Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Pharmacol Res ; 81: 1-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24434421

RESUMEN

Heme oxygenase-1 (HO-1) is a redox sensitive inducible enzyme endowed with important antioxidant and cytoprotective activities. Here we report that two water-soluble isothiocyanate-cysteine conjugates, S-[N-benzyl(thiocarbamoyl)]-l-cysteine (BTTC) and S-[N-(3-phenylpropyl)(thiocarbamoyl)]-l-cysteine (PTTC), potently increase HO-1 protein expression and heme oxygenase activity in renal tubular epithelial cells at 5 and 10µM, while higher concentrations are themselves cytotoxic and pro-apoptotic. Inhibitors of the pro-survival pathways ERK, MAPK and PI3K almost completely abolished the increase in HO-1 induction and heme oxygenase activity, while the JNK pathway appeared to be mainly involved in the apoptosis triggered by the isothiocyanates. We also found that renal cells exposed to 50µM cisplatin (CDDP), a chemotherapeutic agent known for its nephrotoxic actions, displayed a marked increase in caspase-3 activity and the number of apoptotic cells. These effects were abolished by pre-incubation of cells with concentrations of BTCC or PTCC that maximize HO-1 induction and were reversed by the inhibitor of heme oxygenase activity tin protoporphyrin IX (SnPPIX). Moreover, in a model of CDDP-induced nephrotoxicity in vivo, pre-treatment of rats with a daily dose of BTCC or PTCC (25mg/kg, i.p.) completely abolished the increase in serum creatinine and urea levels and markedly reduced the severity of renal tissue apoptosis caused by CDDP. The renoprotective effects of BTCC and PTCC in vivo were markedly attenuated by administration of rats with SnPPIX. These findings indicate that water-soluble isothiocyanates counteract renal dysfunction and apoptosis by up-regulating the HO-1 system and could be used as a supplementary treatment to mitigate CDDP-induced nephrotoxic effects.


Asunto(s)
Cisteína/análogos & derivados , Riñón/efectos de los fármacos , Sustancias Protectoras/farmacología , Tiocarbamatos/farmacología , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Cisplatino , Creatinina/sangre , Cisteína/farmacología , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Riñón/metabolismo , Riñón/patología , Células LLC-PK1 , Masculino , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , ARN Mensajero/metabolismo , Ratas Wistar , Porcinos , Urea/sangre
2.
Pharmacol Res ; 68(1): 108-17, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23253427

RESUMEN

Carbon monoxide-releasing molecules (CO-RMs) are a class of organometallo carbonyl complexes capable of delivering controlled quantities of CO gas to cells and tissues thus exerting a broad spectrum of pharmacological effects. Here we report on the chemical synthesis, CO releasing properties, cytotoxicity profile and pharmacological activities of four novel structurally related iron-allyl carbonyls. The major difference among the new CO-RMs tested was that three compounds (CORM-307, CORM-308 and CORM-314) were soluble in dimethylsulfoxide (DMSO), whereas a fourth one (CORM-319) was rendered water-soluble by reacting the iron-carbonyl with hydrogen tetrafluoroborate. We found that despite the fact all compounds liberated CO, CO-RMs soluble in DMSO caused a more pronounced toxic effect both in vascular and inflammatory cells as well as in isolated vessels. More specifically, iron carbonyls soluble in DMSO released CO with a fast kinetic and displayed a marked cytotoxic effect in smooth muscle cells and RAW 247.6 macrophages despite exerting a rapid and pronounced vasorelaxation ex vivo. In contrast, CORM-319 that is soluble in water and liberated CO with a slower rate, preserved smooth muscle cell viability, relaxed aortic tissue and exerted a significant anti-inflammatory effect in macrophages challenged with endotoxin. These data suggest that iron carbonyls can be used as scaffolds for the design and synthesis of pharmacologically active CO-RMs and indicate that increasing water solubility and controlling the rate of CO release are important parameters for limiting their potential toxic effects.


Asunto(s)
Monóxido de Carbono/metabolismo , Óxido Nítrico/antagonistas & inhibidores , Compuestos Organometálicos/farmacología , Vasodilatadores/farmacología , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiología , Monóxido de Carbono/química , Línea Celular , Técnicas In Vitro , Hierro/química , Macrófagos , Masculino , Ratones , Compuestos Organometálicos/química , Ratas , Ratas Sprague-Dawley , Vasodilatación/efectos de los fármacos , Vasodilatadores/química
3.
Crit Care Med ; 40(2): 544-52, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21926571

RESUMEN

OBJECTIVE: Intracerebral hemorrhage is accompanied by a pronounced inflammatory response that mediates brain damage but is also essential for the tissue reparative process. We assessed the effect of CORM-3, a water-soluble carbon monoxide-releasing molecule possessing anti-inflammatory properties, on inflammation and brain injury after intracerebral hemorrhage. DESIGN: In vivo and in vitro laboratory study. SETTING: Research laboratory. SUBJECTS: Male Sprague-Dawley rats, 250-350 g. INTERVENTIONS: A model of collagenase injection (2 µL) in the brain was established to induce intracerebral hemorrhage. CORM-3 (4 or 8 mg/kg) was administered intravenously at different times as follows: 1) 5 mins before collagenase; 2) 3 hrs after collagenase; and 3) 3 days after collagenase challenge. MEASUREMENTS AND MAIN RESULTS: Saline was used as a negative control. Brain damage, brain water content, and behavioral assessment were evaluated. The inflammatory response was determined at set intervals after intracerebral hemorrhage by counting peripheral neutrophils and lymphocytes, neutrophils, and activated microglia/macrophages in the intracerebral hemorrhage area and measuring plasma tumor necrosis factor-á levels. BV2 microglia and DI-TNC1 astrocytes were exposed to triton (1%) or CORM-3 (10-100 ìM) and cytotoxicity (lactic dehydrogenase assay) measured at 24 hrs. A challenge with collagenase to induce intracerebral hemorrhage caused marked brain damage and modified the levels of inflammatory markers. Pretreatment with CORM-3 significantly prevented injury, modulated inflammation, and reduced plasma tumor necrosis factor-α. CORM-3 given 3 hrs after collagenase significantly increased brain injury and tumor necrosis factor-α production. In contrast, CORM-3 given 3 days after collagenase afforded partial protection, modulated inflammation, and decreased tumor necrosis factor-α starting from the day of application. No dose-dependent effects were observed. CONCLUSIONS: CORM-3 promotes neuroprotection or neurotoxicity after intracerebral hemorrhage depending on the time of administration. Beneficial effects are achieved when CORM-3 is given either before or 3 days after intracerebral hemorrhage, namely, as a prophylactic agent or during the postacute inflammatory phase.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Hemorragia Cerebral/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Compuestos Organometálicos/farmacología , Animales , Lesiones Encefálicas/etiología , Lesiones Encefálicas/mortalidad , Lesiones Encefálicas/patología , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/mortalidad , Hemorragia Cerebral/patología , Modelos Animales de Enfermedad , Inmunohistoquímica , Inflamación/tratamiento farmacológico , Inflamación/etiología , Mediadores de Inflamación/análisis , Mediadores de Inflamación/metabolismo , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Valores de Referencia , Medición de Riesgo , Índice de Severidad de la Enfermedad , Tasa de Supervivencia , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 30(2): 305-12, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19965783

RESUMEN

OBJECTIVE: Sickle cell disease (SCD) is characterized by extensive hemolysis, increased cellular adhesion, and vaso-occlusion. Tissues from sickle patients express heme oxygenase-1 (HO-1), the enzyme that degrades free heme/hemoglobin to the signaling molecule carbon monoxide, and the antioxidants biliverdin/bilirubin. Here, we examined the HO response in endothelial cells exposed to human sickle blood and determined whether this response is beneficial for SCD. METHODS AND RESULTS: We measured HO activity in human and bovine aortic endothelial cells incubated with human sickle or normal blood. Sickle blood increased HO activity, which was enhanced by hypoxia and was caused mainly by the red cell components of sickle blood. Oxidized hemoglobin was higher in sickle blood and increased markedly over time. Interestingly, HO activity correlated inversely with patients' hemoglobin levels and positively with bilirubin and lactate dehydrogenase. HO-1 induction, exogenous biliverdin, or carbon monoxide markedly decreased adhesion of sickle blood to the endothelium, and sickle red cells partially inhibited relaxation mediated by carbon monoxide in isolated aortas. CONCLUSIONS: Our results highlight important associations between SCD and HO byproducts, which may counteract vascular complications of SCD.


Asunto(s)
Anemia de Células Falciformes/sangre , Adhesión Celular , Células Endoteliales/enzimología , Eritrocitos/metabolismo , Hemo-Oxigenasa 1/metabolismo , Hemólisis , Adulto , Anemia de Células Falciformes/enzimología , Anemia de Células Falciformes/fisiopatología , Anemia de Células Falciformes/terapia , Animales , Bilirrubina/sangre , Biliverdina/metabolismo , Boranos/metabolismo , Boranos/farmacología , Dióxido de Carbono/metabolismo , Carbonatos/metabolismo , Carbonatos/farmacología , Estudios de Casos y Controles , Bovinos , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Femenino , Hemoglobinas/metabolismo , Humanos , L-Lactato Deshidrogenasa/sangre , Masculino , Persona de Mediana Edad , Compuestos Organometálicos/metabolismo , Compuestos Organometálicos/farmacología , Ratas , Estudios Retrospectivos , Factores de Tiempo , Vasodilatación , Adulto Joven
5.
J Cardiovasc Pharmacol ; 55(2): 168-75, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19935076

RESUMEN

Carbon monoxide (CO) liberated by a water-soluble carbon monoxide-releasing molecule (CORM-3) induces a positive inotropic effect with a negative chronotropic effect in normal rat hearts. However, the efficacy of CORM-3 under conditions of chronic cardiac insufficiency is unknown. In a rat model of doxorubicin-induced cardiomyopathy, CORM-3 (20 microg/min) produced a positive inotropic effect as demonstrated by significant increases in systolic pressure (P < 0.05) and pressure derivative (dp/dt max) over time (P < 0.05). A similar dose of CO-depleted negative control (inactive CORM-3) failed to cause any change in these parameters. When the inotrope dobutamine was added at a dose of 10 microM following CORM-3, there was no additional increase in systolic pressure or dp/dt max. However, significant rises in systolic pressure and dp/dt max were observed after dobutamine administration to the hearts previously treated with inactive CORM-3. These results suggest that CORM-3 produces a positive inotropic effect in doxorubicin cardiomyopathy rat hearts, similar to that reported previously in normal hearts. The inotropic effect produced by CO in the doxorubicin cardiomyopathy heart was mimicked by a classical inotrope (dobutamine), suggesting that either a maximal inotropic effect is achieved at this dose of CORM-3 or both drugs utilize shared signaling pathways in cardiac muscle.


Asunto(s)
Monóxido de Carbono/uso terapéutico , Cardiomiopatías/inducido químicamente , Cardiomiopatías/tratamiento farmacológico , Cardiotónicos/uso terapéutico , Doxorrubicina/toxicidad , Compuestos Organometálicos/uso terapéutico , Animales , Monóxido de Carbono/farmacocinética , Cardiomiopatías/metabolismo , Cardiotónicos/farmacocinética , Cardiotoxinas/toxicidad , Técnicas In Vitro , Masculino , Compuestos Organometálicos/farmacocinética , Ratas , Ratas Endogámicas Lew
6.
Int J Neurosci ; 120(12): 765-73, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20942593

RESUMEN

In this study, we investigated the inflammatory response to hemorrhagic stroke (HS) as the main mechanism of brain functional recovery. Sprague-Dawley rats (n = 24) underwent surgery with sterile saline (control group, n = 12) and collagenase IV-S (stroke group, n = 12) being injected into the right striatum. White blood cell analysis, histological and immunohistological examination of the brain slices, as well as densitometric analysis of polymorphonuclear and microglial cells/macrophages were correlated with behavioral tests, and the data were subjected to appropriate statistical processing. The results indicate a strong correlation between polymorphonuclear and mononuclear changes in the blood and the zone of hemorrhagic stroke with behavioral tests of functional brain recovery. We propose that the inflammatory response is determined by kinetics of polymorphonuclear and mononuclear cells in both the blood and the hemorrhagic stroke zone. Kinetics of these cells is followed by the restoration of functions, and the maximum functional recovery is observed by the time polymorphonuclear and mononuclear stages have completed. With the development of inflammation and leukocyte kinetics, it is possible to predict functional recovery of hemorrhagic stroke. Improvement of the degree and rate of hemorrhagic stroke functional recovery may be achieved by therapeutic interventions into the inflammatory mechanisms influencing polymorphonuclear and mononuclear cell kinetics.


Asunto(s)
Hemorragia Cerebral/patología , Leucocitos/patología , Accidente Cerebrovascular/patología , Animales , Conducta Animal/fisiología , Encéfalo/metabolismo , Encéfalo/patología , Movimiento Celular/fisiología , Hemorragia Cerebral/complicaciones , Modelos Animales de Enfermedad , Inflamación/etiología , Inflamación/patología , Recuento de Leucocitos/métodos , Leucocitos/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Microglía/metabolismo , Microglía/patología , Examen Neurológico , Neutrófilos/metabolismo , Neutrófilos/patología , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Accidente Cerebrovascular/complicaciones
7.
J Neurosci Res ; 86(9): 1927-35, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18381758

RESUMEN

The inducible protein heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to carbon monoxide (CO) and biliverdin, which play a concerted action in cytoprotection against oxidative stress and in the modulation of cell proliferation and differentiation. Here we report that both HO-1 expression and activity can be highly increased in undifferentiated human mesenchymal stem cells (MSCs) treated with hemin, a known HO-1 inducer. However, HO-1 mRNA and protein expression gradually decrease when MSCs undergo neural differentiation in vitro, making them extremely susceptible to glutamate-mediated cytotoxicity. A time course for HO-1 revealed that this protein is markedly down-regulated after 2 days and returns to control levels 6 days after differentiation. Treatment with glutamate (250 microM) after 2 days of neural differentiation resulted in a more pronounced lactate dehydrogenase release, a marker of cell injury, compared with undifferentiated cells. Notably, cells pretreated with hemin (50 microM) or compounds that release small amounts of CO (10 microM CORM-3 and CORM-A1) rendered cells more resistant to glutamate-induced toxicity; this effect was evident in both undifferentiated and differentiated MSCs. Our findings indicate that MSCs become more vulnerable to oxidative injury during the early stages of differentiation via mechanisms that involve a temporary inhibition of HO-1 expression. Thus, overexpression of HO-1 and CO-releasing molecules could provide a possible therapeutic strategy to improve cell viability during neural differentiation in applications that use stem cell technology.


Asunto(s)
Monóxido de Carbono/metabolismo , Hemo-Oxigenasa 1/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/enzimología , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Diferenciación Celular , Medios de Cultivo , Citoprotección/fisiología , Cartilla de ADN , Hemo-Oxigenasa 1/genética , Humanos , L-Lactato Deshidrogenasa/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Chem Res Toxicol ; 21(7): 1484-94, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18547064

RESUMEN

Chalcones are naturally occurring flavonoids composed of two aromatic rings connected by a three-carbon unit forming an alpha-beta unsaturated carbonyl group. They are pharmacologically relevant because of their ability to exert anticarcinogenic, antimicrobial, and anti-inflammatory activities. Recent evidence indicates that the bioactivity of hydroxy-chalcones is correlated with their intrinsic property to induce the antioxidant and cytoprotective enzyme heme oxygenase-1 (HO-1). In the present study, we assessed how the methoxy substituents positioned on the two aromatic rings affect the anti-inflammatory action of different chalcones in relation to their ability to increase heme oxygenase in RAW246.7 macrophages. Structure-activity relationships of methoxychlacones were qualitatively and quantitatively examined and correlated with inhibition of endotoxin-mediated nitrite production and cytotoxic effects. Our data indicate that (i) a progressive increase in heme oxygenase activity is obtained by sequentially increasing the number of methoxy substituents in the 3,4,5- and 3',4',5'-positions of the aromatic rings; (ii) methoxy substituents placed either in the 2,4,6-positions or alone in the 4- or 4'-position are ineffective; (iii) increased heme oxygenase activity by chalcones is lost when the alpha-beta double bond and the carbonyl group are reduced or protected; (iv) the anti-inflammatory activity and cytotoxicity profiles of the chalcones examined correlate with their potency as HO-1 inducers; and (v) chalcone-mediated HO-1 induction is reduced by thiols or inhibitors of phosphatidylinositol-3 kinase (PI3K) pathway. This study provides additional information on the structural features that methoxychalcones and natural antioxidants need to possess to be considered as therapeutic agents for maximizing HO-1 expression and activity.


Asunto(s)
Antioxidantes/farmacología , Chalconas/farmacología , Hemo-Oxigenasa 1/biosíntesis , Macrófagos/enzimología , Animales , Antioxidantes/química , Biomarcadores/química , Biomarcadores/metabolismo , Chalconas/química , Inducción Enzimática , Hemo-Oxigenasa 1/química , Macrófagos/efectos de los fármacos , Ratones , Relación Estructura-Actividad
9.
Exp Mol Med ; 39(2): 139-48, 2007 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-17464175

RESUMEN

Curcumin is a polyphenolic compound possessing interesting anti-inflammatory and antioxidant properties and has the ability to induce the defensive protein heme oxygenase-1 (HO-1). The objective of this study was to investigate whether curcumin protects against cold storage-mediated damage of human adult atrial myoblast cells (Girardi cells) and to assess the potential involvement of HO-1 in this process. Girardi cells were exposed to either normothermic or hypothermic conditions in Celsior preservation solution in the presence or absence of curcumin. HO-1 protein expression and heme oxygenase activity as well as cellular damage were assessed after cold storage or cold storage followed by re-warming. In additional experiments, an inhibitor of heme oxygenase activity (tin protoporphyrin IX, 10 microM) or siRNA for HO-1 were used to investigate the participation of HO-1 as a mediator of curcumin-induced effects. Treatment with curcumin produced a marked induction of cardiac HO-1 in normothermic condition but cells were less responsive to the polyphenolic compound at low temperature. Cold storage-induced damage was markedly reduced in the presence of curcumin and HO-1 contributed to some extent to this effect. Thus, curcumin added to Celsior preservation solution effectively prevents the damage caused by cold-storage; this effect involves the protective enzyme HO-1 but also other not yet identified mechanisms.


Asunto(s)
Criopreservación , Curcumina/farmacología , Mioblastos Cardíacos/efectos de los fármacos , Mioblastos Cardíacos/patología , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Frío , Crioprotectores/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Hemina/farmacología , Humanos , Peróxido de Hidrógeno/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo
10.
FASEB J ; 19(2): 284-6, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15556971

RESUMEN

Carbon monoxide (CO) is emerging as an important and versatile mediator of physiological processes to the extent that treatment of animals with exogenous CO gas has beneficial effects in a range of vascular- and inflammatory-related disease models. The recent discovery that certain transition metal carbonyls function as CO-releasing molecules (CO-RMs) in biological systems highlighted the potential of exploiting this and similar classes of compounds as a stratagem to deliver CO for therapeutic purposes. Here we describe the biochemical features and pharmacological actions of a newly identified water-soluble CO releaser (CORM-A1) that, unlike the first prototypic molecule recently described (CORM-3), does not contain a transition metal and liberates CO at a much slower rate under physiological conditions. Using a myoglobin assay and an amperometric CO electrode, we demonstrated that the release of CO from CORM-A1 is both pH- and temperature-dependent with a half-life of approximately 21 min at 37 degrees C and pH 7.4. In isolated aortic rings, CORM-A1 promoted a gradual but profound concentration-dependent vasorelaxation over time, which was highly amplified by YC-1 (1 microM) and attenuated by ODQ, a stimulator and inhibitor of guanylate cyclase, respectively. Similarly, administration of CORM-A1 (30 micromol/kg i.v.) in vivo produced a mild decrease in mean arterial pressure, which was markedly potentiated by pretreatment with YC-1 (1.2 micromol/kg i.v.). Interestingly, an inactive form of CORM-A1 that is incapable of releasing CO failed to promote both vasorelaxation and hypotension, thus directly implicating CO as the mediator of the observed pharmacological effects. Our results reveal that the bioactivities exerted by CORM-A1 reflect its intrinsic biochemical behavior of a slow CO releaser, which may be advantageous in the treatment of chronic conditions that require CO to be delivered in a carefully controlled manner.


Asunto(s)
Boranos/farmacología , Monóxido de Carbono/metabolismo , Carbonatos/farmacología , Animales , Aorta/química , Aorta/metabolismo , Sangre/metabolismo , Boranos/síntesis química , Boranos/química , Carbonatos/síntesis química , Carbonatos/química , Carboxihemoglobina/química , Humanos , Concentración de Iones de Hidrógeno , Indazoles/metabolismo , Masculino , Compuestos Organometálicos , Preparaciones Farmacéuticas/síntesis química , Preparaciones Farmacéuticas/química , Ratas , Ratas Endogámicas Lew , Ratas Sprague-Dawley , Temperatura , Vasodilatadores/síntesis química , Vasodilatadores/química , Vasodilatadores/farmacología
11.
Circ Res ; 90(2): E17-24, 2002 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-11834719

RESUMEN

Carbon monoxide (CO) is generated in living organisms during the degradation of heme by the enzyme heme oxygenase, which exists in constitutive (HO-2 and HO-3) and inducible (HO-1) isoforms. Carbon monoxide gas is known to dilate blood vessels in a manner similar to nitric oxide and has been recently shown to possess antiinflammatory and antiapoptotic properties. We report that a series of transition metal carbonyls, termed here carbon monoxide-releasing molecules (CO-RMs), liberate CO to elicit direct biological activities. Specifically, spectrophotometric and NMR analysis revealed that dimanganese decacarbonyl and tricarbonyldichlororuthenium (II) dimer release CO in a concentration-dependent manner. Moreover, CO-RMs caused sustained vasodilation in precontracted rat aortic rings, attenuated coronary vasoconstriction in hearts ex vivo, and significantly reduced acute hypertension in vivo. These vascular effects were mimicked by induction of HO-1 after treatment of animals with hemin, which increases endogenously generated CO. Thus, we have identified a novel class of compounds that are useful as prototypes for studying the bioactivity of CO. In the long term, transition metal carbonyls could be utilized for the therapeutic delivery of CO to alleviate vascular- and immuno-related dysfunctions. The full text of this article is available at http://www.circresaha.org.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Corazón/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Compuestos Organometálicos/farmacología , Sistema Vasomotor/efectos de los fármacos , Animales , Presión Sanguínea/fisiología , Monóxido de Carbono/química , Monóxido de Carbono/metabolismo , Monóxido de Carbono/farmacología , Bovinos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Corazón/fisiología , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemina/farmacología , Técnicas In Vitro , Compuestos de Hierro Carbonilo , Sustancias Macromoleculares , Espectroscopía de Resonancia Magnética , Masculino , Metaloporfirinas/farmacología , Músculo Liso Vascular/metabolismo , Compuestos Organometálicos/química , Compuestos Organometálicos/metabolismo , Protoporfirinas/farmacología , Ratas , Elementos de Transición/química , Vasodilatadores/química , Vasodilatadores/metabolismo , Vasodilatadores/farmacología , Sistema Vasomotor/metabolismo
12.
Circ Res ; 93(2): e2-8, 2003 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-12842916

RESUMEN

Carbon monoxide, which is generated in mammals during the degradation of heme by the enzyme heme oxygenase, is an important signaling mediator. Transition metal carbonyls have been recently shown to function as carbon monoxide-releasing molecules (CO-RMs) and to elicit distinct pharmacological activities in biological systems. In the present study, we report that a water-soluble form of CO-RM promotes cardioprotection in vitro and in vivo. Specifically, we found that tricarbonylchloro(glycinato)ruthenium(II) (CORM-3) is stable in water at acidic pH but in physiological buffers rapidly liberates CO in solution. Cardiac cells pretreated with CORM-3 (10 to 50 micromol/L) become more resistant to the damage caused by hypoxia-reoxygenation and oxidative stress. In addition, isolated hearts reperfused in the presence of CORM-3 (10 micromol/L) after an ischemic event displayed a significant recovery in myocardial performance and a marked and significant reduction in cardiac muscle damage and infarct size. The cardioprotective effects mediated by CORM-3 in cardiac cells and isolated hearts were totally abolished by 5-hydroxydecanoic acid, an inhibitor of mitochondrial ATP-dependent potassium channels. Predictably, cardioprotection is lost when CORM-3 is replaced by an inactive form (iCORM-3) that is incapable of liberating CO. Using a model of cardiac allograft rejection in mice, we also found that treatment of recipients with CORM-3 but not iCORM-3 considerably prolonged the survival rate of transplanted hearts. These data corroborate the notion that transition metal carbonyls could be used as carriers to deliver CO and highlight the bioactivity and potential therapeutic features of CO-RMs in the mitigation of cardiac dysfunction. The full text of this article is available online at http://www.circresaha.org.


Asunto(s)
Monóxido de Carbono/farmacología , Cardiotónicos/farmacología , Rechazo de Injerto/tratamiento farmacológico , Compuestos Organometálicos/farmacología , Daño por Reperfusión/prevención & control , Compuestos de Rutenio/farmacología , Animales , Monóxido de Carbono/química , Monóxido de Carbono/metabolismo , Células Cultivadas , Ácidos Decanoicos/farmacología , Modelos Animales de Enfermedad , Supervivencia de Injerto/efectos de los fármacos , Trasplante de Corazón/efectos adversos , Concentración de Iones de Hidrógeno , Hidroxiácidos/farmacología , Técnicas In Vitro , Ratones , Isquemia Miocárdica/tratamiento farmacológico , Compuestos Organometálicos/química , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Endogámicas Lew , Compuestos de Rutenio/química , Compuestos de Rutenio/metabolismo , Solubilidad , Agua/química
13.
Int Immunopharmacol ; 6(13-14): 1993-2001, 2006 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-17161353

RESUMEN

In an ex vivo mouse model, regulatory transplantation tolerance is not only linked to Foxp3, but also to release of leukaemia inhibitory factor (LIF) and to expression of axotrophin (also known as MARCH-7), a putative ubiquitin E3 ligase associated with feedback control of T cell activation and of T cell-derived LIF. Given this coordinate correlation with tolerance, we now ask if Foxp3 expression is influenced by LIF or by axotrophin. In spleen cells from allo-rejected mice we found that exogenous LIF reduced interferon gamma release in response to donor antigen by 50%, but LIF had no direct effect on levels of Foxp3 protein in allo-primed cells that were either tolerant, or aggressive, for donor antigen. However, we did find an effect of axotrophin on Foxp3: in the axotrophin null mouse, thymic Foxp3 transcripts were reduced compared to axotrophin wildtype littermates. To test whether these findings in the mouse were of potential significance in man we measured transcript levels of axotrophin and LIF in peripheral blood cell samples collected for a recently published clinical study concerning haematopoietic stem cell recipients. In controls, human peripheral blood CD4+CD25+cells contained significantly more FOXP3 and axotrophin than CD4+CD25-cells. In bone marrow autograft recipients, where peripheral blood cell samples directly represent both the grafted tissue and the immune response, both FOXP3 and axotrophin negatively correlated with graft versus host disease (GVHD). These data suggest that (i) thymic Foxp3+T cell development is influenced by axotrophin; and (ii) clinical auto-GVHD inversely correlates with axotrophin transcript expression as has been previously reported for FOXP3.


Asunto(s)
Factores de Transcripción Forkhead/genética , Factor Inhibidor de Leucemia/genética , Tolerancia al Trasplante/inmunología , Ubiquitina-Proteína Ligasas/genética , Animales , Trasplante de Médula Ósea/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Femenino , Factores de Transcripción Forkhead/metabolismo , Expresión Génica/inmunología , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/metabolismo , Humanos , Interferón gamma/metabolismo , Subunidad alfa del Receptor de Interleucina-2/inmunología , Interleucinas/metabolismo , Factor Inhibidor de Leucemia/metabolismo , Factor Inhibidor de Leucemia/farmacología , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Linfocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos CBA , Ratones Noqueados , Factor de Transcripción STAT3/metabolismo , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Timo/inmunología , Timo/metabolismo , Inmunología del Trasplante/genética , Inmunología del Trasplante/inmunología , Tolerancia al Trasplante/genética , Ubiquitina-Proteína Ligasas/metabolismo
14.
Pharmacol Rep ; 58 Suppl: 132-44, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17332683

RESUMEN

The development of carbon monoxide-releasing molecules (CO-RMs) in recent years helped to shed more light on the diverse range of anti-inflammatory and cytoprotective activities of CO gas. In this study, we examined the effect of a ruthenium-based water-soluble CO carrier (CORM-3) on lipopolysaccharide (LPS)- and interferon-gamma (INF-gamma)-induced inflammatory responses in BV-2 microglial cells and explored the possible mechanisms of action. BV-2 microglial cells were stimulated with either LPS or INF-gamma in the presence of CORM-3 and the inflammatory response evaluated by assessing the effect on nitric oxide production (nitrite levels) and tumor necrosis factor-alpha (TNF-alpha) release. Similar experiments were also performed in the presence of inhibitors of guanylate cyclase (ODQ), NO synthase (L-NAME), heme oxygenase activity (tin protoporphyrin IX) or various mitogen-activated protein kinase (MAPK) inhibitors. CORM-3 significantly attenuated the inflammatory response to LPS and INF-gamma as evidenced by a significant reduction (p < 0.001) in nitrite levels and TNF-alpha production (P < 0.05). Such effect was maintained in the presence of ODQ, L-NAME or tin protoporphyrin without showing any cytotoxicity. The use of an inactive form of CORM-3 that does not contain carbonyl groups (Ru(DMSO)(4)Cl(2) failed to inhibit the increase in inflammatory markers suggesting that liberated CO mediates the observed effects. In addition, inhibition of phosphatidylinositol-3-phosphate kinase (PI3K) and extracellular signal-regulated kinase (ERK) pathways seemed to amplify the anti-inflammatory effect of CORM-3, particularly in cells stimulated with INF-gamma. These results suggest that the anti-inflammatory action of CORM-3 could be exploited to mitigate microglia activation in neuro-inflammatory diseases.


Asunto(s)
Monóxido de Carbono/fisiología , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Microglía/metabolismo , Compuestos Organometálicos/farmacología , Animales , Monóxido de Carbono/metabolismo , Línea Celular , Supervivencia Celular , Activación Enzimática , Guanilato Ciclasa/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Inflamación/metabolismo , Microglía/citología , Microglía/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Óxido Nítrico Sintasa/antagonistas & inhibidores , Nitritos/metabolismo , Compuestos Organometálicos/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Factor de Necrosis Tumoral alfa/metabolismo
15.
Transplantation ; 79(6): 726-30, 2005 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-15785381

RESUMEN

BACKGROUND: The specific regulation of allo-tolerance in vivo occurs within a complex microenvironment and involves co-operation between a small proportion of different cell types within the spleen or draining lymph node. By analyzing unmanipulated whole spleen cell populations we have aimed to mimic this in vivo situation to identify critical signaling molecules in regulatory allo-tolerance. METHODS: We compared the kinetics of cytokine release and induction of signaling proteins in (BALB/c-tolerant)CBA, versus (BALB/c-rejected)CBA, spleen cells after challenge with BALB/c antigen. RESULTS: The distinguishing features of allo-tolerance were Foxp3 protein expression, LIF release, and increased levels of STAT3. Comparison of isogenic clones of Tr1, Th1, and Th2 cells revealed that only the regulatory Tr1 cells are characterized by both LIF and IL10 release. CONCLUSIONS: Overall, our findings demonstrate that allo-antigen driven signaling events can be detected within a whole spleen cell population and identify a role for LIF in the regulation of transplantation tolerance in vivo.


Asunto(s)
Proteínas/metabolismo , Tolerancia al Trasplante/inmunología , Animales , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción Forkhead , Interleucina-6 , Factor Inhibidor de Leucemia , Ratones , Ratones Endogámicos BALB C , Factor de Transcripción STAT3 , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transactivadores/metabolismo
16.
Br J Pharmacol ; 145(6): 800-10, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15880142

RESUMEN

The enzyme heme oxygenase-1 (HO-1) is a cytoprotective and anti-inflammatory protein that degrades heme to produce biliverdin/bilirubin, ferrous iron and carbon monoxide (CO). The anti-inflammatory properties of HO-1 are related to inhibition of adhesion molecule expression and reduction of oxidative stress, while exogenous CO gas treatment decreases the production of inflammatory mediators such as cytokines and nitric oxide (NO). CO-releasing molecules (CO-RMs) are a novel group of substances identified by our group that are capable of modulating physiological functions via the liberation of CO. We aimed in this study to examine the potential anti-inflammatory characteristics of CORM-2 and CORM-3 in an in vitro model of lipopolysaccharide (LPS)-stimulated murine macrophages. Stimulation of RAW264.7 macrophages with LPS resulted in increased expression of inducible NO synthase (iNOS) and production of nitrite. CORM-2 or CORM-3 (10-100 microM) reduced nitrite generation in a concentration-dependent manner but did not affect the protein levels of iNOS. CORM-3 also decreased nitrite levels when added 3 or 6 h after LPS exposure. CORM-2 or CORM-3 did not cause any evident cytotoxicity and produced an increase in HO-1 expression and heme oxygenase activity; this effect was completely prevented by the thiol donor N-acetylcysteine. CORM-3 also considerably reduced the levels of tumor necrosis factor-alpha, another mediator of the inflammatory response. The inhibitory effects of CORM-2 and CORM-3 were not observed when the inactive compounds, which do not release CO, were coincubated with LPS. These results indicate that CO liberated by CORM-2 and CORM-3 significantly suppresses the inflammatory response elicited by LPS in cultured macrophages and suggest that CO carriers can be used as an effective strategy to modulate inflammation.


Asunto(s)
Inflamación/prevención & control , Macrófagos/efectos de los fármacos , Nitritos/metabolismo , Compuestos Organometálicos/farmacología , Animales , Monóxido de Carbono , Línea Celular , Supervivencia Celular , Glutatión/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1 , Inflamación/inducido químicamente , Lipopolisacáridos , Macrófagos/metabolismo , Proteínas de la Membrana , Ratones , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Factor de Necrosis Tumoral alfa/metabolismo
17.
FEBS Lett ; 543(1-3): 113-9, 2003 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-12753916

RESUMEN

Bilirubin (BR) and biliverdin (BV), two metabolites produced during haem degradation by haem oxygenase, possess strong antioxidant activities toward peroxyl radical, hydroxyl radical and hydrogen peroxide. Considering the importance attributed to nitric oxide (NO) and its congeners in the control of physiological and pathophysiological processes, we examined the interaction of BR and BV with NO and NO-related species in vitro. Exposure of BR and BV to agents that release NO or nitroxyl resulted in a concentration- and time-dependent loss of BR and BV, as assessed by high performance liquid chromatography. Peroxynitrite, a strong oxidant derived from the reaction of NO with superoxide anion, also showed high reactivity toward BR and BV. The extent of BR and BV consumption largely depended on the NO species being analysed and on the half-lives of the pharmacological compounds considered. Of major importance, BR and BV decomposition occurred also in the presence of pure NO under anaerobic conditions, confirming the ability of bile pigments to scavenge the gaseous free radical. Increasing concentrations of thiols prevented BR consumption by nitroxyl, indicating that bile pigments and thiol groups can compete and/or synergise the cellular defence against NO-related species. In view of the high inducibility of haem oxygenase-1 by NO-releasing agents in different cell types, the present findings highlight novel anti-nitrosative characteristics of BR and BV suggesting a potential function for bile pigments against the damaging effects of uncontrolled NO production.


Asunto(s)
Bilirrubina/metabolismo , Biliverdina/metabolismo , Depuradores de Radicales Libres/metabolismo , Especies de Nitrógeno Reactivo/metabolismo , Anaerobiosis , Cinética , Óxido Nítrico/metabolismo , Nitritos/metabolismo , Óxidos de Nitrógeno/metabolismo , Ácido Peroxinitroso/metabolismo , Compuestos de Sulfhidrilo/farmacología
18.
Antioxid Redox Signal ; 4(4): 615-24, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12230873

RESUMEN

Heme oxygenase-1 (HO-1) is an inducible stress protein the expression of which can be markedly augmented in eukaryotes by a wide range of substances that cause a transient change in the cellular redox state. The importance of this protein in physiology and disease is underlined by the versatility of HO-1 inducers and the functional role attributed to HO-1 products (carbon monoxide and bilirubin) in conditions that are associated with moderate or severe cellular stress. An intriguing aspect is the recent evidence showing that nitric oxide, a ubiquitous signaling molecule, finely modulates the activation of HO-1 expression. As the effects of oxidative stress on the regulation of the HO-1 gene have been well established and characterized, this review will focus on the biological relevance of redox signals involving nitric oxide and reactive nitrogen species that lead to up-regulation of the HO-1 pathway, with particular emphasis on vascular tissues and the cardiovascular system.


Asunto(s)
Hemo Oxigenasa (Desciclizante)/metabolismo , Óxido Nítrico/metabolismo , Estrés Oxidativo , Transducción de Señal/fisiología , Animales , Inducción Enzimática , Glutatión/metabolismo , Hemo Oxigenasa (Desciclizante)/genética , Hemo-Oxigenasa 1 , Humanos , Proteínas de la Membrana , Oxidación-Reducción , Especies de Nitrógeno Reactivo/metabolismo
19.
Antioxid Redox Signal ; 4(2): 229-40, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12006174

RESUMEN

Hypoxia, cytokines, and nitric oxide (NO) stimulate the generation of vascular endothelial growth factor (VEGF) and induce heme oxygenase-1 (HO-1) expression in vascular tissue. HO-1 degrades heme to carbon monoxide (CO), iron, and biliverdin, the latter being reduced to bilirubin by biliverdin reductase. In the present study, we investigated the role of HO-1 in the modulation of VEGF synthesis in rat vascular smooth muscle cells (VSMC). In VSMC stimulated with cytokines, inhibition of NO production significantly, but not completely, reduced VEGF release. In contrast, inhibition of HO activity by tin protoporphyrin IX (SnPPIX) totally prevented cytokine-induced increase in VEGF, despite an augmented synthesis of intracellular NO. Stimulation of HO-1 activity by hemin enhanced VEGF production; this effect was abrogated by blockade of the HO pathway. Similarly, VEGF synthesis induced by hypoxia was down-regulated by SnPPIX, but not by inhibitors of NO synthase. To elucidate further a direct involvement of HO-1 in the observed effects, we generated transfected cells that overexpressed the HO-1 gene. Notably, these cells synthesized significantly more VEGF protein than cells transfected with a control gene. Among the products of HO-1, biliverdin and bilirubin showed no effect, whereas iron ions inhibited VEGF synthesis. Exposure of cells to 1% CO resulted in a marked accumulation of VEGF (20-fold increase) over the basal level. Our data indicate that HO-1 activity influences the generation of VEGF in VSMC in both normoxic and hypoxic conditions. As CO and iron, respectively the inducer and the inhibitor of VEGF synthesis, are concomitantly produced during the degradation of heme, these data indicate that HO by-products may differentially modulate VEGF production.


Asunto(s)
Factores de Crecimiento Endotelial/genética , Hemo Oxigenasa (Desciclizante)/metabolismo , Linfocinas/genética , Músculo Liso Vascular/enzimología , Animales , Bilirrubina/metabolismo , Biliverdina/metabolismo , Células Cultivadas , Deferoxamina/farmacología , Inhibidores Enzimáticos/farmacología , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemo Oxigenasa (Desciclizante)/genética , Hemo-Oxigenasa 1 , Hemina/farmacología , Interleucina-1/farmacología , Hierro/farmacología , Metaloporfirinas/farmacología , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Protoporfirinas/farmacología , Ratas , Proteínas Recombinantes/metabolismo , Transcripción Genética/efectos de los fármacos , Transfección , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
20.
Biochem Soc Symp ; (71): 177-92, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15777021

RESUMEN

The family of haem oxygenase enzymes is unique in nature for its role in haem degradation. Haem is cleaved at the alpha-meso position by haem oxygenase with the support of electrons donated by cytochrome P450 reductase, the first products of this reaction being CO, iron and biliverdin. Biliverdin is then converted to bilirubin by biliverdin reductase. If haem is viewed as a substrate for an anabolic pathway, it becomes evident that haem oxygenases do not break down haem for elimination from the body, but rather use haem to generate crucial molecules that can modulate cellular functions. The facts that biliverdin and bilirubin are potent antioxidants and that CO is both a vasoactive and signalling molecule sustain this idea. The existence of a constitutive haem oxygenase (HO-2), mainly present in the vasculature and nervous system, and an inducible haem oxygenase (HO-1), which is highly expressed during stress conditions in all tissues, also suggests that cells have evolved a fine control of this enzymic pathway to ultimately regulate haem consumption and to ensure production of CO, biliverdin/bilirubin and iron during physiological and pathophysiological situations. This review will focus primarily on the biological actions of biliverdin and bilirubin derived from the haem oxygenase/biliverdin reductase systems and their potential roles in counteracting oxidative and nitrosative stress.


Asunto(s)
Pigmentos Biliares/metabolismo , Citoprotección/fisiología , Hemo Oxigenasa (Desciclizante)/metabolismo , Estrés Oxidativo/fisiología , Animales , Cardiopatías/fisiopatología , Humanos , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA