Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biomed Opt ; 15(1): 017001, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20210475

RESUMEN

Our purpose is to compare cobinamide to hydroxocobalamin in reversing cyanide (CN)-induced physiologic effects in an animal model using diffuse optical spectroscopy (DOS). Cyanide poisoning is a major threat worldwide. Cobinamide is a novel molecule that can bind two molecules of cyanide, has a much higher binding affinity than hydroxocobalamin, and is more water soluble. We investigated the ability of equimolar doses of cobinamide and hydroxocobalamin to reverse the effects of cyanide exposure in an animal model monitored continuously by DOS. Cyanide toxicity was induced in 16 New Zealand white rabbits by intravenous infusion. Animals were divided into three groups: controls (n=5) received saline following cyanide, hydroxocobalamin (N=6) following cyanide, and cobinamide (N=5) following cyanide. Cobinamide caused significantly faster and more complete recovery of oxy- and deoxyhemoglobin concentrations in cyanide-exposed animals than hydroxocobalamin- or saline-treated animals, with a recovery time constant of 13.8+/-7.1 min compared to 75.4+/-25.1 and 76.4+/-42.7 min, for hydroxocobalamin- and saline-treated animals, respectively (p<0.0001). This study indicates that cobinamide more rapidly and completely reverses the physiologic effects of cyanide than equimolar doses of cobalamin at the dose used in this study, and CN effects and response can be followed noninvasively using DOS.


Asunto(s)
Antídotos/farmacología , Cobamidas/farmacología , Cianuros/toxicidad , Hidroxocobalamina/farmacología , Análisis Espectral/métodos , Análisis de Varianza , Animales , Antídotos/química , Cobamidas/química , Cianuros/sangre , Hemoglobinas/análisis , Hidroxocobalamina/química , Óptica y Fotónica , Oxihemoglobinas/análisis , Conejos
2.
Ann Emerg Med ; 55(4): 352-63, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20045579

RESUMEN

STUDY OBJECTIVE: Exposure to cyanide in fires and industrial exposures and intentional cyanide poisoning by terrorists leading to mass casualties is an ongoing threat. Current treatments for cyanide poisoning must be administered intravenously, and no rapid treatment methods are available for mass casualty cyanide exposures. Cobinamide is a cobalamin (vitamin B(12)) analog with an extraordinarily high affinity for cyanide that is more water-soluble than cobalamin. We investigate the use of intramuscular cobinamide sulfite to reverse cyanide toxicity-induced physiologic changes in a sublethal cyanide exposure animal model and determine the ability of an intramuscular cobinamide sulfite injection to rapidly reverse the physiologic effects of cyanide toxicity. METHODS: New Zealand white rabbits were given 10 mg sodium cyanide intravenously over 60 minutes. Quantitative diffuse optical spectroscopy and continuous-wave near-infrared spectroscopy monitoring of tissue oxyhemoglobin and deoxyhemoglobin concentrations were performed concurrently with blood cyanide level measurements and cobinamide levels. Immediately after completion of the cyanide infusion, the rabbits were injected intramuscularly with cobinamide sulfite (n=6) or inactive vehicle (controls, n=5). RESULTS: Intramuscular administration led to rapid mobilization of cobinamide and was extremely effective at reversing the physiologic effects of cyanide on oxyhemoglobin and within deoxyhemoglobin extraction. Recovery time to 63% of their baseline values in the central nervous system occurred within a mean of 1,032 minutes in the control group and 9 minutes in the cobinamide group, with a difference of 1,023 minutes (95% confidence interval 116 to 1,874 minutes). In muscle tissue, recovery times were 76 and 24 minutes, with a difference of 52 minutes (95% confidence interval 7 to 98 minutes). RBC cyanide levels returned toward normal significantly faster in cobinamide sulfite-treated animals than in control animals. CONCLUSION: Intramuscular cobinamide sulfite rapidly and effectively reverses the physiologic effects of cyanide poisoning, suggesting that a compact cyanide antidote kit can be developed for mass casualty cyanide exposures.


Asunto(s)
Antídotos/uso terapéutico , Cobamidas/uso terapéutico , Cianuros/envenenamiento , Animales , Antídotos/administración & dosificación , Antídotos/farmacocinética , Cobamidas/administración & dosificación , Cobamidas/farmacocinética , Modelos Animales de Enfermedad , Hemoglobinas/análisis , Inyecciones Intramusculares , Oxihemoglobinas/análisis , Conejos , Espectroscopía Infrarroja Corta , Factores de Tiempo
3.
J Infect Dis ; 197(3): 457-64, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18199034

RESUMEN

Some Pseudomonas aeruginosa strains are cyanogenic, and cyanide may contribute to the bacterium's virulence. Using human isolates of P. aeruginosa, we have shown that Drosophila melanogaster suspended above cyanogenic strains become motionless and develop bradycardia and that flies injected with cyanogenic bacterial strains die more rapidly than those injected with noncyanogenic strains. Flies exposed to cyanogenic strains had high cyanide and low adenosine triphosphate (ATP) concentrations in body extracts, and treatment with a cyanide antidote equalized survival of flies injected with cyanogenic and noncyanogenic strains. P. aeruginosa PAO1 strain with a mutation in the hydrogen cyanide synthase gene cluster was much less toxic to flies than the parental cyanogenic strain or 2 knock-in strains. Transgenic flies overexpressing rhodanese, which detoxifies cyanide by converting it to thiocyanate, were resistant to cyanide and the increased virulence of cyanogenic strains. We conclude that D. melanogaster is a good model for studying cyanide produced by P. aeruginosa.


Asunto(s)
Cianuros/aislamiento & purificación , Cianuros/toxicidad , Drosophila melanogaster/efectos de los fármacos , Pseudomonas aeruginosa/aislamiento & purificación , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Animales , Drosophila melanogaster/clasificación , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Nitratos/farmacología , Nitritos/farmacología , Pseudomonas aeruginosa/fisiología , Tiocianatos/aislamiento & purificación , Tiocianatos/metabolismo , Tiocianatos/farmacología
4.
Exp Biol Med (Maywood) ; 232(11): 1432-40, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18040067

RESUMEN

A limited number of nitric oxide (NO)-generating drugs are available for clinical use for acute and chronic conditions. Most of these agents are organic nitrates, which do not directly release NO; tolerance to the drugs develops, in part, as a consequence of their conversion to NO. We synthesized nitrosyl-cobinamide (NO-Cbi) from cobinamide, a structural analog of cobalamin (vitamin B12). NO-Cbi is a direct NO-releasing agent that we found was stable in water, but under physiologic conditions, it released NO with a half-life of 30 mins to 1 h. We show in five different biological systems that NO-Cbi is an effective NO-releasing drug. First, in cultured rat vascular smooth muscle cells, NO-Cbi induced phosphorylation of vasodilator-stimulated phosphoprotein, a downstream target of cGMP and cGMP-dependent protein kinase. Second, in isolated Drosophila melanogaster Malpighian tubules, NO-Cbi-stimulated fluid secretion was similar to that stimulated by Deta-NONOate and a cGMP analog. Third, in isolated mouse hearts, NO-Cbi increased coronary flow much more potently than nitroglycerin. Fourth, in contracted mouse aortic rings, NO-Cbi induced relaxation, albeit to a lesser extent than sodium nitroprusside. Fifth, in intact mice, a single NO-Cbi injection rapidly reduced blood pressure, and blood pressure returned to normal after 45 mins; repeated NO-Cbi injections induced the expected fall in blood pressure. These studies indicate that NO-Cbi is a useful NO donor that can be used experimentally in the laboratory; moreover, it could be developed into a vasodilating drug for treating hypertension and potentially other diseases such as angina and congestive heart failure.


Asunto(s)
Cobamidas/farmacología , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico/metabolismo , Compuestos Nitrosos/farmacología , Vasodilatadores/farmacología , Angina de Pecho/tratamiento farmacológico , Angina de Pecho/metabolismo , Animales , Aorta/metabolismo , Presión Sanguínea/efectos de los fármacos , Cobamidas/síntesis química , Cobamidas/química , Circulación Coronaria/efectos de los fármacos , Drosophila melanogaster , Evaluación Preclínica de Medicamentos , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/metabolismo , Hipertensión/tratamiento farmacológico , Hipertensión/metabolismo , Túbulos de Malpighi/metabolismo , Ratones , Relajación Muscular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocardio/metabolismo , Donantes de Óxido Nítrico/síntesis química , Donantes de Óxido Nítrico/química , Compuestos Nitrosos/síntesis química , Compuestos Nitrosos/química , Técnicas de Cultivo de Órganos , Ratas , Vasodilatadores/síntesis química , Vasodilatadores/química
5.
Exp Biol Med (Maywood) ; 232(6): 789-98, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17526771

RESUMEN

Sodium nitroprusside is used to treat hypertensive emergencies and acute heart failure. It acts by releasing nitric oxide (NO), a highly potent vasodilator, but unfortunately, for each NO molecule released, five cyanide ions are released. Thus, nitroprusside therapy is limited by cyanide toxicity. Therefore, a cyanide scavenger could be beneficial when administering nitroprusside. Hydroxocobalamin, which has a relatively high binding affinity for cyanide, has been shown to reduce cyanide levels in nitroprusside-treated patients. Cobinamide, the penultimate precursor in hydroxocobalamin biosynthesis, has a much greater affinity for cyanide than cobalamin, and binds two cyanide ions. We now show that cobinamide is highly effective in neutralizing cyanide ions released by nitroprusside in cultured mammalian cells, Drosophila melanogaster, and mice. Cobinamide also binds NO, but at molar concentrations 2.5-5 times that of nitroprusside, it did not decrease NO concentrations or the physiological effectiveness of nitroprusside. We conclude that cobinamide could be a valuable adjunct to nitroprusside therapy.


Asunto(s)
Antihipertensivos/farmacocinética , Cobamidas/farmacología , Cianuros/metabolismo , Óxido Nítrico/metabolismo , Nitroprusiato/farmacocinética , Adenosina Trifosfato/metabolismo , Animales , Antihipertensivos/farmacología , Células Cultivadas , Drosophila melanogaster , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Túbulos de Malpighi/efectos de los fármacos , Túbulos de Malpighi/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Nitratos/metabolismo , Nitritos/metabolismo , Nitroprusiato/farmacología , Consumo de Oxígeno/efectos de los fármacos , Arteria Pulmonar/citología , Ratas , Tiocianatos/sangre , Tiocianatos/orina , Vitamina B 12
6.
FASEB J ; 20(11): 1865-73, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16940158

RESUMEN

Septic shock has an extremely high mortality rate, with approximately 200,000 people dying from sepsis annually in the U.S. The high mortality results in part from severe hypotension secondary to high serum NO concentrations. Reducing NO levels should be beneficial in sepsis, but NOS inhibitors have had a checkered history in animal models, and one such agent increased mortality in a clinical trial. An alternative approach to reduce NO levels in sepsis is to use an NO scavenger, which should leave sufficient free NO for normal physiological functions. Using a well-established model of bacterial sepsis in Drosophila melanogaster, we found that cobinamide, a B(12) analog and an effective NO scavenger in vitro, dramatically improved fly survival. Cobinamide augmented the effect of an antibiotic and was beneficial even in immune-deficient flies. Cobinamide's mechanism of action appeared to be from reducing NO levels and improving cardiac function.


Asunto(s)
Bacteriemia/fisiopatología , Cobamidas/farmacología , Cobamidas/fisiología , Drosophila melanogaster/microbiología , Animales , Cobamidas/administración & dosificación , Suplementos Dietéticos , Proteínas de Drosophila/deficiencia , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Infecciones por Escherichia coli/fisiopatología , Choque Séptico/prevención & control , Infecciones Estafilocócicas/fisiopatología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/crecimiento & desarrollo
7.
Exp Biol Med (Maywood) ; 231(5): 641-9, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16636313

RESUMEN

Cyanide is a highly toxic agent that inhibits mitochondrial cytochrome-c oxidase, thereby depleting cellular ATP. It contributes to smoke inhalation deaths in fires and could be used as a weapon of mass destruction. Cobalamin (vitamin B12) binds cyanide with a relatively high affinity and is used in Europe to treat smoke inhalation victims. Cobinamide, the penultimate compound in cobalamin biosynthesis, binds cyanide with about 10(10) greater affinity than cobalamin, and we found it was several-fold more effective than cobalamin in (i) reversing cyanide inhibition of oxidative phosphorylation in mammalian cells; (ii) rescuing mammalian cells and Drosophila melanogaster from cyanide toxicity; and (iii) reducing cyanide inhibition of Drosophila Malpighian tubule secretion. Cobinamide could be delivered by oral ingestion, inhalation, or injection to Drosophila, and it was as effective when administered up to 5 mins post-cyanide exposure as when given pre-exposure. We conclude that cobinamide is an effective cyanide detoxifying agent that has potential use as a cyanide antidote, both in smoke inhalation victims and in persons exposed to cyanide used as a weapon of mass destruction.


Asunto(s)
Cobamidas/farmacocinética , Cianuros/toxicidad , Inactivación Metabólica , Vitamina B 12/metabolismo , Complejo Vitamínico B/metabolismo , Animales , Células CHO , Cobamidas/química , Cobamidas/uso terapéutico , Cricetinae , Cianuros/administración & dosificación , Cianuros/metabolismo , Drosophila melanogaster , Humanos , Estructura Molecular , Lesión por Inhalación de Humo/tratamiento farmacológico , Vitamina B 12/química , Vitamina B 12/uso terapéutico
8.
J Biol Chem ; 280(11): 10073-82, 2005 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-15647267

RESUMEN

Methylmalonyl-CoA mutase is a key enzyme in intermediary metabolism, and children deficient in enzyme activity have severe metabolic acidosis. We found that nitric oxide (NO) inhibits methylmalonyl-CoA mutase activity in rodent cell extracts. The inhibition of enzyme activity occurred within minutes and was not prevented by thiols, suggesting that enzyme inhibition was not occurring via NO reaction with cysteine residues to form nitrosothiol groups. Enzyme inhibition was dependent on the presence of substrate, implying that NO was reacting with cobalamin(II) (Cbl(II)) and/or the deoxyadenosyl radical (.CH(2)-Ado), both of which are generated from the co-factor of the enzyme, 5'-deoxyadenosyl-cobalamin (AdoCbl), on substrate binding. Consistent with this hypothesis was the finding that high micromolar concentrations (> or =600 microm) of oxygen also inhibited enzyme activity. To study the mechanism of NO reaction with AdoCbl, we simulated the enzymatic reaction by photolyzing AdoCbl, and found that even at low NO concentrations, NO reacted with both the generated Cbl(II) and .CH(2)-Ado indicating that NO could effectively compete with the back formation of AdoCbl. Thus, NO inhibition of methylmalonyl-CoA mutase appeared to be from the reaction of NO with both AdoCbl intermediates (Cbl(II) and .CH(2)-Ado) generated during the enzymatic reaction. The inhibition of methylmalonyl-CoA mutase by NO was likely of physiological relevance because a NO donor inhibited enzyme activity in intact cells, and scavenging NO from cells or inhibiting cellular NO synthesis increased methylmalonyl-CoA mutase activity when measured subsequently in cell extracts.


Asunto(s)
Metilmalonil-CoA Mutasa/antagonistas & inhibidores , Óxido Nítrico/metabolismo , Animales , Arginina/química , Línea Celular , Proliferación Celular , Cricetinae , Inhibidores Enzimáticos/farmacología , Cinética , Luz , Mercaptoetanol/farmacología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/química , Donantes de Óxido Nítrico , Nitrógeno/química , Oxígeno/química , Oxígeno/metabolismo , Propionatos/química , Unión Proteica , Espectrofotometría , Ácido Succínico/química , Factores de Tiempo
9.
J Biol Chem ; 280(10): 8678-85, 2005 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-15632180

RESUMEN

Nitric oxide (NO) is an important signaling molecule, and a number of NO synthesis inhibitors and scavengers have been developed to allow study of NO functions and to reduce excess NO levels in disease states. We showed previously that cobinamide, a cobalamin (vitamin B12) precursor, binds NO with high affinity, and we now evaluated the potential of cobinamide as a NO scavenger in biologic systems. We found that cobinamide reversed NO-stimulated fluid secretion in Drosophila Malpighian tubules, both when applied in the form of a NO donor and when produced intracellularly by nitricoxide synthase. Moreover, feeding flies cobinamide markedly attenuated subsequent NO-induced increases in tubular fluid secretion. Cobinamide was taken up efficiently by cultured rodent cells and prevented NO-induced phosphorylation of the vasodilator-stimulated phosphoprotein VASP both when NO was provided to the cells and when NO was generated intracellularly. Cobinamide appeared to act via scavenging NO because it reduced nitrite and nitrate concentrations in both the fly and mammalian cell systems, and it did not interfere with cGMP-induced phosphorylation of VASP. In rodent and human cells, cobinamide exhibited toxicity at concentrations > or =50 microM with toxicity completely prevented by providing equimolar amounts of cobalamin. Combining cobalamin with cobinamide had no effect on the ability of cobinamide to scavenge NO. Cobinamide did not inhibit the in vitro activity of either of the two mammalian cobalamin-dependent enzymes, methionine synthase or methylmalonyl-coenzyme A mutase; however, it did inhibit the in vivo activities of the enzymes in the absence, but not presence, of cobalamin, suggesting that cobinamide toxicity was secondary to interference with cobalamin metabolism. As part of these studies, we developed a facile method for producing and purifying cobinamide. We conclude that cobinamide is an effective intra- and extracellular NO scavenger whose modest toxicity can be eliminated by cobalamin.


Asunto(s)
Cobamidas/química , Cobamidas/metabolismo , Óxido Nítrico/metabolismo , Vitamina B 12/química , Vitamina B 12/metabolismo , Animales , Calcimicina/farmacología , Moléculas de Adhesión Celular/metabolismo , División Celular/efectos de los fármacos , Línea Celular , Cobamidas/farmacología , Drosophila , Túbulos de Malpighi/fisiología , Proteínas de Microfilamentos/metabolismo , Estructura Molecular , Nitratos/metabolismo , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Nitritos/metabolismo , Fosfoproteínas/metabolismo , Fosforilación
10.
Biochemistry ; 42(29): 8900-8, 2003 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-12873151

RESUMEN

Despite early claims that nitric oxide does not react with cobalamin under any circumstances, it is now accepted that NO has a high affinity for cobalamin in the 2+ oxidation state [Cbl(II)]. However, it is still the consensus that NO does not react with Cbl(III). We confirmed that NO coordinates to Cbl(II) at all pH values and that Cbl(III) does not react with NO at neutral pH. At low pH, however, Cbl(III) does react with NO by way of a two-step process that also reduces Cbl(III) to Cbl(II). To account for the pH dependence, and because of its intrinsic interest, we also studied reactions of NO with cobinamide [Cbi] in the 2+ and 3+ oxidation states. Both Cbi(II) and Cbi(III) react readily with NO at all pH values. Again, Cbi(III) is reduced during the process of coordinating NO. Compared to cobalamin, cobinamide lacks the tethered 5,6-dimethylbenzamidazolyl moiety bound to the cobalt ion. It may, therefore, be considered a "base-off" form of Cbl. To explain the reaction of Cbl(III) at low pH, we infer that the base-off form of Cbl(III) exists in trace amounts that are rapidly reduced to Cbl(II), which then binds NO efficiently. Base dissociation, we postulate, is the rate-limiting step. Interestingly, Cbi(II) has 100 times greater affinity for NO than does Cbl(II), proving that there is a strong trans effect due to the tethered base in nitrosyl derivatives of both Cbl(II) and Cbl(III). The affinity of Cbi(II) for NO is so high that it is a very efficient NO trap and, consequently, may have important biomedical uses.


Asunto(s)
Cobamidas/química , Óxido Nítrico/química , Vitamina B 12/química , Bencimidazoles/química , Concentración de Iones de Hidrógeno , Cinética , Luz , Modelos Químicos , Nitrógeno/química , Unión Proteica , Factores de Tiempo
11.
Biochemistry ; 42(21): 6500-6, 2003 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-12767233

RESUMEN

Oxygenase and reductase domains in nitric oxide synthase are linked by a peptide region that binds calmodulin. Here we study the effects of modifying the length of the interdomain linker in a deletion mutant lacking 15 amino acids (residues 503-517) in bovine eNOS. The kinetics of CO ligation with the mutant were determined in the presence and absence of tetrahydrobiopterin and arginine and compared with the CO binding kinetics of wild-type eNOS and the eNOS oxygenase domain. In the mutant, electron flow is interrupted. The association kinetics of CO with both mutant and wild-type eNOS can be approximated with two kinetic phases, but the relative proportions change in the mutant. Both the abrogation of electron flow in the mutant and the differences in CO binding may be explained by an alteration in the docking of the FMN domain to the heme domain. We propose that the calmodulin binding residues form a helix that is critical for the proper alignment of the adjacent reductase and oxygenase domains within the active eNOS dimer in achieving proper electron transfer between them.


Asunto(s)
Calmodulina/química , Monóxido de Carbono/química , Óxido Nítrico Sintasa/química , Secuencia de Aminoácidos , Animales , Bovinos , Dimerización , Electrones , Eliminación de Gen , Hemo/química , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Óxido Nítrico Sintasa de Tipo III , Péptidos/química , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...