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










Intervalo de ano de publicação
1.
Curr Vasc Pharmacol ; 21(2): 128-142, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36924093

RESUMO

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.


Assuntos
Doenças Mitocondriais , Neointima , Humanos , Ratos , Animais , Neointima/metabolismo , Neointima/patologia , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Músculo Liso Vascular , Espécies Reativas de Oxigênio/metabolismo , Peróxido de Hidrogênio/toxicidade , Peróxido de Hidrogênio/metabolismo , Células Cultivadas , Becaplermina/metabolismo , Becaplermina/farmacologia , Proliferação de Células , Estresse Oxidativo , Miócitos de Músculo Liso , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/patologia , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Movimento Celular/fisiologia
2.
Bioconjug Chem ; 33(9): 1663-1671, 2022 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-36065131

RESUMO

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.


Assuntos
Antineoplásicos , Curcumina , Neoplasias , Estilbenos , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Apoptose , Cálcio/metabolismo , Carbono/metabolismo , Curcumina/farmacologia , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Mitocôndrias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Polifenóis/metabolismo , Polifenóis/farmacologia , Resveratrol/farmacologia , Estilbenos/farmacologia
3.
Cell Mol Biol Lett ; 27(1): 15, 2022 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-35177003

RESUMO

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.


Assuntos
Aterosclerose , MicroRNAs , RNA Longo não Codificante , Apoptose/genética , Aterosclerose/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Lipoproteínas LDL/farmacologia , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Transdução de Sinais
4.
Biol Trace Elem Res ; 200(12): 5022-5041, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35149956

RESUMO

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.


Assuntos
Estresse do Retículo Endoplasmático , Neuroblastoma , Antioxidantes/farmacologia , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia , Linhagem Celular Tumoral , Cobre/metabolismo , Cobre/farmacologia , Ecossistema , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Proteínas de Choque Térmico/metabolismo , Humanos , Polifenóis/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Quercetina/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Proteína X Associada a bcl-2/metabolismo
5.
Autophagy ; 18(10): 2303-2322, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34964695

RESUMO

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.


Assuntos
Cardiopatias , Doenças Musculares , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Autofagia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Creatina Quinase Forma MM/metabolismo , Óxidos N-Cíclicos , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Proteínas Ativadoras de GTPase/metabolismo , Cardiopatias/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Doenças Musculares/metabolismo , Miocárdio/metabolismo , Estresse Oxidativo , Fatores de Iniciação de Peptídeos/metabolismo , Poliésteres/metabolismo , Poliésteres/farmacologia , Proteínas Quinases S6 Ribossômicas/metabolismo , Proteínas Quinases S6 Ribossômicas/farmacologia , Proteína Sequestossoma-1/metabolismo , Sirolimo/farmacologia , Marcadores de Spin , Succinato Desidrogenase/metabolismo , Succinato Desidrogenase/farmacologia , Superóxidos/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Ubiquitinadas/metabolismo
6.
Autophagy ; 17(10): 3221-3237, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33315519

RESUMO

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.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Doença Hepática Induzida por Substâncias e Drogas , Neoplasias Pulmonares , Animais , Apoptose , Autofagia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proteínas de Ciclo Celular , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Gefitinibe/farmacologia , Neoplasias Pulmonares/metabolismo , Camundongos , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas
7.
Mol Med Rep ; 19(6): 4852-4862, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31059068

RESUMO

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.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Neurônios/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Lobo Temporal/metabolismo , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Transtornos Cerebrovasculares/prevenção & controle , Citocromos c/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/genética , Feminino , Glucose/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neurônios/patologia , Oxigênio/metabolismo , Substâncias Protetoras/metabolismo , Substâncias Protetoras/farmacologia , Ratos , Ratos Sprague-Dawley , Lobo Temporal/patologia , Proteína bcl-X/metabolismo
8.
Microbiology (Reading) ; 157(Pt 3): 685-694, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21148204

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/toxicidade , Diclofenaco/toxicidade , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Mitocôndrias/efeitos dos fármacos , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Anti-Inflamatórios não Esteroides/metabolismo , Diclofenaco/metabolismo , Transporte de Elétrons/fisiologia , Complexo IV da Cadeia de Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Membranas Mitocondriais/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/farmacologia , Consumo de Oxigênio/efeitos dos fármacos , Consumo de Oxigênio/fisiologia , Espécies Reativas de Oxigênio , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/fisiologia , Saccharomyces cerevisiae/ultraestrutura , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/farmacologia
9.
J Biol Chem ; 283(45): 30596-605, 2008 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-18768476

RESUMO

Although neutrophils are known to migrate in response to various chemokines and complement factors, the substances involved in the early stages of their transmigration and activation have been poorly characterized to date. Here we report the discovery of a peptide isolated from healthy porcine hearts that activated neutrophils. Its primary structure is H-Leu-Ser-Phe-Leu-Ile-Pro-Ala-Gly-Trp-Val-Leu-Ser-His-Leu-Asp-His-Tyr-Lys-Arg-Ser-Ser-Ala-Ala-OH, and it was indicated to originate from mitochondrial cytochrome c oxidase subunit VIII. This peptide caused chemotaxis at concentrations lower than that inducing beta-hexosaminidase release. Such responses were observed in neutrophilic/granulocytic differentiated HL-60 cells but not in undifferentiated cells, and G(i2)-type G proteins were suggested to be involved in the peptide signaling. Moreover the peptide activated human neutrophils to induce beta-hexosaminidase secretion. A number of other amphipathic neutrophil-activating peptides presumably originating from mitochondrial proteins were also found. The present results suggest that neutrophils monitor such amphipathic peptides including the identified peptide as an initiation signal for inflammation at injury sites.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/isolamento & purificação , Proteínas Mitocondriais/isolamento & purificação , Proteínas Musculares/isolamento & purificação , Miocárdio/química , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/metabolismo , Peptídeos/isolamento & purificação , Animais , Quimiotaxia/efeitos dos fármacos , Complexo IV da Cadeia de Transporte de Elétrons/química , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Células HL-60 , Humanos , Proteínas Mitocondriais/química , Proteínas Mitocondriais/farmacologia , Proteínas Musculares/química , Proteínas Musculares/farmacologia , Peptídeos/química , Peptídeos/farmacologia , Suínos , beta-N-Acetil-Hexosaminidases/metabolismo
10.
Crit Care Med ; 35(9 Suppl): S468-75, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17713395

RESUMO

Sepsis, the principal cause of death in critically ill patients, is associated with impaired oxygen extraction by tissues. One possible explanation is the development of mitochondrial dysfunction and ineffective oxygen utilization. This abnormality has been termed cytopathic hypoxia. This may be caused by an abnormality in the transport of electrons down the cytochrome chain on the mitochondrial inner membrane. In this article we review our studies on abnormalities in the function of complex IV (cytochrome oxidase), the final electron acceptor in this chain. In addition, we provide evidence that administration of cytochrome c may overcome these abnormalities and provide a novel therapeutic alternative.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Sepse/metabolismo , Animais , Estado Terminal , Citocromos c/biossíntese , Complexo IV da Cadeia de Transporte de Elétrons/administração & dosagem , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Humanos , Mitocôndrias Cardíacas/metabolismo , Relaxamento Muscular , Contração Miocárdica , Miocárdio/metabolismo , Sepse/fisiopatologia
11.
Acta Neurol Scand ; 111(5): 329-32, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15819713

RESUMO

We have investigated the dynamics of cytochrome c oxidase (COX) activity in the cerebrospinal fluid (CSF) and the erythrocyte haemolysate (EH) in 85 patients suffering from brain infarction (BI), reversible (RIA), or transient (TIA) ischemic attack from the perspective of mitochondrial affection in ischemia. In all patients, the COX activity was decreased in the CSF, especially within the first two days, indicating an acute inactivation or modification of mitochondrial proteins, probably mediated by free radicals. The gradual elevation of COX activity until the seventh day suggested that these changes may be reversible. The increase in the COX activity was established in the EH, with the highest values found in the BI, somewhat lower in the RIA, and the lowest in the TIA group, respectively. This could indicate a systemic compensatory response to an acute ischemia. Thus, COX activity in the CSF and EH in acute ischemia could be an indicator of brain metabolic dysfunction.


Assuntos
Isquemia Encefálica/enzimologia , Complexo IV da Cadeia de Transporte de Elétrons/líquido cefalorraquidiano , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Acidente Vascular Cerebral/enzimologia , Doença Aguda , Idoso , Líquido Cefalorraquidiano/enzimologia , Eritrócitos/enzimologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
12.
Arch Biochem Biophys ; 436(1): 91-100, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15752713

RESUMO

Cytochrome c (cyto-c) added to isolated mitochondria promotes the oxidation of extra-mitochondrial NADH and the reduction of molecular oxygen associated to the generation of an electrochemical membrane potential available for ATP synthesis. The electron transport pathway activated by exogenous cyto-c molecules is completely distinct from the one catalyzed by the respiratory chain. Dextran sulfate (500 kDa), known to interact with porin (the voltage-dependent anion channel), other than to inhibit the release of ATP synthesized inside the mitochondria, greatly decreases the activity of exogenous NADH/cyto-c system of intact mitochondria but has no effect on the reconstituted system made of mitoplasts and external membrane preparations. The results obtained are consistent with the existence of specific contact sites containing cytochrome oxidase and porin, as components of the inner and the outer membrane respectively, involved in the oxidation of cytosolic NADH. The proposal is put forward that the bi-trans-membrane electron transport chain activated by cytosolic cyto-c becomes, in physio-pathological conditions: (i) functional in removing the excess of cytosolic NADH; (ii) essential for cell survival in the presence of an impairment of the first three respiratory complexes; and (iii) an additional source of energy at the beginning of apoptosis.


Assuntos
Grupo dos Citocromos c/metabolismo , Citosol/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , NAD/metabolismo , Porinas/metabolismo , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , Fenômenos Fisiológicos Celulares , Sulfato de Dextrana/farmacologia , Eletroquímica , Transporte de Elétrons/efeitos dos fármacos , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/metabolismo , Potenciais da Membrana , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/metabolismo , Oxirredução , Oxigênio/metabolismo , Porinas/farmacologia
13.
Water Environ Res ; 75(5): 434-43, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14587954

RESUMO

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.


Assuntos
Anti-Infecciosos/metabolismo , Benzoatos/metabolismo , Reatores Biológicos , Oxigênio/análise , Purificação da Água/métodos , Bactérias/enzimologia , Biodegradação Ambiental , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia
14.
Biomed Environ Sci ; 16(3): 256-66, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14631831

RESUMO

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.


Assuntos
Antioxidantes/farmacologia , Ácido Ascórbico/farmacologia , Encéfalo/patologia , Estimulantes Ganglionares/toxicidade , Mitocôndrias/patologia , Nicotina/toxicidade , Fumaça/efeitos adversos , Adenosina Trifosfatases/farmacologia , Animais , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Radicais Livres , Camundongos
15.
Folia Biol (Krakow) ; 51(3-4): 201-5, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-15303375

RESUMO

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.


Assuntos
Cobre/envenenamento , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Exposição Ambiental , Troca Materno-Fetal/efeitos dos fármacos , Placenta/efeitos dos fármacos , Placenta/fisiologia , Adulto , Vilosidades Coriônicas/efeitos dos fármacos , Vilosidades Coriônicas/patologia , Complexo IV da Cadeia de Transporte de Elétrons/análise , Feminino , Glutationa Transferase/análise , Glutationa Transferase/farmacologia , Humanos , Imuno-Histoquímica , Lipídeos/análise , Troca Materno-Fetal/fisiologia , Mineração , Placenta/patologia , Polônia , Gravidez
17.
Sci Total Environ ; 290(1-3): 121-9, 2002 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-12083703

RESUMO

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.


Assuntos
Cervos , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Molibdênio/efeitos adversos , Miocárdio/enzimologia , Doenças dos Animais/etiologia , Animais , Cobre/deficiência , Edema/veterinária , Complexo IV da Cadeia de Transporte de Elétrons/análise , Síndrome , Redução de Peso
18.
West Indian Med J ; 50(2): 111-6, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11677906

RESUMO

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.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/fisiologia , Tubarões , Animais , Bovinos , Complexo IV da Cadeia de Transporte de Elétrons/química , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Humanos , Miocárdio/enzimologia
19.
West Indian med. j ; 50(2): 111-6, Jun. 2001. ilus, tab, gra
Artigo em Inglês | MedCarib | ID: med-349

RESUMO

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 are structually 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.(Au)


Assuntos
21003 , Bovinos , Estudo Comparativo , Complexo IV da Cadeia de Transporte de Elétrons/fisiologia , Tubarões , Modelos Animais de Doenças , Complexo IV da Cadeia de Transporte de Elétrons/química , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Miocardite/enzimologia
20.
J Biol Chem ; 275(50): 39625-30, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11006290

RESUMO

10-20% of individuals over the age of 65 suffer from age-related macular degeneration (AMD), the leading cause of severe visual impairment in humans living in developed countries. The pathogenesis of this complex disease is poorly understood, and no efficient therapy or prevention exists to date. A precondition for AMD appears to be the accumulation of the age pigment lipofuscin in lysosomes of retinal pigment epithelial (RPE) cells. In AMD, these cells seem to die by apoptosis with subsequent death of photoreceptor cells, and light may accelerate the disease process. Intracellular factors leading to cell death are not known. Here we show that the lipophilic cation N-retinyl-N-retinylidene ethanolamine (A2E), a lipofuscin component, induces apoptosis in RPE and other cells at concentrations found in human retina. Apoptosis is accompanied by the appearance of the proapoptotic proteins cytochrome c and apoptosis-inducing factor in the cytoplasm and the nucleus. Biochemical examinations show that A2E specifically targets cytochrome oxidase (COX). With both isolated mitochondria and purified COX, A2E inhibits oxygen consumption synergistically with light. Inhibition is reversed by the addition of cytochrome c or cardiolipin, a negatively charged phospholipid that facilitates the binding of cytochrome c to membranes. Succinate dehydrogenase activity is not altered by A2E. We suggest that A2E can act as a proapoptotic molecule via a mitochondria-related mechanism, possibly through site-specific targeting of this cation to COX. Loss of RPE cell viability through inhibition of mitochondrial function might constitute a pivotal step toward the progressive degeneration of the central retina.


Assuntos
Envelhecimento , Apoptose , Células Epiteliais/metabolismo , Lipofuscina/metabolismo , Degeneração Macular/metabolismo , Mitocôndrias/metabolismo , Pigmentos da Retina/metabolismo , Retinoides/metabolismo , Animais , Fator de Indução de Apoptose , Cardiolipinas/farmacologia , Cátions , Núcleo Celular/metabolismo , Sobrevivência Celular , Células Cultivadas , Cerebelo/metabolismo , Cromatografia Líquida de Alta Pressão , Grupo dos Citocromos c/metabolismo , Citoplasma/metabolismo , Fragmentação do DNA , Relação Dose-Resposta a Droga , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/farmacologia , Feminino , Flavoproteínas/metabolismo , Imuno-Histoquímica , Isomerismo , Luz , Potenciais da Membrana , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Muramidase/metabolismo , Consumo de Oxigênio , Células Fotorreceptoras/metabolismo , Ratos , Ratos Wistar , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...