Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Inflammopharmacology ; 32(3): 2049-2060, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38570398

RESUMEN

Gastric ulcers affect approx. 10% of population. Non-steroidal anti-inflammatory drugs (NSAIDs), including acetylsalicylic acid (ASA) predispose to or impair the physiologically complex healing of pre-existing ulcers. Since H2S is an endogenous cytoprotective molecule, we hypothesized that new H2S-releasing ASA-derivative (ATB-340) could overcome pathological impact of NSAIDs on GI regeneration.Clinically translational gastric ulcers were induced in Wistar rats using state-of-the-art microsurgical model employing serosal application of acetic acid. This was followed by 9 days long i.g. daily treatment with vehicle, ATB-340 (6-24 mg/kg) or equimolar ASA doses (4-14 mg/kg). Ulcer area was assessed macro- and microscopically. Prostaglandin (PG)E2  levels, indicating pharmacological activity of NSAIDs and 8-hydroxyguanozine content, reflecting nucleic acids oxidation in serum/gastric mucosa, were determined by ELISA. Qualitative and/or quantitative pathway-specific alterations at the ulcer margin were evaluated using real-time PCR and mass spectrometry-based proteomics.ASA, unlike ATB-340, dose-dependently delayed/impaired gastric tissue recovery, deregulating 310 proteins at the ulcer margin, including Ras signalling, wound healing or apoptosis regulators. ATB-340 maintained NSAIDs-specific cyclooxygenase-inhibiting capacity on systemic and GI level but in time-dependent manner. High dose of ATB-340 (24 mg/kg daily), but not ASA, decreased nucleic acids oxidation and upregulated anti-oxidative/anti-inflammatory heme oxygenase-1, 24-dehydrocholesterol reductase or suppressor of cytokine signalling (SOCS3) at the ulcer margin.Thus, ASA impairs the physiological healing of pre-existing gastric ulcers, inducing the extensive molecularly functional and proteomic alterations at the wound margin. H2S-releasing ATB-340 maintains the target activity of NSAIDs with limited impact on gastric PGE2 signalling and physiological GI regeneration, enhancing anti-inflammatory and anti-oxidative response, and providing the pharmacological advantage.


Asunto(s)
Antiinflamatorios no Esteroideos , Aspirina , Mucosa Gástrica , Sulfuro de Hidrógeno , Proteómica , Ratas Wistar , Úlcera Gástrica , Cicatrización de Heridas , Animales , Úlcera Gástrica/tratamiento farmacológico , Úlcera Gástrica/metabolismo , Aspirina/farmacología , Ratas , Proteómica/métodos , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/farmacología , Masculino , Cicatrización de Heridas/efectos de los fármacos , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Dinoprostona/metabolismo , Enfermedad Crónica , Relación Dosis-Respuesta a Droga , Modelos Animales de Enfermedad , Naproxeno/análogos & derivados
2.
FASEB J ; 35(10): e21913, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34555204

RESUMEN

ATB-346 is a hydrogen sulfide-releasing non-steroidal anti-inflammatory drug (H2 S-NSAID) derived from naproxen, which in preclinical studies has been shown to have markedly reduced gastrointestinal adverse effects. However, its anti-inflammatory properties in humans compared to naproxen are yet to be confirmed. To test this, we used a dermal model of acute inflammation in healthy, human volunteers, triggered by ultraviolet-killed Escherichia coli. This robust model allows quantification of the cardinal signs of inflammation along with cellular and humoral factors accumulating within the inflamed skin. ATB-346 was non-inferior to naproxen in terms of its inhibition of cyclooxygenase activity as well as pain and tenderness. ATB-346 significantly inhibited neutrophil infiltration at the site of inflammation at 4 h, compared to untreated controls. Subjects treated with ATB-346 also experienced significantly reduced pain and tenderness compared to healthy controls. Furthermore, both classical and intermediate monocyte subsets infiltrating the site of inflammation at 48 h expressed significantly lower levels of CD14 compared to untreated controls, demonstrating a shift toward an anti-inflammatory phenotype. Collectively, we have shown for the first time in humans that ATB-346 is potently anti-inflammatory and propose that ATB-346 represents the next generation of H2 S-NSAIDs, as a viable alternative to conventional NSAIDs, with reduced adverse effects profile.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Inflamación/tratamiento farmacológico , Naproxeno/análogos & derivados , Adolescente , Adulto , Dinoprostona/metabolismo , Escherichia coli/inmunología , Escherichia coli/efectos de la radiación , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Masculino , Persona de Mediana Edad , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/inmunología , Naproxeno/metabolismo , Naproxeno/farmacología , Naproxeno/uso terapéutico , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Dolor/metabolismo , Fenotipo , Solubilidad , Rayos Ultravioleta , Vasoconstricción/efectos de los fármacos , Adulto Joven
4.
J Pediatr Gastroenterol Nutr ; 68(5): 623-629, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31022092

RESUMEN

BACKGROUND AND OBJECTIVE: The use of nonsteroidal anti-inflammatory drugs (NSAIDs) is associated with a broad spectrum of life-threatening adverse effects on the immature gastrointestinal tract. NSAID derivatives exploiting the beneficial effects of biologically active gases, such as hydrogen sulfide (H2S), have been developed. Herein, we determined the effects of ketoprofen and ATB-352, a H2S-releasing ketoprofen derivative, on selected metabolic pathways previously identified to be significantly altered by indomethacin in the human immature intestine. METHODS: Ketoprofen and ATB-352 were tested on human mid-gestation small intestinal explants maintained in a serum-free organ culture system for 48 hours. The expression levels of the representative genes involved in selected metabolic pathways were measured by real-time PCR after a treatment of 48 hours. RESULTS: Tested at a concentration that allows more than 80% inhibition of PGE2 production, ketoprofen was found to be less damaging than indomethacin at an equivalent dosage. However, based on the inducibility of cyclooxygenase-2 transcript expression, we were able to discriminate between responder individuals in which the deleterious effects observed with indomethacin were attenuated, and non-responder specimens in which the effects were similar to those observed with indomethacin. ATB-352 did not induce significant changes compared to ketoprofen on these metabolic pathways. CONCLUSIONS: These results show less damaging effects of ketoprofen compared to indomethacin on the immature intestine and indicate that the intestinal response to this NSAID significantly varies between individuals. However, the results did not allow us to demonstrate a specific beneficial effect of H2S release in organ culture.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Indometacina/farmacología , Intestino Delgado/efectos de los fármacos , Cetoprofeno/farmacología , Transducción de Señal/efectos de los fármacos , Ciclooxigenasa 2/efectos de los fármacos , Humanos , Intestino Delgado/embriología , Cetoprofeno/análogos & derivados
5.
Semin Immunol ; 27(3): 227-33, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26095908

RESUMEN

There are numerous gaseous substances that can act as signaling molecules, but the best characterized of these are nitric oxide, hydrogen sulfide and carbon monoxide. Each has been shown to play important roles in many physiological and pathophysiological processes. This article is focused on the effects of these gasotransmitters in the context of inflammation. There is considerable overlap in the actions of nitric oxide, hydrogen sulfide and carbon monoxide with respect to inflammation, and these mediators appear to act primarily as anti-inflammatory substances, promoting resolution of inflammatory processes. They also have protective and pro-healing effects in some tissues, such as the gastrointestinal tract and lung. Over the past two decades, significant progress has been made in the development of novel anti-inflammatory and cytoprotective drugs that release of one or more of these gaseous mediators.


Asunto(s)
Antiinflamatorios/uso terapéutico , Monóxido de Carbono/uso terapéutico , Sulfuro de Hidrógeno/uso terapéutico , Inflamación/tratamiento farmacológico , Óxido Nítrico/uso terapéutico , Apoptosis/efectos de los fármacos , Citoprotección/efectos de los fármacos , Gasotransmisores/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
6.
Proc Natl Acad Sci U S A ; 113(2): 434-9, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26712011

RESUMEN

Cyclooxygenase-2 (COX-2) is an inducible enzyme that drives inflammation and is the therapeutic target for widely used nonsteroidal antiinflammatory drugs (NSAIDs). However, COX-2 is also constitutively expressed, in the absence of overt inflammation, with a specific tissue distribution that includes the kidney, gastrointestinal tract, brain, and thymus. Constitutive COX-2 expression is therapeutically important because NSAIDs cause cardiovascular and renal side effects in otherwise healthy individuals. These side effects are now of major concern globally. However, the pathways driving constitutive COX-2 expression remain poorly understood. Here we show that in the kidney and other sites, constitutive COX-2 expression is a sterile response, independent of commensal microorganisms and not associated with activity of the inflammatory transcription factor NF-κB. Instead, COX-2 expression in the kidney but not other regions colocalized with nuclear factor of activated T cells (NFAT) transcription factor activity and was sensitive to inhibition of calcineurin-dependent NFAT activation. However, calcineurin/NFAT regulation did not contribute to constitutive expression elsewhere or to inflammatory COX-2 induction at any site. These data address the mechanisms driving constitutive COX-2 and suggest that by targeting transcription it may be possible to develop antiinflammatory therapies that spare the constitutive expression necessary for normal homeostatic functions, including those important to the cardiovascular-renal system.


Asunto(s)
Ciclooxigenasa 2/genética , FN-kappa B/metabolismo , Factores de Transcripción NFATC/genética , Transducción de Señal , Transcripción Genética , Animales , Ciclooxigenasa 2/metabolismo , Ciclosporina/farmacología , Citocinas/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Vida Libre de Gérmenes , Riñón/efectos de los fármacos , Riñón/metabolismo , Lipopolisacáridos/farmacología , Luciferasas/metabolismo , Masculino , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Distribución Tisular/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
7.
Am J Physiol Gastrointest Liver Physiol ; 314(2): G143-G149, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29025733

RESUMEN

A diverse range of effects of the intestinal microbiota on mucosal defense and injury has become increasingly clear over the past decade. Hydrogen sulfide (H2S) has emerged as an important mediator of many physiological functions, including gastrointestinal mucosal defense and repair. Hydrogen sulfide is produced by gastrointestinal tract tissues and by bacteria residing within the gut and can influence the function of a wide range of cells. The microbiota also appears to be an important target of hydrogen sulfide. H2S donors can modify the gut microbiota, and the gastrointestinal epithelium is a major site of oxidation of microbial-derived H2S. When administered together with nonsteroidal anti-inflammatory drugs, H2S can prevent some of the dysbiosis those drugs induce, possibly contributing to the observed prevention of gastrointestinal damage. Exogenous H2S can also markedly reduce the severity of experimental colitis and plays important roles in modulating epithelial cell-mucus-bacterial interactions in the intestine, contributing to its ability to promote resolution of inflammation and repair of tissue injury. In this paper we review recent studies examining the roles of H2S in mucosal defense, the possibility that H2S can damage the gastrointestinal epithelium, and effects of H2S on the gut microbiota and on mucus and biofilm interactions in the context of intestinal inflammation.


Asunto(s)
Bacterias/metabolismo , Microbioma Gastrointestinal , Sulfuro de Hidrógeno/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Animales , Antibacterianos/toxicidad , Antiinflamatorios no Esteroideos/toxicidad , Bacterias/efectos de los fármacos , Bacterias/crecimiento & desarrollo , Biopelículas/crecimiento & desarrollo , Disbiosis , Microbioma Gastrointestinal/efectos de los fármacos , Interacciones Huésped-Patógeno , Humanos , Sulfuro de Hidrógeno/farmacología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Probióticos/farmacología
8.
Pharmacol Res ; 132: 220-231, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29287688

RESUMEN

Periodontal disease is the most common cause of tooth loss in humans, is an inflammatory disease initiated by oral microbial biofilm. Given the involvement of the inflammatory pathway in this type of pathology, the main pharmacological strategy for the treatment of periodontitis, is the inhibition of the inflammatory process in order to prevent tissue destruction and bone resorption, a condition associated with a painful state. To do this, the best class of drugs are Non-steroidal anti-inflammatory drugs (NSAIDs), however, the presence of side effects, especially at the gastrointestinal tract, limits their use for long-term therapy. Recently, some evidence shows that derivatives of NSAIDs capable of releasing hydrogen sulphide exhibit lower collateral effects, particularly at the gastric level. In fact, H2S is an endogenous gaseous mediator with a cytoprotective role at the gastric level. In this study, we have compared the protective effects of ketoprofen with ATB-352, a hydrogen sulfide-releasing derivative of ketoprofen, in an experimental model of periodontitis in rat. Periodontitis was induced by a single intragingival injection of 1 µl LPS (10 µg/µl), Our results show that 14 h after intragingival injection of LPS, there was a high tissue damage associated with bone resorption, and in gingivomucosal tissues there was a significant expression of NF-kb p65 and pro-inflammatory cytokine as well as a higher expression of COX-2 and iNOS, activation of the apoptotic process, and also increased levels of NGF expression, often associated with a higher nociceptive perception. Treatment with ATB-352 at the dose of 20mg\kg, was able to reduce the inflammatory process associated with intragingival LPS injection and also had a positive effect on bone resorption and tissue damage.


Asunto(s)
Antiinflamatorios/uso terapéutico , Sulfuro de Hidrógeno/administración & dosificación , Cetoprofeno/análogos & derivados , Cetoprofeno/uso terapéutico , Periodontitis/tratamiento farmacológico , Animales , Antiinflamatorios/farmacología , Resorción Ósea , Ciclooxigenasa 2/metabolismo , Interleucina-1beta/metabolismo , Cetoprofeno/farmacología , Lipopolisacáridos , Masculino , Mucosa Bucal/efectos de los fármacos , Mucosa Bucal/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Periodontitis/inducido químicamente , Periodontitis/metabolismo , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/metabolismo
9.
Dig Dis Sci ; 62(9): 2223-2230, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28733867

RESUMEN

Of the numerous gaseous substances that can act as signaling molecules, the best characterized are nitric oxide, carbon monoxide and hydrogen sulfide. Contributions of each of these low molecular weight substances, alone or in combination, to maintenance of gastrointestinal mucosal integrity have been established. There is considerable overlap in the actions of these gases in modulating mucosal defense and responses to injury, and in some instances they act in a cooperative manner. Each also play important roles in regulating inflammatory and repair processes throughout the gastrointestinal tract. In recent years, significant progress has been made in the development of novel anti-inflammatory and cytoprotective drugs that exploit the beneficial activities of one or more of these gaseous mediators.


Asunto(s)
Monóxido de Carbono/metabolismo , Gasotransmisores/metabolismo , Tracto Gastrointestinal/metabolismo , Sulfuro de Hidrógeno/metabolismo , Óxido Nítrico/metabolismo , Animales , Monóxido de Carbono/antagonistas & inhibidores , Gasotransmisores/antagonistas & inhibidores , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Fármacos Gastrointestinales/farmacología , Fármacos Gastrointestinales/uso terapéutico , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/patología , Humanos , Sulfuro de Hidrógeno/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Óxido Nítrico/antagonistas & inhibidores
10.
Proc Natl Acad Sci U S A ; 111(37): 13559-64, 2014 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-25187563

RESUMEN

Vitamin B deficiencies, which can lead to hyperhomocysteinemia (Hhcy), are commonly reported in patients with inflammatory bowel disease (IBD) and may be a causative underlying factor. However, the mechanism for this effect is not known. Hydrogen sulfide (H2S) is a gaseous mediator that promotes tissue repair and resolution of inflammation. In experimental colitis, a marked increase in colonic H2S synthesis drives ulcer healing and resolution of inflammation. Because H2S synthesis is in part dependent upon enzymes that require vitamin B6 as a cofactor, we tested the hypothesis that Hhcy in rodent models would increase the susceptibility to colitis. In all three models tested, diet-induced Hhcy significantly exacerbated colitis. The usual elevation of colonic H2S synthesis after induction of colitis was absent in all three models of colitis. Administration of an H2S donor to Hhcy rats significantly decreased the severity of colitis. Compared with wild-type mice, interleukin (IL) 10-deficient mice on a normal diet had decreased levels of colonic H2S synthesis, a 40% increase in serum homocysteine, and a phenotype similar to wild-type mice with Hhcy. IL-10-deficient mice fed the vitamin B-deficient diet exhibited more severe colonic inflammation, but the normal elevation of colonic H2S synthesis was absent. Administration of IL-10 to the IL-10-deficient mice restored colonic H2S synthesis and significantly decreased serum homocysteine levels. These results suggest that the exacerbation of colitis in Hhcy is due in part to impaired colonic H2S synthesis. Moreover, IL-10 plays a novel role in promoting H2S production and homocysteine metabolism, which may have therapeutic value in conditions characterized by Hhcy.


Asunto(s)
Colitis/complicaciones , Progresión de la Enfermedad , Sulfuro de Hidrógeno/metabolismo , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/metabolismo , Interleucina-10/metabolismo , Transducción de Señal , Animales , Colitis/inducido químicamente , Colitis/patología , Colon/metabolismo , Colon/patología , Sulfato de Dextran , Dieta , Humanos , Hiperhomocisteinemia/patología , Interleucina-10/deficiencia , Masculino , Ratones Endogámicos C57BL , Peroxidasa/metabolismo , Ratas Wistar , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología
11.
FASEB J ; 29(4): 1591-602, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25550470

RESUMEN

During a course of colitis, production of the gaseous mediator hydrogen sulfide (H2S) is markedly up-regulated at sites of mucosal damage and contributes significantly to healing and resolution of inflammation. The signaling mechanisms through which H2S promotes resolution of colitis are unknown. We hypothesized that the beneficial effects of H2S in experimental colitis are mediated via stabilization of hypoxia-inducible factor (HIF)-1α. The hapten dinitrobenzene sulfonic acid was used to induce colitis in rats and mice. This resulted in an elevated expression of the H2S-producing enzyme, cystathionine γ-lyase (CSE), and HIF-1α at sites of mucosal ulceration, and the expression of these 2 enzymes followed a similar pattern throughout the course of colitis. This represented a functionally important relationship because the loss of CSE-derived H2S production led to decreased HIF-1α stabilization and exacerbation of colitis. Furthermore, application of an H2S-releasing molecule, diallyl disulfide (DADS), stabilized colonic HIF-1α expression, up-regulated hypoxia-responsive genes, and reduced the severity of disease during peak inflammation. Importantly, the ability of DADS to promote the resolution of colitis was abolished when coadministered with an inhibitor of HIF-1α in vivo (PX-478). DADS was also able to maintain HIF-1α expression at a later point in colitis, when HIF-1α levels would have normally returned to control levels, and to enhance resolution. Finally, we found that HIF-1α stabilization inhibited colonic H2S production and may represent a negative feedback mechanism to prevent prolonged HIF-1α stabilization. Our findings demonstrate an important link between H2S and HIF-1α in the resolution of inflammation and injury during colitis and provide mechanistic insights into the therapeutic value of H2S donors.


Asunto(s)
Colitis/metabolismo , Sulfuro de Hidrógeno/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Compuestos Alílicos/farmacología , Animales , Bencenosulfonatos/toxicidad , Colitis/tratamiento farmacológico , Colitis/patología , Cistationina gamma-Liasa/antagonistas & inhibidores , Cistationina gamma-Liasa/metabolismo , Modelos Animales de Enfermedad , Disulfuros/farmacología , Expresión Génica , Células HT29 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Compuestos de Mostaza/farmacología , Fenilpropionatos/farmacología , Estabilidad Proteica , Ratas , Ratas Wistar , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
12.
Pharmacol Res ; 111: 652-658, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27475881

RESUMEN

For many years it has been recognized that inhibition of cyclooxygenase enzymes is effective in reducing the incidence of many types of cancer, but the adverse effects of these drug, particularly in the gastrointestinal and cardiovascular systems, limits their utility. Recently developed hydrogen sulfide-releasing anti-inflammatory drugs may be a promising option for cancer chemoprevention. In this paper we review evidence suggesting that these novel compounds are effective in a range of animal models of various types of cancer, while exhibiting greatly reduced toxicity relative to currently marketed non-steroidal anti-inflammatory drugs. Some of the possible mechanisms of action of hydrogen sulfide-releasing anti-inflammatory drugs are also discussed.


Asunto(s)
Antiinflamatorios/uso terapéutico , Anticarcinógenos/uso terapéutico , Sulfuro de Hidrógeno/uso terapéutico , Animales , Antiinflamatorios/efectos adversos , Antiinflamatorios/farmacología , Anticarcinógenos/efectos adversos , Anticarcinógenos/farmacología , Quimioprevención , Humanos , Sulfuro de Hidrógeno/efectos adversos , Sulfuro de Hidrógeno/farmacología , Neoplasias Intestinales/tratamiento farmacológico , Neoplasias Intestinales/prevención & control , Melanoma/prevención & control
13.
Pharmacol Res ; 114: 67-73, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27777130

RESUMEN

Inflammation plays a key role in tumor promotion and development. Indeed, cyclooxygenase-2 (COX-2) expression is strongly associated with different types of cancer. An emerging class of compounds with significant anti-inflammatory properties is the hydrogen sulfide-releasing non-steroidal anti-inflammatory drugs (H2S-NSAIDs). They consist of a traditional NSAID to which an H2S-releasing moiety is covalently attached. We have recently demonstrated that H2S donors inhibit melanoma cell proliferation. In the current study, we evaluated the potential beneficial effects of a new H2S-releasing derivative of naproxen, ATB-346 [2-(6-methoxynapthalen-2-yl)-propionic acid 4-thiocarbamoyl phenyl ester] which inhibits COX activity but also releases H2S. We used cell culture and a mouse melanoma model to evaluate the effect of ATB-346 on: i) in vitro growth of human melanoma cells; ii) in vivo melanoma development in mice. Cell culture studies demonstrated that ATB-346 reduced the in vitro proliferation of human melanoma cells and this effect was associated to induction of apoptosis and inhibition of NF-κB activation. Moreover, ATB-346 had novel Akt signaling inhibitory properties. Daily oral dosing of ATB-346 (43µmol/kg) significantly reduced melanoma development in vivo. This study shows that ATB-346, a novel H2S-NSAID, inhibits human melanoma cell proliferation by inhibiting pro-survival pathways associated with NF-κB and Akt activation. Furthermore, oral treatment with ATB-346 inhibits melanoma growth in mice. In conclusion, the combination of inhibition of cyclooxygenase and delivery of H2S by ATB-346 may offer a promising alternative to existing therapies for melanoma.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antineoplásicos/uso terapéutico , Melanoma/tratamiento farmacológico , Naproxeno/análogos & derivados , Animales , Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiocinas/inmunología , Femenino , Humanos , Sulfuro de Hidrógeno/inmunología , Melanoma/inmunología , Melanoma/patología , Ratones Endogámicos C57BL , FN-kappa B/inmunología , Naproxeno/farmacología , Naproxeno/uso terapéutico
14.
BMC Gastroenterol ; 16(1): 58, 2016 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-27259970

RESUMEN

BACKGROUND: Rebamipide is a gastroprotective agent with promising results against gastric damage induced by non-steroidal anti-inflammatory drugs. The present study evaluated if rebamipide protects against naproxen-induced gastric damage in healthy volunteers. Changes in gastric PGE2 tissue concentration were also evaluated. METHODS: After a preliminary endoscopy to rule out previous gastric macroscopic damage, twenty-four healthy volunteers of both sexes were divided into 2 groups. One group received sodium naproxen 550 mg b.i.d. plus placebo for 7 days, while the other group received sodium naproxen 550 mg b.i.d. plus rebamipide 100 mg b.i.d. At the end of treatment, a new endoscopy was performed. Gastric macroscopic damage was evaluated by the Cryer score and by the modified Lanza score. The primary outcome measure of the trial was the macroscopic damage observed in each treatment group at the end of treatment. Biopsies were collected at both endoscopies for PGE2 quantification and histopathological analysis (secondary outcomes). Tissue PGE2 was quantified by ELISA. The randomization sequence was generated using 3 blocks of 8 subjects each. Volunteers and endoscopists were blind to whether they were receiving rebamipide or placebo. RESULTS: All recruited volunteers completed the trial. Sodium naproxen induced gastric damage in both groups. At the end of the study, median Cryer score was 4 in both groups (Difference = 0; 95%CI = -1 to 0; p = 0.728). In the placebo group, the mean tissue PGE2 concentration was 1005 ± 129 pg/mL before treatment and 241 ± 41 pg/mL after treatment (p < 0.001). In the rebamipide group, the mean tissue PGE2 concentration was 999 ± 109 pg/mL before treatment, and 168 ± 13 pg/mL after treatment (p < 0.001). There was no difference in mean tissue PGE2 between the two groups (difference = 5; 95%CI from -334.870 to 345.650; p = 0.975). No significant change was observed at the histopathological evaluation, despite the evident macroscopic damage induced by naproxen. CONCLUSION: Rebamipide does not protect against naproxen-induced gastric damage in healthy volunteers. TRIAL REGISTRATION: ClinicalTrials.gov, NCT02632812 . Registered 14 December 2015.


Asunto(s)
Alanina/análogos & derivados , Antiinflamatorios no Esteroideos/efectos adversos , Fármacos Gastrointestinales/uso terapéutico , Naproxeno/efectos adversos , Quinolonas/uso terapéutico , Gastropatías/inducido químicamente , Gastropatías/prevención & control , Adolescente , Adulto , Alanina/uso terapéutico , Dinoprostona/análisis , Método Doble Ciego , Femenino , Mucosa Gástrica/patología , Humanos , Masculino , Persona de Mediana Edad , Gastropatías/patología , Adulto Joven
15.
Am J Physiol Gastrointest Liver Physiol ; 308(12): G994-1003, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25882612

RESUMEN

The small intestine is a significant site of ulceration and bleeding induced by nonsteroidal anti-inflammatory drugs (NSAIDs). The pathogenesis is poorly understood. The present study explored the roles of bile, bacteria, and enterohepatic circulation to NSAID enteropathy, using both a conventional NSAID (naproxen) and a gastrointestinal-safe naproxen derivative (ATB-346), as well as proton pump inhibitors (PPIs). Rats were treated orally with naproxen or equimolar doses of ATB-346 over a 5-day period, with or without PPI administration, and intestinal damage was quantified. The cytotoxicity of bile from the rats was evaluated in vitro. Biliary excretion of naproxen and ATB-346 was determined. The impact of the NSAIDs and of PPIs on the composition of the intestinal microbiota was examined by deep sequencing of 16s rRNA. Naproxen caused significant intestinal damage and inflammation, whereas ATB-346 did not. Naproxen, but not ATB-346, dose dependently increased the cytotoxicity of bile, and it was further increased by PPI coadministration. Whereas biliary excretion of naproxen was significant in naproxen-treated rats, it was greatly reduced in rats treated with ATB-346. The enteric microbiota of naproxen-treated rats was distinct from that in vehicle- or ATB-346-treated rats, and PPI administration caused significant intestinal dysbiosis. The increase in cytotoxicity of bile induced by naproxen and PPIs may contribute significantly to intestinal ulceration and bleeding. Some of these effects may occur secondary to significant changes in the jejunal microbiota induced by both naproxen and PPIs.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Sulfuro de Hidrógeno/farmacología , Enfermedades Intestinales/tratamiento farmacológico , Naproxeno/análogos & derivados , Inhibidores de la Bomba de Protones/farmacología , Animales , Eliminación Hepatobiliar/fisiología , Inflamación/tratamiento farmacológico , Masculino , Naproxeno/farmacología , ARN Ribosómico 16S/genética , Ratas Wistar
16.
Am J Pathol ; 184(8): 2154-5, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25016432

RESUMEN

This Commentary highlights the article by Montero-Melendez et al, revealing novel protective properties of endogenous melanocortin receptor 3 on periodontal status in health and disease and suggesting a new genus of anti-arthritic bone-sparing therapeutics.


Asunto(s)
Artritis Experimental/metabolismo , Artritis Reumatoide/metabolismo , Enfermedades Periodontales/metabolismo , Receptor de Melanocortina Tipo 3/metabolismo , Animales , Masculino
17.
Nitric Oxide ; 46: 25-31, 2015 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-25461269

RESUMEN

Hydrogen sulfide exerts a number of cytoprotective and anti-inflammatory effects in many organ systems. In an effort to exploit these potent and beneficial effects, a number of hydrogen sulfide-releasing derivatives of existing drugs have been developed and extensively tested in pre-clinical models. In particular, efforts have been made by several groups to develop hydrogen sulfide-releasing derivatives of a number of nonsteroidal anti-inflammatory drugs. The main goal of this approach is to reduce the gastrointestinal ulceration and bleeding caused by this class of drugs, particularly when used chronically such as in the treatment of arthritis. However, these drugs may also have utility for prevention of various types of cancer. This paper provides an overview of some of the mechanisms underlying the anti-inflammatory and cytoprotective actions of hydrogen sulfide. It also gives some examples of hydrogen sulfide-releasing anti-inflammatory drugs, and their actions in terms of reducing inflammation and attenuating the development of cancer in experimental models.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Sulfuro de Hidrógeno/química , Sulfuro de Hidrógeno/farmacología , Sustancias Protectoras/farmacología , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/farmacocinética , Antineoplásicos/química , Antineoplásicos/farmacocinética , Descubrimiento de Drogas , Humanos , Sulfuro de Hidrógeno/farmacocinética , Sustancias Protectoras/química , Sustancias Protectoras/farmacocinética
18.
Inflammopharmacology ; 23(2-3): 91-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25711289

RESUMEN

INTRODUCTION: The non-steroid anti-inflammatory drugs (NSAIDs) are among the drugs that can commonly cause injury in the esophagus, such as non-reflux oesophagitis, with important clinical consequences. This injury may be 'silent' and therefore often overlooked. Recently, we established that hydrogen sulfide (H2S) is a critical mediator of esophageal mucosal protection and repair. The aim of the study was to determine the effect of naproxen, the most commonly used NSAIDs, on the oesophagus and oesophagogastric junction and its relation with suppression or stimulation of endogenous H2S synthesis during naproxen-induced oesophageal injury. METHODS: Rats were treated with vehicle (control) or naproxen, with or without being subjected to water immersion restricted stress (Takagi et al. Chem Pharm Bul 12:465-472, 1964). Subgroups of rats were pre-treated with an inhibitor of H2S synthesis cystathionine γ-lyase (CSE) or cystathionine ß-synthase (CBS), or with the Sodium sulphide (NaHS), which spontaneously generates H2S in solution. Damage of the oesophageal mucosa and oesophagogastric junction was estimated and scored using a histological damage index. RESULTS: Treatment with naproxen increased the thickness of the corneal and epithelial layers of the oesophagus, as well as producing disorganization of the muscle plate and irregular submucosal oedema. Both injury factors, stress and suppression of H2S synthesis resulted in the development of severe esophagitis and damage to the oesophagogastric junction. The damage was exacerbated by inhibitors of H2S biosynthesis, and attenuated by treatment with NaHS. CONCLUSIONS: Inhibition of endogenous H2S synthesis provides a novel experimental model that can be useful in preclinical studies NSAID-related non-reflux oesophagitis. H2S contributes significantly to mucosal defence in the oesophagus.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Unión Esofagogástrica/efectos de los fármacos , Unión Esofagogástrica/lesiones , Sulfuro de Hidrógeno/metabolismo , Animales , Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Modelos Animales de Enfermedad , Unión Esofagogástrica/metabolismo , Unión Esofagogástrica/fisiología , Naproxeno/efectos adversos , Ratas , Sulfuros/metabolismo
19.
J Neuroinflammation ; 11: 196, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25472548

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) induces secondary injury mechanisms, including dynamic interplay between ischemic, inflammatory and cytotoxic processes. We recently reported that administration of ATB-346 (2-(6-methoxynapthalen- 2-yl)-propionic acid 4-thiocarbamoyl-phenyl ester), a hydrogen sulfide-releasing cyclooxygenase inhibitor, showed marked beneficial effects in an animal model of spinal cord injury, significantly enhancing recovery of motor function and reducing the secondary inflammation and tissue injury. METHODS: Here we evaluated the neuroprotective potential of ATB-346, a hydrogen sulfide-releasing derivative of naproxen, using the controlled cortical impact (CCI) injury model in mice, one of the most common models of TBI. Moreover, the aim of the present study was to carefully investigate molecular pathways and subtypes of glial cells involved in the protective effect of ATB-346 on inflammatory reaction associated with an experimental model of TBI. In these studies, TBI was induced in mice by CCI and mice were orally administered ATB-346, naproxen (both at 30 µmol/kg) or vehicle (dimethylsulfoxide:1% carboxymethylcellulose [5:95] suspension) one and six hours after brain trauma and once daily for 10 days. RESULTS: Results revealed that ATB-346 attenuated TBI-induced brain edema, suppressed TBI-induced neural cell death and improved neurological function. ATB-346 also significantly reduced the severity of inflammation and restored neurotrophic factors that characterized the secondary events of TBI. CONCLUSIONS: These data demonstrate that ATB-346 can be efficacious in a TBI animal model by reducing the secondary inflammation and tissue injury. Therefore, ATB-346 could represent an interesting approach for the management of secondary damage following CNS diseases, counteracting behavioral changes and inflammatory process.


Asunto(s)
Lesiones Encefálicas/metabolismo , Corteza Cerebral/metabolismo , Inhibidores de la Ciclooxigenasa/uso terapéutico , Sulfuro de Hidrógeno/metabolismo , Trastornos de la Destreza Motora/metabolismo , Naproxeno/análogos & derivados , Animales , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/patología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Inhibidores de la Ciclooxigenasa/farmacología , Masculino , Ratones , Trastornos de la Destreza Motora/tratamiento farmacológico , Trastornos de la Destreza Motora/patología , Naproxeno/farmacología , Naproxeno/uso terapéutico , Tamaño de los Órganos , Distribución Aleatoria
20.
FASEB J ; 27(11): 4489-99, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23901068

RESUMEN

Spinal cord trauma causes loss of motor function that is in part due to the ensuing inflammatory response. Hydrogen sulfide (H2S) is a potent, endogenous anti-inflammatory and neuroprotective substance that has been explored for use in the design of novel nonsteroidal anti-inflammatory drugs. In the current study, we evaluated the potential beneficial effects of ATB-346 [2-(6-methoxynapthalen- 2-yl)-propionic acid 4-thiocarbamoyl-phenyl ester], an H2S-releasing derivative of naproxen, in a murine model of spinal cord injury (SCI). SCI was induced in mice by spinal cord compression, produced through the application of vascular clips to the dura via a T5 to T8 laminectomy. ATB-346, naproxen (both at 30 µmol/kg), or vehicle was orally administered to the mice 1 and 6 h after SCI and once daily thereafter for 10 d. Motor function [Basso Mouse Scale (BMS) of locomotion] improved gradually in the mice treated with naproxen. However, those treated with ATB-346 exhibited a significantly more rapid and sustained recovery of motor function, achieving greater than double the increase in locomotion score of the naproxen group by the 10th day of treatment. ATB-346 also significantly reduced the severity of inflammation (proinflammatory cytokines, apoptosis of neural tissue, and nitrosative stress) that characterized the secondary effects of SCI. Again, the effects of ATB-346 were superior to those of naproxen for several parameters. These results showed marked beneficial effects of an H2S-releasing derivative of naproxen in an animal model of SCI, significantly enhancing recovery of motor function, possibly by reducing the secondary inflammation and tissue injury that characterizes this model. The combination of inhibition of cyclooxygenase and delivery of H2S may offer a promising alternative to existing therapies for traumatic injury.


Asunto(s)
Inhibidores de la Ciclooxigenasa/uso terapéutico , Naproxeno/análogos & derivados , Recuperación de la Función , Traumatismos de la Médula Espinal/tratamiento farmacológico , Administración Oral , Animales , Apoptosis , Inhibidores de la Ciclooxigenasa/administración & dosificación , Inhibidores de la Ciclooxigenasa/farmacología , Citocinas/genética , Citocinas/metabolismo , Sulfuro de Hidrógeno/farmacología , Locomoción/efectos de los fármacos , Masculino , Ratones , Naproxeno/administración & dosificación , Naproxeno/farmacología , Naproxeno/uso terapéutico , Médula Espinal/metabolismo , Médula Espinal/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA