of the second generation of coxibs, and the newer NO NSAIDs is now awaited.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Mucosa Gástrica/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Antiinflamatorios no Esteroideos/uso terapéutico , Ensayos Clínicos como Asunto , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/efectos adversos , Inhibidores de la Ciclooxigenasa/metabolismo , Diseño de Fármacos , Mucosa Gástrica/patología , Enfermedades Gastrointestinales/inducido químicamente , Enfermedades Gastrointestinales/patología , Enfermedades Gastrointestinales/prevención & control , Humanos , Mucosa Intestinal/patología , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Proteínas de la Membrana , Donantes de Óxido Nítrico/efectos adversos , Donantes de Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/uso terapéutico , Prostaglandina-Endoperóxido Sintasas/metabolismo , Prostaglandinas/metabolismo
10.
IDrugs ; 5(6): 590-3, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12802708

RESUMEN

Ono Pharmaceuticals is developing ONO-1714, a selective inducible NO synthase modulator, for the potential treatment of sepsis and hypotension. By March 1999, ONO-1714 had entered phase I trials [354023]. By June 2002, a phase II trial in hypotension during dialysis was ongoing in Japan and clinical pharmacology trials were underway in the UK [446138]. In August 1999, Lehmann Brothers gave ONO-1714 a 10% probability of reaching the market with an expected launch in 2004. Sales were expected to peak at 50 million US dollars in 2012 [349228].

11.
Methods Mol Biol ; 644: 189-200, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20645175

RESUMEN

A prevalent unwanted action of cyclo-oxygenase (COX) inhibitors, as exemplified by the non-steroidal anti-inflammatory drugs (NSAIDs), is their potential to produce gastrointestinal side effects in clinical use. The injury provoked by such agents includes rapid superficial disruption to the surface layer of the gastric mucosa, the production of acute gastric erosions in the corpus region and the formation of ulcers in the antral region of the stomach. The small intestine is also adversely affected, with a developing enteropathy over a more protracted period that causes lesions and inflammation in the gut. From experimental work, the interactive mechanisms of such damage in the stomach differ distinctly from those that underlie the intestinal injury, yet the damage in both regions involves the inhibition of both COX-1 and COX-2 isoforms. This chapter outlines the in vivo methods that can be used to identify the potential for novel NSAIDs and selective COX-inhibitors to produce acute gastric corpus lesions and more-chronic antral ulcers in the rat, as well as causing small intestinal enteropathy. Such methods can also be utilized to evaluate the ability of novel agents to prevent the gastrointestinal injury provoked by NSAIDs or COX-inhibitors.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Inhibidores de la Ciclooxigenasa/efectos adversos , Tracto Gastrointestinal/efectos de los fármacos , Prostaglandina-Endoperóxido Sintasas/metabolismo , Animales , Mucosa Gástrica/efectos de los fármacos , Intestino Delgado/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Úlcera Gástrica/inducido químicamente
12.
Pharmacol Rep ; 62(3): 548-56, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20631420

RESUMEN

Although a variety of pharmaceutical preparations of aminosalicylate are commonly used in the clinic for the control of inflammatory bowel disease, the mechanisms underlying their therapeutic actions remain unclear. Recent in vivo and in vitro studies have demonstrated that 5-aminosalicylic acid (5-ASA), regarded as the active moiety in aminosalicylate preparations such as sulfasalazine, can induce the heat shock protein, heme oxygenase-1 (HO-1) and up-regulate HO enzyme activity in the colon. As HO-1 can produce endogenous anti-oxidant and anti-inflammatory moieties such as bilirubin and carbon monoxide (CO), these findings suggest a novel mechanism of action for aminosalicylates, acting as anti-colitic agents through the up-regulation of HO-1 enzyme expression and activity.


Asunto(s)
Ácidos Aminosalicílicos/uso terapéutico , Colon/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Ácidos Aminosalicílicos/metabolismo , Ácidos Aminosalicílicos/farmacología , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Colitis/tratamiento farmacológico , Colitis/enzimología , Colitis/metabolismo , Colon/enzimología , Colon/metabolismo , Humanos , Enfermedades Inflamatorias del Intestino/enzimología , Enfermedades Inflamatorias del Intestino/metabolismo , Mesalamina/farmacología , Mesalamina/uso terapéutico , Ratones , Ratas , Regulación hacia Arriba
14.
IDrugs ; 5(7): 623-5, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-14686336
15.
IDrugs ; 5(12): 1113-7, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14689951
16.
Expert Opin Investig Drugs ; 14(11): 1347-58, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16255675

RESUMEN

Almost 20 years after the identification of the biological role of nitric oxide (NO), the full therapeutic potential of novel agents that mimic the activity of NO or interfere with its synthesis has yet to be realised for utilities involving the gastrointestinal tract. New utilities for classical NO donors, which were used as vasodilators for decades, in the treatment of motility disorders have been explored and a product for treating anal fissure was recently launched. New classes of compounds incorporating a NO-donating moiety into standard non-steroidal anti-inflammatory drugs, the NO-non-steroidal anti-inflammatory drugs (NO-NSAIDs) or COX-inhibiting nitric oxide donors (CINODs) have also been developed. These have been shown to exhibit reduced gastrointestinal injury in experimental models, and reports on their efficacy and safety in Phase I and II studies are now available. Modulation of the inducible NO synthase isoform that generates excessive NO that can lead to subsequent cytotoxic moieties, such as peroxynitrite, may have therapeutic possibilities in a range of inflammatory diseases of the gut. Likewise, agents that promote the decomposition of peroxynitrite or removal of its other component, superoxide, may also prove to be of use. Further targets for pharmaceutical exploitation are likely to come from both genomic and molecular insights into the processes that regulate the NO system.


Asunto(s)
Enfermedades Gastrointestinales/tratamiento farmacológico , Donantes de Óxido Nítrico/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Aspirina/análogos & derivados , Aspirina/uso terapéutico , Enteritis/etiología , Esófago/efectos de los fármacos , Esófago/fisiología , Fisura Anal/tratamiento farmacológico , Vaciamiento Gástrico/efectos de los fármacos , Motilidad Gastrointestinal/efectos de los fármacos , Humanos , Naproxeno/análogos & derivados , Naproxeno/uso terapéutico , Nitratos/uso terapéutico , Óxido Nítrico/fisiología , Óxido Nítrico/uso terapéutico , Óxido Nítrico Sintasa de Tipo II/efectos de los fármacos , Ácido Peroxinitroso/fisiología
17.
Inflammopharmacology ; 11(4): 415-22, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-15035794

RESUMEN

Nitric oxide (NO) can protect the gastrointestinal tract from injury, including that provoked by non-steroidal anti-inflammatory drugs (NSAIDs). This protective profile of NO, which predominantly reflects actions on the microcirculation, is mimicked by NO donors. Moreover, the NO-donating agents know as the NO-NSAIDs or CINODs (cyclo-oxygenase-inhibiting nitric oxide-donating drugs) exhibit reduced gut injury in experimental models, which is considered to reflect these local beneficial actions of NO. NSAIDs cause chronic inflammatory lesions in the small intestine in experimental models. This injury results from initial COX inhibition and other local events, with translocation of indigenous luminal bacteria, leading to induction of NO synthase isoform, iNOS, and subsequent production of the cytotoxic moiety, peroxynitrite from NO and superoxide. Agents that inhibit iNOS or superoxide production can attenuate such intestinal injury. In the absence of reactive oxygen moieties, NO may play a beneficial role in the resolution of inflammatory damage to the gut, thus reconciling the potential opposing properties of NO in tissue inflammation and injury.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda