Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Bioconjug Chem ; 33(9): 1663-1671, 2022 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-36065131

RESUMEN

Resveratrol, a natural polyphenol, exhibits beneficial health properties and has been touted as a potential anti-tumor agent. Here, we demonstrate potent anti-cancer effects of carbon dots (C-dots) synthesized from resveratrol. The mild synthesis conditions retained resveratrol functional moieties upon the carbon dots' (C-dots) surface, an important requisite for achieving specificity toward cancer cells and biological activities. Indeed, the disruptive effects of the resveratrol-C-dot were more pronounced in several cancer cell types compared to normal cells, underscoring targeting capabilities of the C-dots, a pertinent issue for the development of cancer therapeutics. In particular, we observed impairment of mitochondrial functionalities, including intracellular calcium release, inhibition of cytochrome-C oxidase enzyme activity, and mitochondrial membrane perturbation. Furthermore, the resveratrol C-dots were more potent than either resveratrol molecules alone, known anti-cancer polyphenolic agents such as curcumin and triphenylphosphonium, or C-dots prepared from different carbonaceous precursors. This study suggests that resveratrol-synthesized C-dots may have promising therapeutic potential as anti-cancer agents.


Asunto(s)
Antineoplásicos , Curcumina , Neoplasias , Estilbenos , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Apoptosis , Calcio/metabolismo , Carbono/metabolismo , Curcumina/farmacología , Complejo IV de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/farmacología , Mitocondrias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Polifenoles/metabolismo , Polifenoles/farmacología , Resveratrol/farmacología , Estilbenos/farmacología
2.
Cell Mol Biol Lett ; 27(1): 15, 2022 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-35177003

RESUMEN

BACKGROUND: Although long non-coding RNA differentiation antagonizing non-protein coding RNA (DANCR) has been reported to be involved in atherosclerosis (AS) development, its specific mechanism remains unclear. METHODS: DANCR expression levels in blood samples of AS patients and oxidized low-density lipoprotein (ox-LDL) treated vascular smooth muscle cells (VSMCs) and human umbilical vein endothelial cells (HUVECs) were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The small interfering RNA targeting DANCR (si-DANCR) was used to silence DANCR expression. Cell viability was assessed by CCK-8 assay. Cell apoptosis was evaluated by flow cytometry. Levels of inflammatory cytokines, anti-oxidative enzyme superoxide dismutase (SOD) activity, and malonaldehyde (MDA) were detected by specific commercial kits. An animal AS model was established to confirm the role of DANCR/microR-214-5p/COX20 (the chaperone of cytochrome c oxidase subunit II COX2) in AS development. RESULTS: DANCR was significantly increased in the blood samples of AS patients and ox-LDL treated VSMCs and HUVECs. DANCR downregulation obviously increased viability and reduced apoptosis of ox-LDL-treated VSMCs and HUVECs. Meanwhile, DANCR downregulation reduced the levels of inflammatory cytokines, including interleukin (IL)-6 (IL-6), IL-1beta (IL-1ß), IL-6 and tumor necrosis factor (TNF)-alpha (TNF-α) and MDA while increasing the SOD level in ox-LDL-treated VSMCs and HUVECs. DANCR regulated COX20 expression by acting as a competing endogenous RNA (ceRNA) of miR-214-5p. Rescue experiments demonstrated that miR-214-5p downregulation obviously attenuated si-DANCR-induced protective effects on ox-LDL-caused endothelial injury. CONCLUSIONS: Our results revealed that DANCR promoted AS progression by targeting the miR-214-5p/COX20 axis, suggesting that DANCR might be a potential therapeutic target for AS.


Asunto(s)
Aterosclerosis , MicroARNs , ARN Largo no Codificante , Apoptosis/genética , Aterosclerosis/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/farmacología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Lipoproteínas LDL/farmacología , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Transducción de Señal
3.
Curr Vasc Pharmacol ; 21(2): 128-142, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36924093

RESUMEN

BACKGROUND: The pathological role of cytochrome c oxidase 5A (COX5A) in vascular neointima formation remains unknown. AIM: This study aims to investigate the role of COX5A on platelet-derived growth factor BB (PDGFBB)- mediated smooth muscle phenotypic modulation and neointima formation and clarify the molecular mechanisms behind this effect. METHODS: For in vitro assays, human aortic vascular smooth muscle cells (HA-VSMCs) were transfected with pcDNA3.1-COX5A and COX5A siRNA to overexpress and knockdown COX5A, respectively. Mitochondrial complex IV activity, oxygen consumption rate (OCR), H2O2 and ATP production, reactive oxygen species (ROS) generation, cell proliferation, and migration were measured. For in vivo assays, rats after balloon injury (BI) were injected with recombinant lentivirus carrying the COX5A gene. Mitochondrial COX5A expression, carotid arterial morphology, mitochondrial ultrastructure, and ROS were measured. RESULTS: The results showed that PDGF-BB reduced the level and altered the distribution of COX5A in mitochondria, as well as reduced complex IV activity, ATP synthesis, and OCR while increasing H2O2 synthesis, ROS production, and cell proliferation and migration. These effects were reversed by overexpression of COX5A and aggravated by COX5A knockdown. In addition, COX5A overexpression attenuated BI-induced neointima formation, muscle fiber area ratio, VSMC migration to the intima, mitochondrial ultrastructural damage, and vascular ROS generation. CONCLUSION: The present study demonstrated that COX5A protects VSMCs against phenotypic modulation by improving mitochondrial respiratory function and attenuating mitochondrial damage, as well as reducing oxidative stress, thereby preventing neointima formation.


Asunto(s)
Enfermedades Mitocondriales , Neointima , Humanos , Ratas , Animales , Neointima/metabolismo , Neointima/patología , Complejo IV de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/farmacología , Músculo Liso Vascular , Especies Reactivas de Oxígeno/metabolismo , Peróxido de Hidrógeno/toxicidad , Peróxido de Hidrógeno/metabolismo , Células Cultivadas , Becaplermina/metabolismo , Becaplermina/farmacología , Proliferación Celular , Estrés Oxidativo , Miocitos del Músculo Liso , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/patología , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Movimiento Celular/fisiología
4.
Biol Trace Elem Res ; 200(12): 5022-5041, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35149956

RESUMEN

An increase in anthropogenic activities results in metal contamination in the ecosystem which has proven to be a major health risk in humans, as they make entry into cellular organelles via agricultural products. Copper (Cu) is one such metal that acts as an essential cofactor for the activity of several enzymes, one being the cytochrome c oxidase. The increasing number of evidence suggests a substantial correlation of Cu overload with neurodegenerative disorders, including Parkinson's disease (PD). We aim to explore quercetin, a well-known polyphenol, as an alternative for combating Cu-induced toxicity in human neuroblastoma SH-SY5Y secondary cell lines. We observed that Cu increased intracellular reactive oxygen species (ROS) levels, triggered morphological deformities and condensation of nuclei, caused an imbalance in the mitochondrial membrane potential (MMP), and finally induced apoptotic cell deaths. We further investigated the effects of Cu in modulating the pro- and anti-apoptotic proteins, such as Bax, Bcl-2, etc. However, quercetin reversed these changes owing to its antioxidant and anti-apoptotic properties, resulting in autophagy induction as an outcome of upregulation of autophagosome-bound microtubules-associated protein light chain-3 (LC3II). Besides, we investigated the role of Cu in stimulating ER stress proteins, viz. PERK, CHOP, and the concomitant responses of quercetin in restoring the ER homeostasis in cellular organelles like mitochondria and ER, against Cu-induced toxic insults by modulating autophagic pathways. Overall, this research work proposes a remedial approach for Cu-mediated neurotoxicity through understanding the diverse molecular signaling inside a cell with an aim to develop effective therapeutics.


Asunto(s)
Estrés del Retículo Endoplásmico , Neuroblastoma , Antioxidantes/farmacología , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia , Línea Celular Tumoral , Cobre/metabolismo , Cobre/farmacología , Ecosistema , Complejo IV de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/farmacología , Proteínas de Choque Térmico/metabolismo , Humanos , Polifenoles/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Quercetina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Proteína X Asociada a bcl-2/metabolismo
5.
Autophagy ; 18(10): 2303-2322, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34964695

RESUMEN

By promoting anabolism, MTORC1 is critical for muscle growth and maintenance. However, genetic MTORC1 upregulation promotes muscle aging and produces age-associated myopathy. Whether MTORC1 activation is sufficient to produce myopathy or indirectly promotes it by accelerating tissue aging is elusive. Here we examined the effects of muscular MTORC1 hyperactivation, produced by simultaneous depletion of TSC1 and DEPDC5 (CKM-TD). CKM-TD mice produced myopathy, associated with loss of skeletal muscle mass and force, as well as cardiac failure and bradypnea. These pathologies were manifested at eight weeks of age, leading to a highly penetrant fatality at around twelve weeks of age. Transcriptome analysis indicated that genes mediating proteasomal and macroautophagic/autophagic pathways were highly upregulated in CKM-TD skeletal muscle, in addition to inflammation, oxidative stress, and DNA damage signaling pathways. In CKM-TD muscle, autophagosome levels were increased, and the AMPK and ULK1 pathways were activated; in addition, autophagy induction was not completely blocked in CKM-TD myotubes. Despite the upregulation of autolysosomal markers, CKM-TD myofibers exhibited accumulation of autophagy substrates, such as SQSTM1/p62 and ubiquitinated proteins, suggesting that the autophagic activities were insufficient. Administration of a superoxide scavenger, tempol, normalized most of these molecular pathologies and subsequently restored muscle histology and force generation. However, CKM-TD autophagy alterations were not normalized by rapamycin or tempol, suggesting that they may involve non-canonical targets other than MTORC1. These results collectively indicate that the concomitant muscle deficiency of TSC1 and DEPDC5 can produce early-onset myopathy through accumulation of oxidative stress, which dysregulates myocellular homeostasis.Abbreviations: AMPK: AMP-activated protein kinase; CKM: creatine kinase, M-type; COX: cytochrome oxidase; DEPDC5: DEP domain containing 5, GATOR1 subcomplex subunit; DHE: dihydroethidium; EDL: extensor digitorum longus; EIF4EBP1: eukaryotic translation initiation factor 4E binding protein 1; GAP: GTPase-activating protein; GTN: gastrocnemius; MTORC1: mechanistic target of rapamycin kinase complex 1; PLA: plantaris; QUAD: quadriceps; RPS6KB/S6K: ribosomal protein S6 kinase beta; SDH: succinate dehydrogenase; SOL: soleus; SQSTM1: sequestosome 1; TA: tibialis anterior; TSC1: TSC complex subunit 1; ULK1: unc-51 like autophagy activating kinase 1.


Asunto(s)
Cardiopatías , Enfermedades Musculares , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Autofagia , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Forma MM de la Creatina-Quinasa/metabolismo , Óxidos N-Cíclicos , Complejo IV de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/farmacología , Proteínas Activadoras de GTPasa/metabolismo , Cardiopatías/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Enfermedades Musculares/metabolismo , Miocardio/metabolismo , Estrés Oxidativo , Factores de Iniciación de Péptidos/metabolismo , Poliésteres/metabolismo , Poliésteres/farmacología , Proteínas Quinasas S6 Ribosómicas/metabolismo , Proteínas Quinasas S6 Ribosómicas/farmacología , Proteína Sequestosoma-1/metabolismo , Sirolimus/farmacología , Marcadores de Spin , Succinato Deshidrogenasa/metabolismo , Succinato Deshidrogenasa/farmacología , Superóxidos/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Ubiquitinadas/metabolismo
6.
Microbiology (Reading) ; 157(Pt 3): 685-694, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21148204

RESUMEN

The widely used drug diclofenac can cause serious heart, liver and kidney injury, which may be related to its ability to cause mitochondrial dysfunction. Using Saccharomyces cerevisiae as a model system, we studied the mechanisms of diclofenac toxicity and the role of mitochondria therein. We found that diclofenac reduced cell growth and viability and increased levels of reactive oxygen species (ROS). Strains increasingly relying on respiration for their energy production showed enhanced sensitivity to diclofenac. Furthermore, oxygen consumption was inhibited by diclofenac, suggesting that the drug inhibits respiration. To identify the site of respiratory inhibition, we investigated the effects of deletion of respiratory chain subunits on diclofenac toxicity. Whereas deletion of most subunits had no effect, loss of either Rip1p of complex III or Cox9p of complex IV resulted in enhanced resistance to diclofenac. In these deletion strains, diclofenac did not increase ROS formation as severely as in the wild-type. Our data are consistent with a mechanism of toxicity in which diclofenac inhibits respiration by interfering with Rip1p and Cox9p in the respiratory chain, resulting in ROS production that causes cell death.


Asunto(s)
Antiinflamatorios no Esteroideos/toxicidad , Diclofenaco/toxicidad , Complejo IV de Transporte de Electrones/metabolismo , Mitocondrias/efectos de los fármacos , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Antiinflamatorios no Esteroideos/metabolismo , Diclofenaco/metabolismo , Transporte de Electrón/fisiología , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/farmacología , Membranas Mitocondriales/metabolismo , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/farmacología , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Especies Reactivas de Oxígeno , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/ultraestructura , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/farmacología
7.
Autophagy ; 17(10): 3221-3237, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33315519

RESUMEN

Liver dysfunction is an outstanding dose-limiting toxicity of gefitinib, an EGFR (epidermal growth factor receptor)-tyrosine kinase inhibitor (TKI), in the treatment of EGFR mutation-positive non-small cell lung cancer (NSCLC). We aimed to elucidate the mechanisms underlying gefitinib-induced hepatotoxicity, and provide potentially effective intervention strategy. We discovered that gefitinib could sequentially activate macroautophagy/autophagy and apoptosis in hepatocytes. The inhibition of autophagy alleviated gefitinib-induced apoptosis, whereas the suppression of apoptosis failed to lessen gefitinib-induced autophagy. Moreover, liver-specific Atg7+/- heterozygous mice showed less severe liver injury than vehicle, suggesting that autophagy is involved in the gefitinib-promoted hepatotoxicity. Mechanistically, gefitinib selectively degrades the important anti-apoptosis factor COX6A1 (cytochrome c oxidase subunit 6A1) in the autophagy-lysosome pathway. The gefitinib-induced COX6A1 reduction impairs mitochondrial respiratory chain complex IV (RCC IV) function, which in turn activates apoptosis, hence causing liver injury. Notably, this autophagy-promoted apoptosis is dependent on PLK1 (polo like kinase 1). Both AAV8-mediated Plk1 knockdown and PLK1 inhibitor BI-2536 could mitigate the gefitinib-induced hepatotoxicity in vivo by abrogating the autophagic degradation of the COX6A1 protein. In addition, PLK1 inhibition could not compromise the anti-cancer activity of gefitinib. In conclusion, our findings reveal the gefitinib-hepatotoxicity pathway, wherein autophagy promotes apoptosis through COX6A1 degradation, and highlight pharmacological inhibition of PLK1 as an attractive therapeutic approach toward improving the safety of gefitinib-based cancer therapy.Abbreviations: 3-MA: 3-methyladenine; AAV8: adeno-associated virus serotype 8; ATG5: autophagy related 5; ATG7: autophagy related 7; B2M: beta-2-microglobulin; CCCP: carbonyl cyanide m-chlorophenylhydrazone; CHX: cycloheximide; COX6A1: cytochrome c oxidase subunit 6A1; c-PARP: cleaved poly(ADP-ribose) polymerase; CQ: chloroquine; GOT1/AST: glutamic-oxaloacetic transaminase 1, soluble; GPT/ALT: glutamic pyruvic transaminase, soluble; HBSS: Hanks´ balanced salt solution; H&E: hematoxylin and eosin; MAP1LC3/LC3: microtubule associated proteins 1 light chain 3; PLK1: polo like kinase 1; RCC IV: respiratory chain complex IV; ROS: reactive oxygen species; TUBB8: tubulin beta 8 class VIII.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Enfermedad Hepática Inducida por Sustancias y Drogas , Neoplasias Pulmonares , Animales , Apoptosis , Autofagia , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proteínas de Ciclo Celular , Complejo IV de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/farmacología , Gefitinib/farmacología , Neoplasias Pulmonares/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas , Quinasa Tipo Polo 1
8.
J Exp Med ; 155(1): 264-75, 1982 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-6274994

RESUMEN

Mitochondria synthesize several hydrophobic proteins. Like bacteria, mitochondria initiate protein synthesis with an N-formylmethionine residue. Because N-formylmethionyl peptides have been found to be chemotactic for polymorphonuclear leukocytes (PMN), mitochondria isolated from cultured human cells and purified bovine mitochondrial proteins were tested for PMN chemotactic activity in vitro. Nondisrupted mitochondria were not chemotactic. However, intact mitochondria that had been incubated with a lysosomal lysate did stimulate PMN migration. Antibodies directed against two mitochondrial enzymes, cytochrome oxidase and ATPase, (both of which contain mitochondrially synthesized subunits) but not anti-C3 or anti-C5 decreased mitochondrially derived chemotactic activity. In addition, purified bovine mitochondrial N-formylmethionyl proteins stimulated PMN migration in vitro, whereas nonformylated mitochondrial proteins did not. Furthermore, the chemotactic activity of purified mitochondrial proteins and disrupted mitochondria was decreased by the formyl peptide antagonist butyloxycarbonyl-phenylalanine-leucine-phenylalanine-leucine-phenylalanine. Finally, disrupted mitochondria and purified mitochondrial proteins stimulated PMN-directed migration (chemotaxis), according to accepted criteria. In addition to other chemotactic factors, release of N-formylmethionyl proteins from mitochondria at sites of tissue damage, may play a role in the accumulation of inflammatory cells at these sites.


Asunto(s)
Quimiotaxis de Leucocito/efectos de los fármacos , Metionina/análogos & derivados , Mitocondrias/metabolismo , N-Formilmetionina/farmacología , Adenosina Trifosfatasas/farmacología , Complejo IV de Transporte de Electrones/farmacología , Fibroblastos/metabolismo , Células HeLa/metabolismo , Humanos , Neutrófilos , Oligopéptidos/farmacología
9.
Mol Med Rep ; 19(6): 4852-4862, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31059068

RESUMEN

Cerebrovascular disease (CVD) is one of the leading causes of mortality worldwide. The role of cytochrome c oxidase subunit 6B1 (COX6B1) in the central nervous system remains unclear. The present study aimed to analyze the role of COX6B1 in rat hippocampal neurons extracted from fetal rats. The subcellular localization of the neuron­specific marker microtubule­associated protein 2 was detected by immunofluorescence assay. Cell viability was assessed using a cell counting kit, and the levels of apoptosis and cytosolic Ca2+ were analyzed by flow cytometry. The expression levels of the molecular factors downstream to COX6B1 were determined using reverse transcription­quantitative polymerase chain reaction and western blotting. Reoxygenation following oxygen­glucose deprivation (OGD) decreased cell viability and the expression levels of COX6B1 in a time­dependent manner, and 60 min of reoxygenation was identified as the optimal time period for establishing an ischemia/reperfusion (I/R) model. Overexpression of COX6B1 was demonstrated to reverse the viability of hippocampal neurons following I/R treatment. Specifically, COX6B1 overexpression decreased the cytosolic concentration of Ca2+ and suppressed neuronal apoptosis, which were increased following I/R treatment. Furthermore, overexpression of COX6B1 increased the protein expression levels of apoptosis regulator BCL­2 and mitochondrial cytochrome c (cyt c), and decreased the protein expression levels of apoptosis regulator BCL2­associated X and cytosolic cyt c in I/R model cells. Collectively, the present study results suggested that COX6B1 overexpression may reverse I/R­induced neuronal damage by increasing the viability of neurons, by decreasing the cytosolic levels of Ca2+ and by suppressing apoptosis. These results may facilitate the development of novel strategies for the prevention and treatment of CVD.


Asunto(s)
Complejo IV de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/farmacología , Neuronas/metabolismo , Daño por Reperfusión/prevención & control , Lóbulo Temporal/metabolismo , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Trastornos Cerebrovasculares/prevención & control , Citocromos c/metabolismo , Complejo IV de Transporte de Electrones/genética , Femenino , Glucosa/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neuronas/patología , Oxígeno/metabolismo , Sustancias Protectoras/metabolismo , Sustancias Protectoras/farmacología , Ratas , Ratas Sprague-Dawley , Lóbulo Temporal/patología , Proteína bcl-X/metabolismo
10.
FEBS Lett ; 359(1): 93-6, 1995 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-7851539

RESUMEN

We have studied the interactions between various synthetic peptides and two model unfolded proteins, reduced alpha-lactalbumin and reduced and carboxymethylated alpha-lactalbumin. We found that mitochondrial presequences could induce aggregation of the unfolded alpha-lactalbumins but not of the native alpha-lactalbumin. The presequence-induced aggregation of unfolded alpha-lactalbumin was dependent on electrostatic interactions and on the amphiphilicity of the presequences. Since positive charge and amphiphilicity are necessary for the targeting function of mitochondrial presequences, presequence-induced aggregation may be responsible for the instability of mitochondrial precursor proteins and may need to be inhibited by binding factors in the cytosol.


Asunto(s)
Lactalbúmina/química , Mitocondrias/química , Pliegue de Proteína , Precursores de Proteínas/química , Secuencia de Aminoácidos , Animales , Bovinos , Electroquímica , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/farmacología , Cinética , Sustancias Macromoleculares , Metilación , Datos de Secuencia Molecular , Oxidación-Reducción , Fragmentos de Péptidos/farmacología , Saccharomyces cerevisiae/enzimología
11.
Brain Res ; 777(1-2): 103-9, 1997 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-9449418

RESUMEN

We previously reported the existence of motilin receptors in the cerebellum of the rabbit. We now explored the existence of motilin receptors in other brain regions and determined their association with neurons by subcellular fractionation studies. Autoradiographic studies with [(125)I]nle13-porcine motilin on rabbit coronal brain sections revealed specific binding sites in the hippocampus, thalamus, hypothalamus and amygdaloid body. Receptor binding studies allowed the identification of two binding sites. In all regions the density of the high-affinity binding site was lower than in the cerebellum, but its affinity was the same, except for the hypothalamus. No differences were found for affinity or density of the low-affinity receptor site. Homogenates of rabbit cerebellum were subjected to differential centrifugation. The highest motilin binding (10-times more than in the postnuclear supernatant) was found in the fraction which also showed maximal enrichment of [11-3H]saxitoxin binding (selective marker for voltage-sensitive Na+ channels), 6.9-fold, and cytochrome c oxidase activity (mitochondrial marker), 2.4-fold. In discontinuous sucrose density gradient centrifugation the motilin and saxitoxin binding both peaked in the 0.85-1 M layer, while cytochrome c oxidase was maximal in the 1.2 M layer. In conclusion, motilin receptors exist in several regions of the rabbit brain and are probably associated with synaptosomes. These findings further support a neurotransmitter role for motilin in the brain.


Asunto(s)
Química Encefálica , Motilina/farmacología , Receptores de Superficie Celular/análisis , 5'-Nucleotidasa/farmacología , Animales , Autorradiografía , Membrana Celular/química , Cerebelo/química , Complejo IV de Transporte de Electrones/farmacología , Radioisótopos de Yodo , Motilina/metabolismo , Prosencéfalo/química , Conejos , Ensayo de Unión Radioligante , Saxitoxina/metabolismo , Saxitoxina/farmacología , Fracciones Subcelulares/química , Sinaptosomas/química , Tritio
12.
Life Sci ; 53(26): 1975-80, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8255160

RESUMEN

Using absorption spectroscopy we have found that metallothionein I at concentrations as low as 1 microM reduces ferricytochrome c. This reduction is more potent than that of another abundant intracellular thiol, glutathione. Also, oxidation of ferricytochrome c which had been reduced by metallothionein I occurs more slowly than when the reductant was glutathione. Over a pH range of 6 through 9 there is no variation in the initial rate of metallothionein I reduction of ferricytochrome c. These data demonstrate a potent interaction between metallothionein I and ferricytochrome c.


Asunto(s)
Grupo Citocromo c/metabolismo , Metalotioneína/farmacología , Animales , Complejo IV de Transporte de Electrones/farmacología , Cinética , Hígado/química , Oxidación-Reducción , Conejos , Espectrofotometría/métodos
13.
Sci Total Environ ; 290(1-3): 121-9, 2002 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-12083703

RESUMEN

Since the mid-1980s, a 'mysterious' wasting disease has been afflicting the moose (Alces alces L.) population of south-western Sweden. In 1994, molybdenosis combined with copper deficiency was suggested as the cause of this complex syndrome of clinical signs, diversity of necropsy findings and changes in trace element concentrations. These findings were corroborated by scientists in many countries by similar observations in other ruminants, particularly cattle and sheep, and also by changes in trace element concentrations and clinical chemical findings in our model experiments with goats. The biochemistry of copper is dependent on a number of copper-dependent enzymes in the animal organism. An important example is cytochrome c oxidase (COX), responsible for oxidative phosphorylation and energy production within the cell. In the present study, COX activity and trace element concentrations were determined in myocardium from affected and healthy moose. Citrate synthase (CS) activity was also measured for use as a mitochondrial marker. COX activity had decreased by 45% and the COX/CS ratio by 37%, while Mo and Na were found to have increased by 140% and 25%, respectively. The increase in Na was indicative of the frequently reported oedematous changes in 'flabby' moose heart. The concentrations of the elements Cu, Mg, Mn, P and Zn had decreased by 20%, 20%, 35%, 7% and 19%, respectively. The simultaneous decrease in COX activity and Cu concentration and the increase in Mo further support the hypothesis that molybdenosis is the cause of the moose disease.


Asunto(s)
Ciervos , Complejo IV de Transporte de Electrones/farmacología , Molibdeno/efectos adversos , Miocardio/enzimología , Enfermedades de los Animales/etiología , Animales , Cobre/deficiencia , Edema/veterinaria , Complejo IV de Transporte de Electrones/análisis , Síndrome , Pérdida de Peso
14.
Biomed Environ Sci ; 16(3): 256-66, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-14631831

RESUMEN

OBJECTIVE: To investigate the toxicity of cigarette smoke extract (CSE) and nicotine on mouse brain mitochondria as well as the protective effect of vitamin C in vitro. METHOD: Mouse brain mitochondria in vitro was incubated with CSE or nicotine in the absence or presence of vitamin C for 60 minutes, and the changes of mitochondrial function and structure were measured. RESULTS: CSE inhibited mitochondrial ATPase and cytochrome C oxidase activities in a dose-dependent manner. However, no significant changes in the peroxidation indices were observed when mitochondrial respiratory enzymes activity was inhibited, and protection of mitochondria from CSE-induced injury by vitamin C was not displayed in vitro. The effect of CSE on mouse brain mitochondria swelling response to calcium stimulation was dependent on calcium concentrations. CSE inhibited swelling of mitochondria at 6.5 mumol/L Ca2+, but promoted swelling response at 250 mumol/L Ca2+. Nicotine, the major component of cigarette smoke, showed no significant damage in mouse brain mitochondria in vitro. The CSE treatment induced mitochondrial inner membrane damage and vacuolization of the matrix, whereas the outer mitochondrial membrane appeared to be preserved. CONCLUSION: The toxic effect of CSE on brain mitochondria may be due to its direct action on enzymatic activity rather than through oxygen free radical injury. Nicotine is not the responsible component for the toxicity of CSE to brain mitochondria.


Asunto(s)
Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Encéfalo/patología , Estimulantes Ganglionares/toxicidad , Mitocondrias/patología , Nicotina/toxicidad , Humo/efectos adversos , Adenosina Trifosfatasas/farmacología , Animales , Complejo IV de Transporte de Electrones/farmacología , Radicales Libres , Ratones , Nicotiana
15.
Water Environ Res ; 75(5): 434-43, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14587954

RESUMEN

In this study, various amounts of oxygen were added to denitrifying chemostats receiving benzoate to mimic the input of oxygen to anoxic zones of biological nutrient removal systems. The effect of oxygen on the biodegradative capability of the mixed-microbial culture for benzoate was investigated. The anoxic benzoate biodegradative capability of the culture was not significantly changed as the mass flowrate of oxygen was increased to 40% of the input benzoate chemical oxygen demand (COD) mass flowrate, but was decreased approximately 70% when the mass flowrate of oxygen was increased to 70% of the input benzoate COD mass flowrate. The decrease in the anoxic benzoate biodegradative capability was due primarily to the loss of the denitrifying enzymes (measured by the anoxic pyruvate-degrading ability) and not to the loss of the key anoxic catabolic enzyme (benzoyl-coenzyme A reductase). The proportional increase in the concentration of nitrate as the residual terminal electron acceptor and the lack of synthesis of aerobic ring-cleavage enzymes as the oxygen input to the chemostat was increased suggest that the mixed microbial culture preferred oxygen to nitrate as the terminal electron acceptor, but degraded benzoate using the anoxic metabolic pathway. The concentration of the mixed microbial culture increased as the oxygen input to the chemostat was increased, suggesting that the oxygen was used by cytochrome cbb3 rather than quinol oxidase because the energetic yield of cytochrome cbb3 is higher than that of quinol oxidase or the nitrogen oxide reductases.


Asunto(s)
Antiinfecciosos/metabolismo , Benzoatos/metabolismo , Reactores Biológicos , Oxígeno/análisis , Purificación del Agua/métodos , Bacterias/enzimología , Biodegradación Ambiental , Complejo IV de Transporte de Electrones/farmacología
16.
West Indian Med J ; 50(2): 111-6, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11677906

RESUMEN

Cytochrome c oxidase, the final member of the electron transport chain, is crucial to respiration and also contributes to the synthesis of cellular ATP. The total absence of this enzyme is incompatible with life and its deficiency or malfunction leads to a number of serious disease states. Understanding the mechanism of action of this enzyme, which is an important prerequisite to unravelling its role in the pathogenesis of disease states, is hampered by the lack of suitable enzyme models. The bovine enzyme, which is commonly used, is enormously complex and the bacterial enzymes, which are structurally simple, appear to follow a different mechanism of action. The hammer head shark is a seasonal resident of the warm waters of the Caribbean Sea. The work presented here indicates that, like the bovine enzyme, the enzyme of the heart of this shark (i) possesses thirteen subunits and two substrate binding sites and (ii) exhibits biphasic kinetics. The work also confirms that, unlike the bovine enzyme which is dimeric, the shark enzyme functions as a monomer. Given this latter simplifying feature, in conjunction with its kinetic and structural similarities to the more complex mammalian varieties, we propose that shark heart cytochrome c oxidase replace the bovine and bacterial forms as the enzyme of choice for model studies.


Asunto(s)
Complejo IV de Transporte de Electrones/fisiología , Tiburones , Animales , Bovinos , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/farmacología , Humanos , Miocardio/enzimología
17.
Folia Biol (Krakow) ; 51(3-4): 201-5, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-15303375

RESUMEN

This study examined morphochemical differences between 49 full term human placentas collected from healthy non-smoking women living in the high polluted region, i.e. the Copper Mining Territory (CMT) and the 38 control placentas (C) obtained from little polluted eastern Carpathian regions. The placentas were studied by histochemical, immunohistochemical and morphometric methods. In CMT placentas a decrease in the cytochrome c oxidase and glucose-6-phosphate activities and the immunoreactivity of glutathione S-transferase pi in the villous syncytiotrophoblast and amniotic epithelium was noted. All CMT placentas showed abundance of mineral and fibrinoid deposits and of lipid droplets. This produced a compensatory increase in the mother-fetus exchange area due to excessive proliferation of placental villi which in turn decreased the intervillous space and thus the influx of indispensable maternal blood. Lately slight signs of increase in the cytochrome c oxidase activity accompanied by a noticeable decrease in number of the thinnest (most abundant) terminal villi is observed.


Asunto(s)
Cobre/envenenamiento , Complejo IV de Transporte de Electrones/farmacología , Exposición a Riesgos Ambientales , Intercambio Materno-Fetal/efectos de los fármacos , Placenta/efectos de los fármacos , Placenta/fisiología , Adulto , Vellosidades Coriónicas/efectos de los fármacos , Vellosidades Coriónicas/patología , Complejo IV de Transporte de Electrones/análisis , Femenino , Glutatión Transferasa/análisis , Glutatión Transferasa/farmacología , Humanos , Inmunohistoquímica , Lípidos/análisis , Intercambio Materno-Fetal/fisiología , Minería , Placenta/patología , Polonia , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA