Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
J Cell Biol ; 220(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34328510

RESUMEN

Ferroptosis is a form of iron-dependent regulated cell death driven by uncontrolled lipid peroxidation. Mitochondria are double-membrane organelles that have essential roles in energy production, cellular metabolism, and cell death regulation. However, their role in ferroptosis has been unclear and somewhat controversial. In this Perspective, I summarize the diverse metabolic processes in mitochondria that actively drive ferroptosis, discuss recently discovered mitochondria-localized defense systems that detoxify mitochondrial lipid peroxides and protect against ferroptosis, present new evidence for the roles of mitochondria in regulating ferroptosis, and outline outstanding questions on this fascinating topic for future investigations. An in-depth understanding of mitochondria functions in ferroptosis will have important implications for both fundamental cell biology and disease treatment.


Asunto(s)
Ferroptosis/genética , GTP Ciclohidrolasa/antagonistas & inhibidores , Hierro/metabolismo , Mitocondrias/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Sistema de Transporte de Aminoácidos y+/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos y+/genética , Sistema de Transporte de Aminoácidos y+/metabolismo , Células Cultivadas , Ferroptosis/efectos de los fármacos , GTP Ciclohidrolasa/genética , GTP Ciclohidrolasa/metabolismo , Regulación de la Expresión Génica , Glutatión/antagonistas & inhibidores , Glutatión/biosíntesis , Humanos , Peroxidación de Lípido/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Oxidantes/farmacología , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Especies Reactivas de Oxígeno/agonistas , Especies Reactivas de Oxígeno/metabolismo , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Proteína de Unión al Calcio S100A4/genética , Proteína de Unión al Calcio S100A4/metabolismo , Transducción de Señal , Ubiquinona/antagonistas & inhibidores , Ubiquinona/metabolismo
2.
FEBS J ; 288(6): 1956-1974, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32898935

RESUMEN

Coenzyme Q10 (CoQ, ubiquinone) is a redox-active lipid endogenously synthesized by the cells. The final stage of CoQ biosynthesis is performed at the mitochondrial level by the 'complex Q', where coq2 is responsible for the prenylation of the benzoquinone ring of the molecule. We report that the competitive coq2 inhibitor 4-nitrobenzoate (4-NB) decreased the cellular CoQ content and caused severe impairment of mitochondrial function in the T67 human glioma cell line. In parallel with the reduction in CoQ biosynthesis, the cholesterol level increased, leading to significant perturbation of the plasma membrane physicochemical properties. We show that 4-NB treatment did not significantly affect the cell viability, because of an adaptive metabolic rewiring toward glycolysis. Hypoxia-inducible factor 1α (HIF-1α) stabilization was detected in 4-NB-treated cells, possibly due to the contribution of both reduction in intracellular oxygen tension and ROS overproduction. Exogenous CoQ supplementation partially recovered cholesterol content, HIF-1α degradation, and ROS production, whereas only weakly improved the bioenergetic impairment induced by the CoQ depletion. Our data provide new insights on the effect of CoQ depletion and contribute to shed light on the pathogenic mechanisms of ubiquinone deficiency syndrome.


Asunto(s)
Metabolismo Energético , Glucólisis , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ubiquinona/análogos & derivados , Transferasas Alquil y Aril/antagonistas & inhibidores , Transferasas Alquil y Aril/metabolismo , Ataxia/metabolismo , Línea Celular Tumoral , Colesterol/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Debilidad Muscular/metabolismo , Nitrobenzoatos/farmacología , Estabilidad Proteica/efectos de los fármacos , Ubiquinona/antagonistas & inhibidores , Ubiquinona/biosíntesis , Ubiquinona/deficiencia , Ubiquinona/metabolismo
3.
J Biochem ; 163(4): 329-339, 2018 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-29319808

RESUMEN

Coenzyme Q (CoQ) is an essential factor of the mitochondrial respiratory chain. CoQ homologues with different lengths of the isoprenoid side chain are widely distributed in nature, but little is known about the relationship between the isoprenoid side chain length and biological function; therefore, we examined the effects of CoQ homologues on HeLa cells. When CoQ homologues with a shorter isoprenoid side chain than CoQ4 were added to HeLa cells, they induced cell death, and the order of cytotoxic intensity was as follows: CoQ0 ≫ CoQ3 ≈ CoQ1 > CoQ2 ≫ CoQ4. Furthermore, we found that CoQ1, CoQ2 and CoQ3 could induce caspase-mediated apoptosis, and the order of intensity was as follows: CoQ3 > CoQ2 ≥ CoQ1. We could not identify the participation of reactive oxygen species in the apoptosis induction, but observed that an NAD(P)H dehydrogenase (quinone) 1 (NQO1) inhibitor, dicumarol, could inhibit not only the intracellular reduction of the homologues but also apoptosis. However, because dicumarol did not affect well-known apoptosis inducers, such as anti-Fas IgG, tumor necrosis factor (TNF)-α, TNF-related apoptosis-inducing ligand, UV-B and H2O2 of HeLa cells at all, we concluded that NQO1-related intracellular reduction of CoQ, or its reduced product, ubiquinol, may participate in the apoptosis induction of HeLa cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Terpenos/farmacología , Ubiquinona/metabolismo , Dicumarol/farmacología , Células HeLa , Humanos , Oxidación-Reducción , Terpenos/química , Terpenos/metabolismo , Ubiquinona/antagonistas & inhibidores
4.
Arch Biochem Biophys ; 580: 75-83, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26116786

RESUMEN

Manganese-induced toxicity has been recently associated with an increased ROS generation from mitochondrial complex II (succinate:ubiquinone oxidoreductase). To achieve a deeper mechanistic understanding how divalent manganese ions (Mn(2+)) could stimulate mitochondrial ROS production we performed investigations with bovine heart submitochondrial particles (SMP). In succinate fueled SMP, the Mn(2+) induced hydrogen peroxide (H2O2) production was blocked by the specific complex II ubiquinone binding site (IIQ) inhibitor atpenin A5 while a further downstream block at complex III increased the rate markedly. This suggests that site IIQ was the source of the reactive oxygen species. Moreover, Mn(2+) ions also accelerated the rate of superoxide dismutation, explaining the general increase in the measured rates of H2O2 production and an attenuation of direct superoxide detection.


Asunto(s)
Complejo II de Transporte de Electrones/metabolismo , Peróxido de Hidrógeno/metabolismo , Manganeso/farmacología , Membranas Mitocondriales/efectos de los fármacos , Partículas Submitocóndricas/efectos de los fármacos , Animales , Cationes Bivalentes , Bovinos , Complejo III de Transporte de Electrones/metabolismo , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Membranas Mitocondriales/metabolismo , Piridonas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Partículas Submitocóndricas/metabolismo , Ácido Succínico/metabolismo , Ácido Succínico/farmacología , Superóxido Dismutasa/metabolismo , Ubiquinona/antagonistas & inhibidores , Ubiquinona/metabolismo
5.
PLoS One ; 10(5): e0126220, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25970180

RESUMEN

Today HIV-1 infection is recognized as a chronic disease with obligatory lifelong treatment to keep viral titers below detectable levels. The continuous intake of antiretroviral drugs however, leads to severe and even life-threatening side effects, supposedly by the deleterious impact of nucleoside-analogue type compounds on the functioning of the mitochondrial DNA polymerase. For detailed investigation of the yet partially understood underlying mechanisms, the availability of a versatile model system is crucial. We therefore set out to develop the use of Caenorhabditis elegans to study drug induced mitochondrial toxicity. Using a combination of molecular-biological and functional assays, combined with a quantitative analysis of mitochondrial network morphology, we conclude that anti-retroviral drugs with similar working mechanisms can be classified into distinct groups based on their effects on mitochondrial morphology and biochemistry. Additionally we show that mitochondrial toxicity of antiretroviral drugs cannot be exclusively attributed to interference with the mitochondrial DNA polymerase.


Asunto(s)
Fármacos Anti-VIH/toxicidad , Caenorhabditis elegans/efectos de los fármacos , ADN Mitocondrial/antagonistas & inhibidores , Evaluación de Medicamentos/métodos , Mitocondrias/efectos de los fármacos , Inhibidores de la Transcriptasa Inversa/toxicidad , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , ADN Mitocondrial/metabolismo , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Didanosina/toxicidad , Didesoxinucleósidos/toxicidad , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Modelos Biológicos , Consumo de Oxígeno/efectos de los fármacos , Estavudina/toxicidad , Ubiquinona/antagonistas & inhibidores , Ubiquinona/metabolismo , Zalcitabina/toxicidad , Zidovudina/toxicidad
6.
Metabolism ; 63(6): 735-45, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24641882

RESUMEN

New-onset diabetes has been observed in clinical trials and meta-analyses involving statin therapy. To explain this association, three major mechanisms have been proposed and discussed in the literature. First, certain statins affect insulin secretion through direct, indirect or combined effects on calcium channels in pancreatic ß-cells. Second, reduced translocation of glucose transporter 4 in response to treatment results in hyperglycemia and hyperinsulinemia. Third, statin therapy decreases other important downstream products, such as coenzyme Q10, farnesyl pyrophosphate, geranylgeranyl pyrophosphate, and dolichol; their depletion leads to reduced intracellular signaling. Other possible mechanisms implicated in the effect of statins on new-onset diabetes are: statin interference with intracellular insulin signal transduction pathways via inhibition of necessary phosphorylation events and reduction of small GTPase action; inhibition of adipocyte differentiation leading to decreased peroxisome proliferator activated receptor gamma and CCAAT/enhancer-binding protein which are important pathways for glucose homeostasis; decreased leptin causing inhibition of ß-cells proliferation and insulin secretion; and diminished adiponectin levels. Given that the magnitude of the risk of new-onset diabetes following statin use remains to be fully clarified and the well-established beneficial effect of statins in reducing cardiovascular risk, statins remain the first-choice treatment for prevention of CVD. Elucidation of the mechanisms underlying the development of diabetes in association with statin use may help identify novel preventative or therapeutic approaches to this problem and/or help design a new generation statin without such side-effects.


Asunto(s)
Diabetes Mellitus/inducido químicamente , Diabetes Mellitus/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Adipocitos/efectos de los fármacos , Adiponectina/metabolismo , Animales , Canales de Calcio/efectos de los fármacos , Canales de Calcio/metabolismo , Caveolinas/metabolismo , Diferenciación Celular/efectos de los fármacos , Dolicoles/antagonistas & inhibidores , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Hiperglucemia/inducido químicamente , Hiperglucemia/metabolismo , Hiperinsulinismo/inducido químicamente , Hiperinsulinismo/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Canales Iónicos/metabolismo , Leptina/metabolismo , MicroARNs/metabolismo , Proteínas Mitocondriales/metabolismo , Terpenos/antagonistas & inhibidores , Ubiquinona/análogos & derivados , Ubiquinona/antagonistas & inhibidores , Proteína Desacopladora 3
7.
J Antibiot (Tokyo) ; 67(2): 147-51, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24169795

RESUMEN

Polymyxin B and colistin were examined for their ability to inhibit the type II NADH-quinone oxidoreductases (NDH-2) of three species of Gram-negative bacteria. Polymyxin B and colistin inhibited the NDH-2 activity in preparations from all of the isolates in a concentration-dependent manner. The mechanism of NDH-2 inhibition by polymyxin B was investigated in detail with Escherichia coli inner membrane preparations and conformed to a mixed inhibition model with respect to ubiquinone-1 and a non-competitive inhibition model with respect to NADH. These suggest that the inhibition of vital respiratory enzymes in the bacterial inner membrane represents one of the secondary modes of action for polymyxins.


Asunto(s)
Colistina/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/enzimología , Polimixina B/farmacología , Quinona Reductasas/antagonistas & inhibidores , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/enzimología , Antibacterianos/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Colistina/análogos & derivados , Escherichia coli/efectos de los fármacos , Escherichia coli/enzimología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/enzimología , NAD/antagonistas & inhibidores , Quinona Reductasas/efectos de los fármacos , Ubiquinona/antagonistas & inhibidores
8.
J Clin Invest ; 123(12): 5179-89, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24270420

RESUMEN

Identification of single-gene causes of steroid-resistant nephrotic syndrome (SRNS) has furthered the understanding of the pathogenesis of this disease. Here, using a combination of homozygosity mapping and whole human exome resequencing, we identified mutations in the aarF domain containing kinase 4 (ADCK4) gene in 15 individuals with SRNS from 8 unrelated families. ADCK4 was highly similar to ADCK3, which has been shown to participate in coenzyme Q10 (CoQ10) biosynthesis. Mutations in ADCK4 resulted in reduced CoQ10 levels and reduced mitochondrial respiratory enzyme activity in cells isolated from individuals with SRNS and transformed lymphoblasts. Knockdown of adck4 in zebrafish and Drosophila recapitulated nephrotic syndrome-associated phenotypes. Furthermore, ADCK4 was expressed in glomerular podocytes and partially localized to podocyte mitochondria and foot processes in rat kidneys and cultured human podocytes. In human podocytes, ADCK4 interacted with members of the CoQ10 biosynthesis pathway, including COQ6, which has been linked with SRNS and COQ7. Knockdown of ADCK4 in podocytes resulted in decreased migration, which was reversed by CoQ10 addition. Interestingly, a patient with SRNS with a homozygous ADCK4 frameshift mutation had partial remission following CoQ10 treatment. These data indicate that individuals with SRNS with mutations in ADCK4 or other genes that participate in CoQ10 biosynthesis may be treatable with CoQ10.


Asunto(s)
Síndrome Nefrótico/genética , Proteínas Quinasas/fisiología , Ubiquinona/análogos & derivados , Adolescente , Corticoesteroides/farmacología , Corticoesteroides/uso terapéutico , Secuencia de Aminoácidos , Animales , Células Cultivadas , Niño , Consanguinidad , Secuencia Conservada , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/genética , Resistencia a Medicamentos , Exoma/genética , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Mitocondrias/fisiología , Datos de Secuencia Molecular , Mutación , Síndrome Nefrótico/tratamiento farmacológico , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/patología , Podocitos/metabolismo , Podocitos/ultraestructura , Proteínas Quinasas/deficiencia , Proteínas Quinasas/genética , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Ubiquinona/antagonistas & inhibidores , Ubiquinona/biosíntesis , Ubiquinona/metabolismo , Ubiquinona/uso terapéutico , Adulto Joven , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética
9.
Biochem Biophys Res Commun ; 425(2): 348-52, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22842579

RESUMEN

Ubiquinone (Q) is a product in the cholesterol synthesis pathway and is an essential component of the respiratory chain in the mitochondrial membrane. In addition, extra-mitochondrial Q has anti-oxidative properties and this fraction is increased during carcinogenesis. The aim of the present study was to investigate if extra-mitochondrial level of Q is affected by statin treatment in a rat model for liver cancer, and if this change correlates with inhibited carcinogenesis. To do this we isolated sub-cellular fractions of rat livers from a previous experiment where we have shown anti-carcinogenic effects of statins. The levels of Q(8), Q(9) and Q(10) were analysed with liquid chromatography-mass spectrometry. The Q(9)-levels, constituting the major part of Q in rats, were not significantly affected in any of the sub-cellular compartments. The levels of Q(10), constituting a minor part of Q in rats but the major part of Q in humans, were significantly decreased by about 60% in the statin treated rats. The decrease was present in all sub-cellular compartments, but was most pronounced in the cytosol. There was a significant correlation between extra-mitochondrial Q(10) levels and inhibited carcinogenesis. No such correlation was observed with extra-mitochondrial Q(9). The reduced Q(10)-levels might be explained by the reduced availability of isoprene units during statin treatment, shifting the synthesis towards isoforms with shorter side-chains. In line with this hypothesis there were increased levels of Q(8)-levels during statin treatment. The results support our previous suggestion that at least part of the anti-carcinogenic effect of statins in our rat model is mediated by effects on synthesis of Q. We also demonstrate a shift in the Q-synthesis pathway towards isoforms with shorter side-chains during statin treatment. The ratio between the different Q-isoforms might be used as a more sensitive marker of statin-induced inhibition of Q than measuring total Q levels.


Asunto(s)
Anticarcinógenos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Neoplasias Hepáticas/enzimología , Ubiquinona/biosíntesis , Animales , Modelos Animales de Enfermedad , Isoenzimas/antagonistas & inhibidores , Isoenzimas/biosíntesis , Masculino , Mitocondrias Hepáticas/enzimología , Ratas , Ratas Endogámicas F344 , Ubiquinona/antagonistas & inhibidores
10.
PLoS One ; 7(2): e30606, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22359546

RESUMEN

Coenzyme Q(10) (CoQ(10)) is a potent lipophilic antioxidant in cell membranes and a carrier of electrons in the mitochondrial respiratory chain. We previously characterized the effects of varying severities of CoQ(10) deficiency on ROS production and mitochondrial bioenergetics in cells harboring genetic defects of CoQ(10) biosynthesis. We observed a unimodal distribution of ROS production with CoQ(10) deficiency: cells with <20% of CoQ(10) and 50-70% of CoQ(10) did not generate excess ROS while cells with 30-45% of CoQ(10) showed increased ROS production and lipid peroxidation. Because our previous studies were limited to a small number of mutant cell lines with heterogeneous molecular defects, here, we treated 5 control and 2 mildly CoQ(10) deficient fibroblasts with varying doses of 4-nitrobenzoate (4-NB), an analog of 4-hydroxybenzoate (4-HB) and inhibitor of 4-para-hydroxybenzoate:polyprenyl transferase (COQ2) to induce a range of CoQ(10) deficiencies. Our results support the concept that the degree of CoQ(10) deficiency in cells dictates the extent of ATP synthesis defects and ROS production and that 40-50% residual CoQ(10) produces maximal oxidative stress and cell death.


Asunto(s)
Fibroblastos/efectos de los fármacos , Nitrobenzoatos/farmacología , Ubiquinona/análogos & derivados , Adenosina Trifosfato/biosíntesis , Muerte Celular , Relación Dosis-Respuesta a Droga , Humanos , Estrés Oxidativo , Especies Reactivas de Oxígeno , Ubiquinona/antagonistas & inhibidores , Ubiquinona/biosíntesis
11.
Protein Cell ; 2(7): 531-42, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21822798

RESUMEN

The mitochondrial respiratory complex II or succinate: ubiquinone oxidoreductase (SQR) is a key membrane complex in both the tricarboxylic acid cycle and aerobic respiration. Five disinfectant compounds were investigated with their potent inhibition effects on the ubiquinone reduction activity of the porcine mitochondrial SQR by enzymatic assay and crystallography. Crystal structure of the SQR bound with thiabendazole (TBZ) reveals a different inhibitor-binding feature at the ubiquinone binding site where a water molecule plays an important role. The obvious inhibitory effect of TBZ based on the biochemical data (IC(50) ~100 µmol/L) and the significant structure-based binding affinity calculation (~94 µmol/L) draw the suspicion of using TBZ as a good disinfectant compound for nematode infections treatment and fruit storage.


Asunto(s)
Complejo II de Transporte de Electrones/efectos de los fármacos , Mitocondrias/enzimología , Oxidorreductasas/antagonistas & inhibidores , Tiabendazol/farmacología , Ubiquinona/antagonistas & inhibidores , Agua/metabolismo , Animales , Antihelmínticos/metabolismo , Antihelmínticos/farmacología , Sitios de Unión , Cristalografía por Rayos X , Concentración 50 Inhibidora , Mitocondrias/efectos de los fármacos , Estructura Molecular , Oxidorreductasas/química , Relación Estructura-Actividad , Porcinos , Tiabendazol/química , Tiabendazol/metabolismo , Agua/química
12.
Neurosci Lett ; 501(2): 92-5, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21763754

RESUMEN

Statins have been suggested to protect against Alzheimer's disease (AD). Recently, however, we reported that aged dogs that underwent chronic statin treatment exhibited cognitive deficits compared with age matched controls. In human studies, blood levels of Coenzyme Q10 (CoQ10) decrease with statin use. CoQ10 is important for proper mitochondrial function and is a powerful antioxidant, two important factors for cognitive health in aging. Thus, the current study tested the hypothesis that CoQ10 levels in the serum and/or parietal cortex are decreased in statin treated dogs and are associated with poorer cognition. Six aged beagles (>8 years) were administered 80 mg/day of atorvastatin for 14.5 months and compared with placebo-treated animals. As predicted, serum CoQ10 was significantly lower in statin-treated dogs. Parietal cortex CoQ10 was not different between the two groups. However, poorer cognition was correlated with lower parietal cortex CoQ10. This study in dogs suggests that serum CoQ10 is reduced with atorvastatin treatment. CoQ10 levels in brain may be linked to impaired cognition in response to atorvastatin, in agreement with previous reports that statins may have a negative impact on cognition in the elderly.


Asunto(s)
Trastornos del Conocimiento/metabolismo , Ácidos Heptanoicos/administración & dosificación , Pirroles/administración & dosificación , Ubiquinona/análogos & derivados , Envejecimiento/metabolismo , Animales , Atorvastatina , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/fisiopatología , Perros , Femenino , Ácidos Heptanoicos/efectos adversos , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Lóbulo Parietal/efectos de los fármacos , Lóbulo Parietal/metabolismo , Lóbulo Parietal/fisiopatología , Pirroles/efectos adversos , Ubiquinona/antagonistas & inhibidores , Ubiquinona/deficiencia , Ubiquinona/fisiología
14.
J Am Coll Cardiol ; 56(15): 1196-204, 2010 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-20883926

RESUMEN

OBJECTIVES: The purpose of this study was to determine whether coenzyme Q10 is an independent predictor of prognosis in heart failure. BACKGROUND: Blood and tissue concentrations of the essential cofactor coenzyme Q10 are decreased by statins, and this could be harmful in patients with heart failure. METHODS: We measured serum coenzyme Q10 in 1,191 patients with ischemic systolic heart failure enrolled in CORONA (Controlled Rosuvastatin Multinational Study in Heart Failure) and related this to clinical outcomes. RESULTS: Patients with lower coenzyme Q10 concentrations were older and had more advanced heart failure. Mortality was significantly higher among patients in the lowest compared to the highest coenzyme Q10 tertile in a univariate analysis (hazard ratio: 1.50, 95% confidence interval: 1.04 to 2.6, p = 0.03) but not in a multivariable analysis. Coenzyme Q10 was not an independent predictor of any other clinical outcome. Rosuvastatin reduced coenzyme Q10 but there was no interaction between coenzyme Q10 and the effect of rosuvastatin. CONCLUSIONS: Coenzyme Q10 is not an independent prognostic variable in heart failure. Rosuvastatin reduced coenzyme Q10, but even in patients with a low baseline coenzyme Q10, rosuvastatin treatment was not associated with a significantly worse outcome. (Controlled Rosuvastatin Multinational Study in Heart Failure [CORONA]; NCT00206310).


Asunto(s)
Fluorobencenos/uso terapéutico , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/tratamiento farmacológico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Pirimidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Ubiquinona/análogos & derivados , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Femenino , Fluorobencenos/efectos adversos , Estudios de Seguimiento , Insuficiencia Cardíaca/diagnóstico , Hospitalización/tendencias , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Internacionalidad , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Pirimidinas/efectos adversos , Rosuvastatina Cálcica , Sulfonamidas/efectos adversos , Resultado del Tratamiento , Ubiquinona/antagonistas & inhibidores , Ubiquinona/sangre
15.
Anticancer Res ; 30(4): 1105-12, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20530415

RESUMEN

AIM: This study tests the hypothesis that statins (HMGCoA reductase inhibitors) inhibit carcinogenesis and that this effect may be mediated by the statin-induced inhibition of ubiquinone synthesis. MATERIALS AND METHODS: The effects of lovastatin, with and without addition of ubiquinone, were studied in a rat model for chemically induced hepatocarcinogenesis. Intermediates in the mevalonate pathway were measured. RESULTS: Lovastatin treatment reduced the volume fraction of liver nodules by 50% and the cell proliferation within the liver nodules was reduced to one third. Ubiquinone (Q10) treatment reversed the statin-induced inhibition of cell proliferation. Lathosterol levels were reduced significantly in the statin-treated rats, indicating inhibition of the mevalonate pathway, but cholesterol levels were not affected. CONCLUSION: Lovastatin inhibits carcinogenesis in a rat model for liver cancer, despite unaffected cholesterol levels. The statin-induced inhibition of cell proliferation may, at least in part, be explained by the inhibition of ubiquinone synthesis.


Asunto(s)
Anticarcinógenos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Neoplasias Hepáticas Experimentales/prevención & control , Lovastatina/farmacología , Ubiquinona/análogos & derivados , Ubiquinona/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Colesterol/metabolismo , Modelos Animales de Enfermedad , Hígado/anatomía & histología , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Masculino , Ácido Mevalónico/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Lesiones Precancerosas/tratamiento farmacológico , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Ratas , Ratas Endogámicas F344 , Ubiquinona/biosíntesis , Ubiquinona/farmacología
16.
Mini Rev Med Chem ; 10(3): 232-62, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20105132

RESUMEN

Elevated lipid level is supposed to be one of the main risk factors of atherosclerosis and related cardiovascular diseases and stroke (and is connected to mortality and morbidity). Therefore, lipid lowering is one of the major approaches in prevention of coronary heart diseases and stroke. Though drugs of various categories acting through different mechanisms are available in the antihyperlipidemic therapy, there are still a few problems associated with the currently available lipid lowering drugs. Therefore, medicinal chemists worldwide are designing, synthesizing and evaluating a variety of new molecules for antihyperlipidemic activity to address these problems. One of the important approaches to this is identifying new drug targets for antihyperlipidemic activity. This review summarizes nineteen recently identified and currently being exploited targets for the ongoing research by researchers world over to discover novel leads as potential drugs for antihyperlipidemic therapy.


Asunto(s)
Hipolipemiantes/química , ATP Citrato (pro-S)-Liasa/antagonistas & inhibidores , ATP Citrato (pro-S)-Liasa/metabolismo , Proteína C-Reactiva/antagonistas & inhibidores , Proteína C-Reactiva/metabolismo , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Proteínas de Transferencia de Ésteres de Colesterol/antagonistas & inhibidores , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Humanos , Hiperlipidemias/tratamiento farmacológico , Hiperlipidemias/etiología , Hipolipemiantes/uso terapéutico , Lipoproteínas/metabolismo , Receptores Activados del Proliferador del Peroxisoma/antagonistas & inhibidores , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Esterol O-Aciltransferasa/antagonistas & inhibidores , Esterol O-Aciltransferasa/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/antagonistas & inhibidores , Ubiquinona/metabolismo
18.
Biofizika ; 52(3): 503-9, 2007.
Artículo en Ruso | MEDLINE | ID: mdl-17633540

RESUMEN

It has been shown that various nitric oxide donors and metabolites have similar effects on lipid peroxidation in rat myocardium homogenate. The formation of malondialdehyde, a secondary product of lipid peroxidation, was inhibited in a dose-dependent manner by PAPA/NONO (a synthetic nitric oxide donor), S-nitrosoglutathione, nitrite, and nitroxyl anion. The inhibition of lipid peroxidation was provided most efficiently by the administration of dinitrosyl-iron complexes with dextran and PAPA/NONO. S-nitrosoglutathione also inhibited the destruction of coenzymes Q9 and Q10 during free radical oxidation of myocardium homogenate. Low-molecular-weight dinitrosyl iron complexes with cysteine also promoted lipid peroxidation, which is probably due to iron release during the destruction dinitrosyl iron complexes. It is likely that the antioxidant action of nitric oxide derivatives is related to the reduction of ferry forms of hemoproteins and interaction of nitric oxide with lipid radicals.


Asunto(s)
Antioxidantes/farmacología , Peroxidación de Lípido/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico/metabolismo , Oxidantes/farmacología , Animales , Coenzimas/antagonistas & inhibidores , Hidrazinas/farmacología , Hierro/farmacología , Masculino , Malondialdehído/análisis , Miocardio/química , Óxido Nítrico/farmacología , Óxidos de Nitrógeno/farmacología , Ratas , Ratas Wistar , S-Nitrosoglutatión/farmacología , Ubiquinona/análogos & derivados , Ubiquinona/antagonistas & inhibidores
19.
Am J Obstet Gynecol ; 194(5): e35-8, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16647895

RESUMEN

OBJECTIVE: The purpose of this study was to examine the influence of menstrual cycle and oral contraceptive use on serum levels of lipid-soluble antioxidants. STUDY DESIGN: In this cross-section study, nonfasting blood samples were collected twice from 10 healthy premenopausal women during the follicular phase (between days 8 and 11) and the luteal phase (between days 18 and 22) of their same menstrual cycle. In addition, blood samples from 15 premenopausal women who used oral contraceptive for at least 6 months and 40 women who did not use oral contraceptive were collected randomly at any day of the menstrual cycle. Serum levels of coenzyme Q10, alpha-tocopherol, gamma-tocopherol, beta-carotene, alpha-carotene, and lycopene were determined using high pressure liquid chromatography. RESULTS: Serum coenzyme Q10 and alpha-tocopherol levels were significantly lower during the follicular phase compared with the luteal phase of the same menstrual cycle (P < .05). Oral contraceptive use also significantly decreased coenzyme Q10 and alpha-tocopherol (P < .001). Other antioxidant levels were comparable. CONCLUSION: Alterations in coenzyme Q10 and alpha-tocopherol levels during the menstrual cycle and in oral contraceptive users should be taken into consideration, concerning the future antioxidant research in premenopausal women. Further studies are needed to investigate the potential role of endogenous and exogenous ovarian hormones on oxidative stress in women.


Asunto(s)
Antioxidantes/metabolismo , Anticonceptivos Orales/farmacología , Ciclo Menstrual/sangre , Adulto , Antioxidantes/química , Coenzimas , Estudios Transversales , Femenino , Fase Folicular/sangre , Humanos , Lípidos , Fase Luteínica/sangre , Premenopausia/sangre , Solubilidad , Ubiquinona/análogos & derivados , Ubiquinona/antagonistas & inhibidores , Ubiquinona/sangre , alfa-Tocoferol/antagonistas & inhibidores , alfa-Tocoferol/sangre
20.
Neurochem Res ; 31(3): 303-11, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16733807

RESUMEN

The neurotoxic effects of cocaine and methamphetamine (METH) were studied in mice brain with a primary objective to determine the neuroprotective potential of coenzyme Q10 (CoQ10) in drug addiction. Repeated treatment of cocaine or METH induced significant reduction in the striatal dopamine and CoQ10 in mice. Cocaine or METH-treated mice exhibited increased thiobarbituric acid reactive substances (TBARs) in the striatum and cerebral cortex without any significant change in the cerebellum. Complex I immunoreactivity was inhibited in both cocaine and METH-treated mice, whereas tyrosine hydroxylase (TH) immunoreactivity was decreased in METH-treated mice and increased in cocaine-treated mice. Neither cocaine nor METH could induce significant change in alpha-synuclein expression at the doses and duration we have used in the present study. CoQ10 treatment attenuated cocaine and METH-induced inhibition in the striatal 18F-DOPA uptake as determined by high-resolution microPET neuroimaging. Hence exogenous administration of CoQ10 may provide neuroprotection in drug addiction.


Asunto(s)
Estimulantes del Sistema Nervioso Central/toxicidad , Cocaína/toxicidad , Cuerpo Estriado/efectos de los fármacos , Metanfetamina/toxicidad , Ubiquinona/análogos & derivados , Animales , Coenzimas , Cuerpo Estriado/metabolismo , Dihidroxifenilalanina/análogos & derivados , Dihidroxifenilalanina/metabolismo , Dopamina/biosíntesis , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/biosíntesis , Immunoblotting , Peroxidación de Lípido/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Microdiálisis , Especies Reactivas de Oxígeno/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Ubiquinona/antagonistas & inhibidores , Ubiquinona/biosíntesis , alfa-Sinucleína/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...