Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
1.
Chem Biol Interact ; 351: 109677, 2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-34634269

RESUMEN

Methylglyoxal (MG) is a reactive metabolite derived from different physiological pathways. Its production can be harmful to cells via glycation reactions of lipids, DNA, and proteins. But, the effects of MG on mitochondrial functioning and bioenergetic responses are still elusive. Then, the effects of MG on key parameters of mitochondrial functionality were examined here. Isolated rat liver mitochondria were exposed to 0.1-10 mM of MG to determine its toxicity in the mitochondrial viability, membrane potential (Δψm), swelling and the superoxide (O2•-) production. Besides, mitochondrial oxidative phosphorylation parameters were analyzed by high-resolution respiratory (HRR) assay. In this set of experiments, routine state, PM state (pyruvate/malate), oxidative phosphorylation (OXPHOS), LEAK respiration, electron transport system (ETS) and oxygen residual (ROX) states were evaluated. HRR showed that PM state, OXPHOS CI-Linked, LEAK respiration, ETS CI/CII-Linked and ETS CII-Linked/ROX were significantly inhibited by MG exposure. MG also inhibited the complex II activity, and decreased Δψm and the viability of mitochondria. Taken together, our data indicates that MG is an inductor of mitochondrial dysfunctions and impairs important steps of respiratory chain, effects that can alter bioenergetics responses.


Asunto(s)
Inhibidores Enzimáticos/toxicidad , Mitocondrias/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Piruvaldehído/toxicidad , Animales , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratas Wistar
2.
J Pharmacol Sci ; 147(4): 367-375, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34663519

RESUMEN

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by CAG repeat expansion in the huntingtin (HTT) gene. Here, we examined the effects of antioxidants on 3-nitropropionic acid (3-NP; a mitochondrial complex II inhibitor)-induced mitochondrial dysfunction and cell death in STHdhQ111 striatal cells carrying homozygous mutant HTT with extended CAG repeats compared with those in STHdhQ7 striatal cells. 3-NP reduced cell viability and increased cell death both in STHdhQ111 and STHdhQ7, and the cytotoxicity was markedly attenuated by antioxidants (N-acetyl-l-cysteine and edaravone). Furthermore, 3-NP increased intracellular reactive oxygen species (ROS) production in both cell lines, and this increase was inhibited by antioxidants. Mitochondrial ROS was also increased by 3-NP in STHdhQ111 but not in STHdhQ7, and this increase was significantly inhibited by edaravone. Mitochondrial membrane potential (MMP) was lower in STHdhQ111 than that in STHdhQ7, and antioxidants prevented 3-NP-induced MMP decrease in STHdhQ111.3-NP enhanced oligomerization of dynamin-related protein 1 (Drp1), a protein that promotes mitochondrial fission in both cells, and both antioxidants prevented the increase in oligomerization. These results suggest that reduced mitochondrial complex II activity enhances cell death via intracellular ROS production and Drp1 oligomerization in striatal cells with mutant HTT and antioxidants may reduce striatal cell death.


Asunto(s)
Antioxidantes/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Complejo II de Transporte de Electrones/metabolismo , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mutación/genética , Especies Reactivas de Oxígeno/metabolismo , Expansión de Repetición de Trinucleótido/genética , Animales , Edaravona/farmacología , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Transgénicos , Dinámicas Mitocondriales/efectos de los fármacos , Nitrocompuestos/efectos adversos , Nitrocompuestos/antagonistas & inhibidores , Propionatos/efectos adversos , Propionatos/antagonistas & inhibidores
3.
Biochim Biophys Acta Proteins Proteom ; 1869(9): 140679, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34089891

RESUMEN

The quinone binding site (Q-site) of Mitochondrial Complex II (succinate-ubiquinone oxidoreductase) is the target for a number of inhibitors useful for elucidating the mechanism of the enzyme. Some of these have been developed as fungicides or pesticides, and species-specific Q-site inhibitors may be useful against human pathogens. We report structures of chicken Complex II with six different Q-site inhibitors bound, at resolutions 2.0-2.4 Å. These structures show the common interactions between the inhibitors and their binding site. In every case a carbonyl or hydroxyl oxygen of the inhibitor is H-bonded to Tyr58 in subunit SdhD and Trp173 in subunit SdhB. Two of the inhibitors H-bond Ser39 in subunit SdhC directly, while two others do so via a water molecule. There is a distinct cavity that accepts the 2-substituent of the carboxylate ring in flutolanil and related inhibitors. A hydrophobic "tail pocket" opens to receive a side-chain of intermediate-length inhibitors. Shorter inhibitors fit entirely within the main binding cleft, while the long hydrophobic side chains of ferulenol and atpenin A5 protrude out of the cleft into the bulk lipid region, as presumably does that of ubiquinone. Comparison of mitochondrial and Escherichia coli Complex II shows a rotation of the membrane-anchor subunits by 7° relative to the iron­sulfur protein. This rotation alters the geometry of the Q-site and the H-bonding pattern of SdhB:His216 and SdhD:Asp57. This conformational difference, rather than any active-site mutation, may be responsible for the different inhibitor sensitivity of the bacterial enzyme.


Asunto(s)
Complejo II de Transporte de Electrones/antagonistas & inhibidores , Complejo II de Transporte de Electrones/ultraestructura , Ubiquinona/ultraestructura , Secuencia de Aminoácidos/genética , Animales , Benzoquinonas , Sitios de Unión , Pollos/genética , Complejo II de Transporte de Electrones/metabolismo , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Mutagénesis Sitio-Dirigida , Quinonas/química , Alineación de Secuencia , Sus scrofa/genética , Ubiquinona/química
4.
J Biol Chem ; 296: 100515, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33676890

RESUMEN

Heat-modified citrus pectin, a water-soluble indigestible polysaccharide fiber derived from citrus fruits and modified by temperature treatment, has been reported to exhibit anticancer effects. However, the bioactive fractions and their mechanisms remain unclear. In this current study, we isolated an active compound, trans-4,5-dihydroxy-2-cyclopentene-l-one (DHCP), from heat-treated citrus pectin, and found that is induces cell death in colon cancer cells via induction of mitochondrial ROS. On the molecular level, DHCP triggers ROS production by inhibiting the activity of succinate ubiquinone reductase (SQR) in mitochondrial complex II. Furthermore, cytotoxicity, apoptotic activity, and activation of caspase cascades were determined in HCT116 and HT-29 cell-based systems, the results indicated that DHCP enhances the sensitivity of cancer cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), with DHCP-induced ROS accounting for the synergistic effect between DHCP and TRAIL. Furthermore, the combination of DHCP and TRAIL inhibits the growth of HCT116 and HT-29 xenografts synergistically. ROS significantly increases the expression of TRAIL death receptor 5 (DR5) via the p53 and C/EBP homologous protein pathways. Collectively, our findings indicate that DHCP has a favorable toxicity profile and is a new TRAIL sensitizer that shows promise in the development of pectin-based pharmaceuticals, nutraceuticals, and dietary agents aimed at combating human colon cancer.


Asunto(s)
Citrus/química , Neoplasias del Colon/tratamiento farmacológico , Ciclopentanos/farmacología , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Ciclo Celular , Proliferación Celular , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
BMC Cardiovasc Disord ; 21(1): 142, 2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33731005

RESUMEN

BACKGROUND: The aim of the present study was to investigate whether the disappearance of ischemic post-processing (IPO) in the myocardium of diabetes mellitus (DM) is associated with the increase of succinate dehydrogenase-flavin protein (SDHA). METHODS: A total of 50 Sprague Dawley rats, weighing 300-400 g, were divided into 5 groups according to the random number table method, each with 10 rats. After DM rats were fed a high-fat and -sugar diet for 4 weeks, they were injected with Streptozotocin to establish the diabetic rat model. Normal rats were fed the same regular diet for the same number of weeks. Next, the above rats were taken to establish a cardiopulmonary bypass (CPB) model. Intraperitoneal glucose tolerance test (IPGTT) and oral glucose tolerance test (OGTT) were used to detect whether the DM rat model was established successfully. Taking blood from the femoral artery to collect the blood-gas analysis indicators, and judged whether the CPB model is established. After perfusion was performed according to the experimental strategy, the area of myocardial infarction (MI), and serum creatine kinase isoenzyme (CK-MB) and cardiac troponin (CTnI) levels were measured. Finally, the relative mRNA and protein expression of SDHA was detected. RESULTS: The OGTT and IPGTT suggested that the DM rat model was successfully established. The arterial blood gas analysis indicated that the CPB model was successfully established. As compared with the N group, the heart function of the IR group was significantly reduced, the levels of myocardial enzyme markers, the area of MI, as well as the relative mRNA and protein expression of SDHA, were all increased. As compared with the IR group, the CK-MB and CTnI levels in the IPO group, the MI area, relative mRNA and protein expression of SDHA decreased. As compared with the IPO group, the myocardial enzyme content in the DM + IPO group, the MI area and the relative mRNA and protein expression of SDHA increased. As compared with the DM + IPO group, in the DM + IPO + dme group, the myocardial enzyme content, area of MI and relative mRNA and protein expression were all decreased. CONCLUSION: IPO can inhibit the expression of SDHA, reduce MIRI and exert a cardioprotective effect in the normal rats. However, the protective effect of IPO disappears in the diabetic rats. The inhibitor dme combined with IPO can increase the expression of SDHA and restore the protective effect of IPO in DM myocardia.


Asunto(s)
Diabetes Mellitus Experimental/enzimología , Complejo II de Transporte de Electrones/metabolismo , Poscondicionamiento Isquémico , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/enzimología , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/complicaciones , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Complejo II de Transporte de Electrones/genética , Inhibidores Enzimáticos/farmacología , Masculino , Infarto del Miocardio/complicaciones , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Ratas Sprague-Dawley , Regulación hacia Arriba
6.
Sci Rep ; 11(1): 1483, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33452321

RESUMEN

Mitochondrial dysfunction and neurodegeneration underlie movement disorders such as Parkinson's disease, Huntington's disease and Manganism among others. As a corollary, inhibition of mitochondrial complex I (CI) and complex II (CII) by toxins 1-methyl-4-phenylpyridinium (MPP+) and 3-nitropropionic acid (3-NPA) respectively, induced degenerative changes noted in such neurodegenerative diseases. We aimed to unravel the down-stream pathways associated with CII inhibition and compared with CI inhibition and the Manganese (Mn) neurotoxicity. Genome-wide transcriptomics of N27 neuronal cells exposed to 3-NPA, compared with MPP+ and Mn revealed varied transcriptomic profile. Along with mitochondrial and synaptic pathways, Autophagy was the predominant pathway differentially regulated in the 3-NPA model with implications for neuronal survival. This pathway was unique to 3-NPA, as substantiated by in silico modelling of the three toxins. Morphological and biochemical validation of autophagy markers in the cell model of 3-NPA revealed incomplete autophagy mediated by mechanistic Target of Rapamycin Complex 2 (mTORC2) pathway. Interestingly, Brain Derived Neurotrophic Factor (BDNF), which was elevated in the 3-NPA model could confer neuroprotection against 3-NPA. We propose that, different downstream events are activated upon neurotoxin-dependent CII inhibition compared to other neurotoxins, with implications for movement disorders and regulation of autophagy could potentially offer neuroprotection.


Asunto(s)
Autofagia/fisiología , Complejo II de Transporte de Electrones/metabolismo , Neuronas/metabolismo , 1-Metil-4-fenilpiridinio/farmacología , Animales , Muerte Celular , Línea Celular , Supervivencia Celular , Células Cultivadas , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Mitocondrias/metabolismo , Trastornos del Movimiento/fisiopatología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Neuroprotección , Neurotoxinas/toxicidad , Nitrocompuestos/farmacología , Propionatos/farmacología , Ratas , Transcriptoma/genética
7.
Arch Toxicol ; 95(2): 591-615, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33512557

RESUMEN

Inhibition of complex I of the mitochondrial respiratory chain (cI) by rotenone and methyl-phenylpyridinium (MPP +) leads to the degeneration of dopaminergic neurons in man and rodents. To formally describe this mechanism of toxicity, an adverse outcome pathway (AOP:3) has been developed that implies that any inhibitor of cI, or possibly of other parts of the respiratory chain, would have the potential to trigger parkinsonian motor deficits. We used here 21 pesticides, all of which are described in the literature as mitochondrial inhibitors, to study the general applicability of AOP:3 or of in vitro assays that are assessing its activation. Five cI, three complex II (cII), and five complex III (cIII) inhibitors were characterized in detail in human dopaminergic neuronal cell cultures. The NeuriTox assay, examining neurite damage in LUHMES cells, was used as in vitro proxy of the adverse outcome (AO), i.e., of dopaminergic neurodegeneration. This test provided data on whether test compounds were unspecific cytotoxicants or specifically neurotoxic, and it yielded potency data with respect to neurite degeneration. The pesticide panel was also examined in assays for the sequential key events (KE) leading to the AO, i.e., mitochondrial respiratory chain inhibition, mitochondrial dysfunction, and disturbed proteostasis. Data from KE assays were compared to the NeuriTox data (AO). The cII-inhibitory pesticides tested here did not appear to trigger the AOP:3 at all. Some of the cI/cIII inhibitors showed a consistent AOP activation response in all assays, while others did not. In general, there was a clear hierarchy of assay sensitivity: changes of gene expression (biomarker of neuronal stress) correlated well with NeuriTox data; mitochondrial failure (measured both by a mitochondrial membrane potential-sensitive dye and a respirometric assay) was about 10-260 times more sensitive than neurite damage (AO); cI/cIII activity was sometimes affected at > 1000 times lower concentrations than the neurites. These data suggest that the use of AOP:3 for hazard assessment has a number of caveats: (i) specific parkinsonian neurodegeneration cannot be easily predicted from assays of mitochondrial dysfunction; (ii) deriving a point-of-departure for risk assessment from early KE assays may overestimate toxicant potency.


Asunto(s)
Proteínas del Complejo de Cadena de Transporte de Electrón/antagonistas & inhibidores , Transporte de Electrón/efectos de los fármacos , Inhibidores Enzimáticos/toxicidad , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Plaguicidas/toxicidad , Biomarcadores , Línea Celular , Línea Celular Tumoral , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Complejo III de Transporte de Electrones/antagonistas & inhibidores , Humanos , Proteostasis/efectos de los fármacos , Medición de Riesgo , Transcriptoma
8.
Pflugers Arch ; 472(12): 1743-1755, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32940784

RESUMEN

Nitric oxide (NO) affects mitochondrial activity through its interactions with complexes. Here, we investigated regulations of complex I (C-I) and complex II (C-II) by neuronal NO synthase (nNOS) in the presence of fatty acid supplementation and the impact on left ventricular (LV) mitochondrial activity from sham and angiotensin II (Ang-II)-induced hypertensive (HTN) rats. Our results showed that nNOS protein was expressed in sham and HTN LV mitochondrial enriched fraction. In sham, oxygen consumption rate (OCR) and intracellular ATP were increased by palmitic acid (PA) or palmitoyl-carnitine (PC). nNOS inhibitor, S-methyl-l-thiocitrulline (SMTC), did not affect OCR or cellular ATP increment by PA or PC. However, SMTC increased OCR with PA + malonate (a C-II inhibitor), but not with PA + rotenone (a C-I inhibitor), indicating that nNOS attenuates C-I with fatty acid supplementation. Indeed, SMTC increased C-I activity but not that of C-II. Conversely, nNOS-derived NO was increased by rotenone + PA in LV myocytes. In HTN, PC increased the activity of C-I but reduced that of C-II, consequently OCR was reduced. SMTC increased both C-I and C-II activities with PC, resulted in OCR enhancement in the mitochondria. Notably, SMTC increased OCR only with rotenone, suggesting that nNOS modulates C-II-mediated OCR in HTN. nNOS-derived NO was partially reduced by malonate + PA. Taken together, nNOS attenuates C-I-mediated mitochondrial OCR in the presence of fatty acid in sham and C-I modulates nNOS activity. In HTN, nNOS attenuates C-I and C-II activities whereas interactions between nNOS and C-II maintain mitochondrial activity.


Asunto(s)
Complejo II de Transporte de Electrones/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Hipertensión/metabolismo , Mitocondrias Cardíacas/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Angiotensina II/toxicidad , Animales , Células Cultivadas , Citrulina/análogos & derivados , Citrulina/farmacología , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Hipertensión/etiología , Hipertensión/fisiopatología , Masculino , Malonatos/farmacología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Consumo de Oxígeno , Ratas , Ratas Sprague-Dawley , Rotenona/farmacología , Tiourea/análogos & derivados , Tiourea/farmacología
9.
Neurochem Res ; 45(10): 2487-2498, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32789797

RESUMEN

Obesity is characterized by chronic inflammation of low grade. The cholinergic anti-inflammatory pathway favors the reduction of the inflammatory response. In this work the effect of stimulation of the cholinergic anti-inflammatory pathway on SHIRPA behavioral test and mitochondrial respiratory chain activity in obese mice was evaluated. The animals were paired in four groups: saline + control diet; donepezil + control diet; saline + high-fat diet and donepezil + high-fat diet. 5 mg/kg/day orally of donepezil or saline were given 7 days before the beginning of the diet until completing 11 weeks of the experiment. Food intake and body weight were measured. At the end of the experiment the animals were submitted to the SHIRPA behavioral test, soon after they were killed by decapitation, the open abdominal cavity and the mesenteric fat were removed. The hypothalamus, hippocampus, prefrontal cortex, and striatum were removed for evaluation of the mitochondrial respiratory chain. It can be observed that donepezil prevented weight gain and food consumption, as well as a tendency to prevent the accumulation of mesenteric fat in obese animals. There was no behavioral change in obese animals, nor did the influence of donepezil on these parameters. On the other hand, donepezil did not prevent inhibition of complex I activity, prevented the inhibition of complex II, and showed a tendency to prevent IV complex activity inhibited in obesity. With these results it can be concluded that the activation of the cholinergic anti-inflammatory pathway is promising for the alterations found in obesity.


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Encéfalo/metabolismo , Donepezilo/uso terapéutico , Metabolismo Energético/efectos de los fármacos , Obesidad/prevención & control , Animales , Peso Corporal/efectos de los fármacos , Dieta Alta en Grasa , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Complejo II de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Complejo IV de Transporte de Electrones/metabolismo , Masculino , Ratones , Obesidad/metabolismo
10.
Biochimie ; 176: 150-157, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32721502

RESUMEN

Itaconic acid (methylene-succinic acid, ItA) is an unsaturated dicarboxylic acid that is secreted by mammalian macrophages in response to a pro-inflammatory stimulus and shows an anti-inflammatory/antibacterial effect. Being a mitochondrial metabolite, it exhibits an inhibitory activity on succinate dehydrogenase and subsequently induces mitochondrial dysfunction. The present study has shown that ItA dose-dependently inhibited ADP- and DNP-stimulated (uncoupled) respiration of rat liver mitochondria energized with succinate. This effect of ItA could be related to the suppression of the activity of complex II and the combined activity of complexes II + III of the respiratory chain. At the same time, ItA had no effect on the activity of the dicarboxylate carrier, which catalyzes the transport of succinate across the inner mitochondrial membrane. It was found that 4 mM ItA diminished the rates of ADP- and DNP-stimulated mitochondrial respiration supported by the substrates of complex I glutamate and malate. A study of the effect of ItA on the activity of complexes of the respiratory chain showed that it decreases the activity of complex IV. It was observed that 4 mM ItA inhibited the rate of H2O2 production by mitochondria. At the same time, ItA promoted the opening of the cyclosporin A-sensitive Ca2+-dependent permeability transition pore. The latter was revealed as the decrease in the calcium retention capacity of mitochondria and the stimulation of release of cytochrome c from the organelles. ItA by itself promoted the cytochrome c release from mitochondria. Possible mechanisms of the effect of ItA on mitochondrial function are discussed.


Asunto(s)
Complejo II de Transporte de Electrones , Complejo IV de Transporte de Electrones , Mitocondrias Hepáticas/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Succinatos/farmacología , Animales , Calcio/metabolismo , Citocromos c/metabolismo , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Complejo II de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Complejo IV de Transporte de Electrones/metabolismo , Masculino , Poro de Transición de la Permeabilidad Mitocondrial/antagonistas & inhibidores , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Ratas , Ratas Wistar
11.
Biomed Pharmacother ; 128: 110291, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32526455

RESUMEN

The antitumor activity of atypical adamantyl retinoid ST1926 has been frequently reported in cancer studies; nevertheless, its effect on glioma has not been fully understood. Mitochondria are critical in regulating tumorigenesis and are defined as a promising target for anti-tumor therapy. In the present study, we found that ST1926 might be a mitochondria-targeting anti-glioma drug. ST1926 showed significantly inhibitory role in the viability of glioma cells mainly through inducing apoptosis and autophagy. The results showed that ST1926 alleviated mitochondria-regulated bioenergetics in glioma cells via reducing ATP production and promoting reactive oxygen species production. Importantly, ST1926 significantly impaired complex II (CII) function, which was associated with the inhibition of succinate dehydrogenase (SDH) activity. In addition, the effects of ST1926 on the induction of apoptosis and ROS were further promoted by the treatment of CII inhibitors, including TTFA and 3-NPA. Furthermore, the in vivo experiments confirmed the role of ST1926 in suppressing xenograft tumor growth with few toxicity. Therefore, ST1926 might be an effective anti-glioma drug through targeting CII.


Asunto(s)
Adamantano/análogos & derivados , Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Cinamatos/farmacología , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Metabolismo Energético/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Glioma/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Adamantano/farmacología , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Complejo II de Transporte de Electrones/metabolismo , Glioma/enzimología , Glioma/patología , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/enzimología , Mitocondrias/patología , Especies Reactivas de Oxígeno/metabolismo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Bioorg Med Chem ; 28(11): 115509, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32354673

RESUMEN

The application of a carbon-silicon bioisosteric replacement strategy to find new acaricides with improved properties led to the discovery of Sila-Cyflumetofen 6B, a novel and highly potent acaricide. The essential t-butyl group in the beta-ketonitrile acaricide Cyflumetofen 6A could be swapped with the bioisosteric trimethyl-silyl group with retention of high level acaricidal activity and favourable pharmacological properties. Sila-Cyflumetofen 6B was found to possess similar preferred energy-minimized conformation and electrostatic potential surface compare to Cyflumetofen 6A. Herein we also report the development and application of the first homology model of the spider mite mitochondrial electron transport complex II (succinate ubiquinone oxidoreductase; SQR) and demonstrated that the active metabolite AB-1 of Cyflumetofen 6A and its sila-analogue Sila-AB-1 bind to the Qp site in same binding pose and that both compounds form two H-bonds and a cation-π interaction with Trp 165, Tyr 433 and Arg 279, respectively. Furthermore, we also developed a new mode of action test for spider mite Complex II using cytochrome c as electron acceptor and blocking its re-oxidation by addition of KCN resulting in a sensitive and convenient colorimetric assay. This new method avoids the use of non-specific artificial electron acceptors and allows to measure SQR inhibition in crude extracts of Tetranychus urtice. In this assay Sila-AB-1, the intrinsically active metabolite of Sila-Cyflumetofen, 6A exhibited even a somewhat lower IC50 value than the metabolite of Cyflumetofen AB-1. Synthetic methodologies are described for the preparation of Sila-Cyflumetofen 6B and its active metabolite Sila-AB-1 which enable an efficient synthesis of these compounds in only 5 and 4 steps, respectively, from cheap commercial starting materials. Although the value of carbon-silicon bioisosteric replacements has already be demonstrated in the past it is to the best of our knowledge the first report of a successful application in crop protection research in the last two decades.


Asunto(s)
Acaricidas/farmacología , Carbono/química , Diseño de Fármacos , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Propionatos/farmacología , Silicio/química , Acaricidas/síntesis química , Acaricidas/química , Animales , Cianatos/farmacología , Relación Dosis-Respuesta a Droga , Complejo II de Transporte de Electrones/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Estructura Molecular , Propionatos/química , Relación Estructura-Actividad , Tetranychidae/enzimología
13.
Nat Commun ; 11(1): 1228, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-32144272

RESUMEN

The BCL-2 antagonist venetoclax is highly effective in multiple myeloma (MM) patients exhibiting the 11;14 translocation, the mechanistic basis of which is unknown. In evaluating cellular energetics and metabolism of t(11;14) and non-t(11;14) MM, we determine that venetoclax-sensitive myeloma has reduced mitochondrial respiration. Consistent with this, low electron transport chain (ETC) Complex I and Complex II activities correlate with venetoclax sensitivity. Inhibition of Complex I, using IACS-010759, an orally bioavailable Complex I inhibitor in clinical trials, as well as succinate ubiquinone reductase (SQR) activity of Complex II, using thenoyltrifluoroacetone (TTFA) or introduction of SDHC R72C mutant, independently sensitize resistant MM to venetoclax. We demonstrate that ETC inhibition increases BCL-2 dependence and the 'primed' state via the ATF4-BIM/NOXA axis. Further, SQR activity correlates with venetoclax sensitivity in patient samples irrespective of t(11;14) status. Use of SQR activity in a functional-biomarker informed manner may better select for MM patients responsive to venetoclax therapy.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Complejo II de Transporte de Electrones/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Transporte de Electrón/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Sulfonamidas/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Línea Celular Tumoral , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 14/genética , Resistencia a Antineoplásicos , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana/genética , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Mutación , Oxidación-Reducción/efectos de los fármacos , Selección de Paciente , Pronóstico , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Sulfonamidas/uso terapéutico , Tenoiltrifluoroacetona/farmacología , Translocación Genética
14.
Bioorg Med Chem ; 28(5): 115299, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31980361

RESUMEN

Mitochondrial complex II and complex III are two promising targets for the development of numerous pharmaceuticals and pesticides. Although tremendous inhibitors of either complex II or complex III were identified, compounds which are capable of prohibiting the activities of both complexes have been rarely reported. Since multi-target drugs can interact with several drug targets simultaneously, we were keen on discovering new and potent dual-target inhibitors of both complex II and complex III. Therefore, a new series of structurally simplified sulfonamides bearing a diaryl ether scaffold were designed and synthesized in this paper. Afterwards, the biological activities of the newly synthesized compounds were evaluated. The results implied that several compounds demonstrated outstanding potency against succinate-cytochrome c reductase (SCR, a mixture of complex II and complex III). Further studies confirmed that N-(3,5-Dichloro-4-(2,4,6-trichlorophenoxy)phenyl)benzenesulfonamide (3f), a representative compound herein, was identified as a dual-target inhibitor of both complexes. Furthermore, computational simulations were also performed to have a better understanding about binding of 3f to the enzyme complexes, which concluded that 3f should bind to complex II and the Qo site of complex III. Consequently, we harbor the idea that this work can be beneficial for the synthesis and discovery of more dual- or multi-target inhibitors.


Asunto(s)
Complejo III de Transporte de Electrones/antagonistas & inhibidores , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Mitocondrias/enzimología , Relación Dosis-Respuesta a Droga , Complejo II de Transporte de Electrones/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Estructura Molecular , Relación Estructura-Actividad
15.
Chemosphere ; 243: 125472, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31995896

RESUMEN

Repression of the electron transport in mitochondria can result in an increase of reactive oxygen species (ROS) in plant cells. This study was to clarify inhibition of the mitochondrial respiratory components (Complex I and Complex III) as stimuli to induce oxidative damage in Oryza sativa L. under exogenous SCN- exposure with special emphasis on lipid peroxidation, protein modification, and DNA damage at the biochemical and molecular levels. Our results showed that enzymatic activity and gene expression of cytochrome c reductase (Complex III) in roots and shoots of rice seedlings were significantly repressed by SCN- exposure, where significant inhibition of NADH dehydrogenase (Complex I) was only detected in shoots, suggesting that Complex III was the main target attacked by SCN- ligand in rice roots, and both components were arrested in shoots. ROS analysis in tissues indicated that SCN- exposure caused significant accumulation of H2O2 and O2-•, increased malondialdehyde (MDA) and carbonyl content in rice materials in a dose-dependent manner. Similarly, a remarkable elevation of electrolyte leakage was observed in rice tissue samples. The comet assay indicated a positive correlation between DNA damage and external SCN- exposure. In conclusion, oxidative burst generated from the inhibitions of the electron transport in mitochondria in rice seedlings under SCN- exposure can cause lipid peroxidation, protein modification and DNA damage, eventually decreasing fresh weight of rice seedlings.


Asunto(s)
Flavoproteínas Transportadoras de Electrones/antagonistas & inhibidores , Mitocondrias/química , Oryza/metabolismo , Estrés Oxidativo , Tiocianatos/farmacología , Daño del ADN , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Peroxidación de Lípido , Mitocondrias/metabolismo , Proteínas de Plantas/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Plantones/metabolismo
16.
Chem Res Toxicol ; 32(7): 1423-1431, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31251588

RESUMEN

Idiosyncratic drug reactions (IDRs) significantly increase the risk of failure in drug development. The major IDR leading to drug candidate failure is idiosyncratic drug-induced liver injury (IDILI). Although most evidence suggests that IDRs are mediated by the immune system, there are other hypotheses, such as mitochondrial dysfunction. Many pharmaceutical companies routinely screen for mitochondrial toxicity in an attempt to "derisk" drug candidates. However, the basic hypothesis has never been rigorously tested. A major assay used for this screening involves measurement of inhibition of the mitochondrial electron transport chain. One study found that the combination of rotenone and isoniazid, which inhibit mitochondrial complex I and II, respectively, were synergistic in causing hepatocyte toxicity in vitro and suggested the combination of another drug that inhibited complex I would increase the risk of isoniazid-induced liver injury in patients. We tested this hypothesis in vivo where wild-type and PD-1-/- mice administered anti-CTLA-4, our impaired immune tolerance mouse model, were given 0.02% (w/v) rotenone in water or 0.1%, 0.05%, and 0.01% (w/w) rotenone alone or in combination with isoniazid in food. The cotreatment led to lethality in 100% of the animals receiving 0.1% rotenone and 0.2% isoniazid and 83% of the animals cotreated with 0.05% rotenone and 0.2% isoniazid in food. Nevertheless, there was no significant increase in GLDH or histological evidence of liver injury. No signs of toxicity were observed in any of the mice given rotenone or isoniazid alone. Even though inhibition of the mitochondrial electron transport chain did not lead to significant liver toxicity, it could provide danger signals that promote immune-mediated liver injury. However, rotenone did not significantly increase the liver injury induced by isoniazid in our impaired immune tolerance model. Overall, we conclude that inhibition of the mitochondrial electron transport chain is not a significant mechanism of IDILI.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Transporte de Electrón/efectos de los fármacos , Inhibidores Enzimáticos/toxicidad , Isoniazida/toxicidad , Mitocondrias/efectos de los fármacos , Rotenona/toxicidad , Animales , Sinergismo Farmacológico , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Femenino , Hígado/efectos de los fármacos , Ratones Endogámicos C57BL
17.
Blood ; 134(4): 389-394, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31101624

RESUMEN

We have previously demonstrated that oxidative phosphorylation is required for the survival of human leukemia stem cells (LSCs) from patients with acute myeloid leukemia (AML). More recently, we demonstrated that LSCs in patients with de novo AML rely on amino acid metabolism to drive oxidative phosphorylation. Notably, although overall levels of amino acids contribute to LSC energy metabolism, our current findings suggest that cysteine may be of particular importance for LSC survival. We demonstrate that exogenous cysteine is metabolized exclusively to glutathione. Upon cysteine depletion, glutathione synthesis is impaired, leading to reduced glutathionylation of succinate dehydrogenase A (SDHA), a key component of electron transport chain complex (ETC) II. Loss of SDHA glutathionylation impairs ETC II activity, thereby inhibiting oxidative phosphorylation, reducing production of ATP, and leading to LSC death. Given the role of cysteine in driving LSC energy production, we tested cysteine depletion as a potential therapeutic strategy. Using a novel cysteine-degrading enzyme, we demonstrate selective eradication of LSCs, with no detectable effect on normal hematopoietic stem/progenitor cells. Together, these findings indicate that LSCs are aberrantly reliant on cysteine to sustain energy metabolism, and that targeting this axis may represent a useful therapeutic strategy.


Asunto(s)
Cisteína/metabolismo , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Leucemia Mieloide Aguda/metabolismo , Células Madre Neoplásicas/metabolismo , Adenosina Trifosfato/metabolismo , Biomarcadores , Metabolismo Energético , Glutatión/metabolismo , Humanos , Oxidación-Reducción , Fosforilación Oxidativa , Especies Reactivas de Oxígeno/metabolismo , Succinato Deshidrogenasa/metabolismo
18.
Environ Toxicol Pharmacol ; 66: 36-42, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30594848

RESUMEN

Glyphosate-based herbicides, such as Touchdown (TD) and Roundup, are among the most heavily-used herbicides in the world. While the active ingredient is generally considered non-toxic, the toxicity resulting from exposure to commercially-sold formulations is less clear. In many cases, cell cultures or various model organisms exposed to glyphosate formulations show toxicity and, in some cases, lethality. Using Caenorhabditis elegans, we assessed potential toxic mechanisms through which a highly-concentrated commercial formulation of TD promotes neurodegeneration. Following a 30-min treatment, we assayed mitochondrial electron transport chain function and reactive oxygen species (ROS) production. Initial oxygen consumption studies indicated general mitochondrial inhibition compared to controls (*p < 0.05). When Complex II activity was further assessed, inhibition was observed in all TD-treated groups (*p < 0.05). Complex IV activity, however, was not adversely affected by TD. This electron transport chain inhibition also resulted in reduced ATP levels (*p < 0.05). Furthermore, hydrogen peroxide levels, but not other ROS, were increased (*p < 0.05). Taken together, these data indicate that commercially-available formulations of TD may exert neurotoxicity through Complex II (succinate dehydrogenase) inhibition, decreased ATP levels, and increased hydrogen peroxide production.


Asunto(s)
Caenorhabditis elegans/efectos de los fármacos , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Glicina/análogos & derivados , Herbicidas/toxicidad , Adenosina Trifosfato/metabolismo , Animales , Caenorhabditis elegans/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Glicina/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Glifosato
19.
J Biol Chem ; 293(51): 19932-19941, 2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30385511

RESUMEN

We recently reported a previously unrecognized mitochondrial respiratory phenomenon. When [ADP] was held constant ("clamped") at sequentially increasing concentrations in succinate-energized muscle mitochondria in the absence of rotenone (commonly used to block complex I), we observed a biphasic, increasing then decreasing, respiratory response. Here we investigated the mechanism. We confirmed decades-old reports that oxaloacetate (OAA) inhibits succinate dehydrogenase (SDH). We then used an NMR method to assess OAA concentrations (known as difficult to measure by MS) as well as those of malate, fumarate, and citrate in isolated succinate-respiring mitochondria. When these mitochondria were incubated at varying clamped ADP concentrations, respiration increased at low [ADP] as expected given the concurrent reduction in membrane potential. With further increments in [ADP], respiration decreased associated with accumulation of OAA. Moreover, a low pyruvate concentration, that alone was not enough to drive respiration, was sufficient to metabolize OAA to citrate and completely reverse the loss of succinate-supported respiration at high [ADP]. Further, chemical or genetic inhibition of pyruvate uptake prevented OAA clearance and preserved respiration. In addition, we measured the effects of incremental [ADP] on NADH, superoxide, and H2O2 (a marker of reverse electron transport from complex II to I). In summary, our findings, taken together, support a mechanism (detailed within) wherein succinate-energized respiration as a function of increasing [ADP] is initially increased by [ADP]-dependent effects on membrane potential but subsequently decreased at higher [ADP] by inhibition of succinate dehydrogenase by OAA. The physiologic relevance is discussed.


Asunto(s)
Adenosina Difosfato/metabolismo , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ácido Oxaloacético/farmacología , Animales , Respiración de la Célula/efectos de los fármacos , Complejo II de Transporte de Electrones/metabolismo , Metabolismo Energético/efectos de los fármacos , Mitocondrias/enzimología , Células Musculares/citología , Oxígeno/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
20.
Arch Toxicol ; 92(10): 3191-3205, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30143847

RESUMEN

Propofol is the most frequently used intravenous anesthetic for induction and maintenance of anesthesia. Propofol acts first and formost as a GABAA-agonist, but effects on other neuronal receptors and voltage-gated ion channels have been described. Besides its direct effect on neurotransmission, propofol-dependent impairment of mitochondrial function in neurons has been suggested to be responsible for neurotoxicity and postoperative brain dysfunction. To clarify the potential neurotoxic effect in more detail, we investigated the effects of propofol on neuronal energy metabolism of hippocampal slices of the stratum pyramidale of area CA3 at different activity states. We combined oxygen-measurements, electrophysiology and flavin adenine dinucleotide (FAD)-imaging with computational modeling to uncover molecular targets in mitochondrial energy metabolism that are directly inhibited by propofol. We found that high concentrations of propofol (100 µM) significantly decrease population spikes, paired pulse ratio, the cerebral metabolic rate of oxygen consumption (CMRO2), frequency and power of gamma oscillations and increase FAD-oxidation. Model-based simulation of mitochondrial FAD redox state at inhibition of different respiratory chain (RC) complexes and the pyruvate-dehydrogenase show that the alterations in FAD-autofluorescence during propofol administration can be explained with a strong direct inhibition of the complex II (cxII) of the RC. While this inhibition may not affect ATP availability under normal conditions, it may have an impact at high energy demand. Our data support the notion that propofol may lead to neurotoxicity and neuronal dysfunction by directly affecting the energy metabolism in neurons.


Asunto(s)
Región CA3 Hipocampal/efectos de los fármacos , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Síndromes de Neurotoxicidad/etiología , Propofol/efectos adversos , Adenosina Trifosfato/metabolismo , Anestésicos Intravenosos/efectos adversos , Animales , Región CA3 Hipocampal/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Flavina-Adenina Dinucleótido/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Técnicas de Cultivo de Órganos , Consumo de Oxígeno/efectos de los fármacos , Ratas Wistar , Transmisión Sináptica/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...