Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neurochem Int ; 148: 105050, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33945834

RESUMO

Neurons spurn hydrogen-rich fatty acids for energizing oxidative ATP synthesis, contrary to other cells. This feature has been mainly attributed to a lower yield of ATP per reduced oxygen, as compared to glucose. Moreover, the use of fatty acids as hydrogen donor is accompanied by severe ß-oxidation-associated ROS generation. Neurons are especially susceptible to detrimental activities of ROS due to their poor antioxidative equipment. It is also important to note that free fatty acids (FFA) initiate multiple harmful activities inside the cells, particularly on phosphorylating mitochondria. Several processes enhance FFA-linked lipotoxicity in the cerebral tissue. Thus, an uptake of FFA from the circulation into the brain tissue takes place during an imbalance between energy intake and energy expenditure in the body, a situation similar to that during metabolic syndrome and fat-rich diet. Traumatic or hypoxic brain injuries increase hydrolytic degradation of membrane phospholipids and, thereby elevate the level of FFA in neural cells. Accumulation of FFA in brain tissue is markedly associated with some inherited neurological disorders, such as Refsum disease or X-linked adrenoleukodystrophy (X-ALD). What are strategies protecting neurons against FFA-linked lipotoxicity? Firstly, spurning the ß-oxidation pathway in mitochondria of neurons. Secondly, based on a tight metabolic communication between neurons and astrocytes, astrocytes donate metabolites to neurons for synthesis of antioxidants. Further, neuronal autophagy of ROS-emitting mitochondria combined with the transfer of degradation-committed FFA for their disposal in astrocytes, is a potent protective strategy against ROS and harmful activities of FFA. Finally, estrogens and neurosteroids are protective as triggers of ERK and PKB signaling pathways, consequently initiating the expression of various neuronal survival genes via the formation of cAMP response element-binding protein (CREB).


Assuntos
Encéfalo/metabolismo , Ácidos Graxos/toxicidade , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Metabolismo Energético/fisiologia , Ácidos Graxos/metabolismo , Humanos , Mitocôndrias/efeitos dos fármacos , Neurônios/metabolismo , Estresse Oxidativo/fisiologia
2.
Z Gastroenterol ; 58(3): 245-253, 2020 Mar.
Artigo em Alemão | MEDLINE | ID: mdl-32018318

RESUMO

The abdominal surgeon may have the opportunity to steadily learn on the (patho-)biochemical and (-)physiological consequences of his disease-related surgical activity (change of anatomy of the GI tract and its surrounding organs, medication and so on) if he refers closely to several medical disciplines as specifically indicated. AIM & METHOD: By means of a short compact overview based on (i) topic-related references from the scientific medical literature and (ii) own surgery-specific perceptions, interrelation of cholic acids (CA) with metabolism, in particular, with planned or performed (abdomino-)surgical procedures should be illustrated. RESULTS (CORNER POINTS): 1. Surgery and biochemistry have a common and traditionally matured matter of consideration with regard to the consequences of an altered portal vein circulation and liver cirrhosis. 2. CA are (i) natural detergents, (ii) components of cholesterol-associated gall stones and (iii) essential signal molecules of intestine-liver metabolic interaction. CA and chenodesoxycholic acid [CDCA] dominate the CA pool with approximately 35 %. By conjugation of CA with taurine und glycine, its solubility is increased. The enterohepatic circulation minimizes the excretion of CA. 3. The generation of CA out of cholesterol within the liver (turnover/day: 0.2-0.6 g cholesterol) is controlled by cholesterol-7α-hydroxylase (CYP7A1). A toxic CA accumulation is prevented by a CA-induced repression of CYP7A1 expression and sulfation of CA (resulting in an increase of urine solubility). 4. CA show regulatory activities in the energy, glucose, lipid and lipoprotein metabolism and connate immune system. By binding of the CA to the farnesoid X-nuclear receptor [FXR] and the membranous G-protein-coupled CA receptor-1 [GPBAR1, TGR5], mannifold effects within the fat and carbohydrate metabolism are induced. 5. CA trigger the expression of the iodothyronine-dejodinase (DIO2) within the brown fat tissue and skelet muscles by activation of the GPBAR1-MAPK signal pathways. Hence, thyroxine (T4) is transformed to trijodthyronine (T3) and, subsequently, fat oxidation and thermogenesis are increased. 6. CA change the intestinal microbioma by bacteriolytic activities and, on the other hand, the CA profile is modulated by the microbioma. Typical microbial effects of the CA pool are (i) separation of glycine and taurine residuals of conjugated CA by "bile salt hydrolases" and (ii) chemical modification of free, primary CA by re-amidation, oxidation-reduction, esterification and desulfation. 7. CA inhibit the endotoxin-based inflammatory response induced by lipopolysaccharides (LPS; membranous component of gram-negative bacteria). Via binding of CA to macrophages-associated receptors (GPBAR1 and FXR), (i) the LPS-induced proinflammatory cytokine generation is reduced and the expression of antiinflammatory IL-10 is promoted. In addition, (ii) white-blood cell "trafficking" is regulated and (iii) inflammasoma is activated by macrophages and neutrophil granulocytes. 8. The body weight-independent changes after bariatric surgery (e. g., in case of "Roux-en-Y gastric bypass" [RYGB]) correlate with an increased CA serum level and an altered intestinal CA profile. The latter leads secundarily to a modification of the microbioma. RYGB has - among others - positive effects onto the carbohydrate metabolism. Thus, insulin sensitivity of the liver is improved and the secretion of the glucagon-like peptide 1 is enhanced. CONCLUSION: CA are a parade example for metabolic regulators, the interactions of which have an impact onto various (patho-)biochemical and (-)physiological processes, (abdomino-)surgically relevant diseases and (abdomino-)surgical measures. Their biochemical/physiological activities and insight into associated molecular processes should be part of the medical and scientific skills of a modern (abdominal) surgeon with a developed pathophysiological expertise.


Assuntos
Ácidos Cólicos , Microbiota , Cirurgiões , Ácidos e Sais Biliares , Circulação Êntero-Hepática , Humanos , Intestinos , Fígado , Masculino , Receptores Acoplados a Proteínas G
4.
J Biomed Mater Res B Appl Biomater ; 107(4): 1246-1253, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30261124

RESUMO

Total joint replacement is one of the most successful procedures in orthopedic surgery today. However, metal implant materials undergo wear and corrosion processes. Generated particles and ions can cause a variety of cellular reactions. Cobalt-containing alloys are used frequently in implant materials. Some studies suggest that cobalt exhibits potential cytotoxic effects, for example, via generation of reactive oxygen species (ROS). To further elucidate the effects of cobalt on human cells, we determined cell viability and cytosolic and mitochondrial superoxide formation after incubation of either ions or particles with different cells. MM-6 and Jurkat cell lines were treated for 24, 48 and 72 h with either CoCrMo particles or cobalt ions (supplied as CoCl2 ). A total of 24 h exposure of both forms of cobalt did not induce cell death using terminal deoxynucleotidyl transferase (TUNEL) and trypan blue assay. Interestingly, the formation of superoxide (O2.- ) is evoked mainly by ionic CoCl2 but not cobalt particles. Cobalt alloy particles are likely to even suppress O2.- formation in mitochondria in both used cell lines. Furthermore, we did not observe any effect of cobalt particles on O2.- formation in peripheral blood mononuclear cells (PBMCs) from healthy donors. We also found that the O2- formation by CoCl2 within mitochondria is a generalized effect for all cell types used, while the formation of superoxide in cytosolic compartment is cell-type dependent. In summary, our data suggest that cobalt ions specifically induce the formation of O2.- , whereas the cobalt particles were better tolerated. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1246-1253, 2019.


Assuntos
Ligas/farmacologia , Artroplastia de Quadril , Cobalto/farmacologia , Prótese de Quadril , Leucócitos Mononucleares/imunologia , Superóxidos/imunologia , Humanos , Células Jurkat
5.
Mycotoxin Res ; 33(3): 207-218, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28474303

RESUMO

The aim of the present study was to examine the role of chronic deoxynivalenol (DON) exposition on the liver morphology and function in combination with pre- and post-hepatic lipopolysaccharide (LPS) stress in young pigs fed for 4 weeks with a DON-contaminated diet (4.59 mg/kg feed). At the end of the experiment, LPS (7.5 µg/kg BW) was administered for 1 h pre-hepatically (Vena portae hepatis) or post-hepatically (Vena jugularis). Liver morphology was macroscopically checked and showed haemorrhage in all LPS groups, significantly higher relative liver weights, accompanied by marked oedema in the gallbladder wall. Histological changes were judged by a modified histology activity index (HAI). Liver HAI score was significantly increased in all LPS groups compared to placebo, primarily due to neutrophil infiltration and haemorrhage. DON feed alone was without effect on the liver HAI. Liver function was characterized by (i) hepatic biochemical markers, (ii) mitochondrial respiration and (iii) Ca2+ accumulation capacity of isolated mitochondria. Clinical chemical parameters characterizing liver function were initially (<3 h) slightly influenced by LPS. After 3 h, bilirubin and alkaline phosphatase were increased significantly, in DON-fed, jugular-infused LPS group. Respiration and Ca2+ accumulation capacity of isolated liver mitochondria was not impaired by chronic DON exposure, acute LPS challenge or combined treatments. DON-contaminated feed did not change macroscopy and histology of the liver, but modified the function under LPS stress. The different function was not linked to modifications of liver mitochondria.


Assuntos
Fígado/efeitos dos fármacos , Tricotecenos/toxicidade , Ração Animal , Animais , Dieta/veterinária , Contaminação de Alimentos , Inflamação , Lipopolissacarídeos , Fígado/patologia , Mitocôndrias Hepáticas/efeitos dos fármacos , Suínos
6.
Neurochem Int ; 109: 68-77, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28366720

RESUMO

The brain uses long-chain fatty acids (LCFAs) to a negligible extent as fuel for the mitochondrial energy generation, in contrast to other tissues that also demand high energy. Besides this generally accepted view, some studies using cultured neural cells or whole brain indicate a moderately active mitochondrial ß-oxidation. Here, we corroborate the conclusion that brain mitochondria are unable to oxidize fatty acids. In contrast, the combustion of liver-derived ketone bodies by neural cells is long-known. Furthermore, new insights indicate the use of odd-numbered medium-chain fatty acids as valuable source for maintaining the level of intermediates of the citric acid cycle in brain mitochondria. Non-esterified LCFAs or their activated forms exert a large variety of harmful side-effects on mitochondria, such as enhancing the mitochondrial ROS generation in distinct steps of the ß-oxidation and therefore potentially increasing oxidative stress. Hence, the question arises: Why do in brain energy metabolism mitochondria selectively spurn LCFAs as energy source? The most likely answer are the relatively higher content of peroxidation-sensitive polyunsaturated fatty acids and the low antioxidative defense in brain tissue. There are two remarkable peroxisomal defects, one relating to α-oxidation of phytanic acid and the other to uptake of very long-chain fatty acids (VLCFAs) which lead to pathologically high tissue levels of such fatty acids. Both, the accumulation of phytanic acid and that of VLCFAs give an enlightening insight into harmful activities of fatty acids on neural cells, which possibly explain why evolution has prevented brain mitochondria from the equipment with significant ß-oxidation enzymatic capacity.


Assuntos
Encéfalo/metabolismo , Metabolismo Energético/fisiologia , Ácidos Graxos/metabolismo , Mitocôndrias/metabolismo , Doenças Neurodegenerativas/metabolismo , Estresse Oxidativo/fisiologia , Animais , Humanos , Espécies Reativas de Oxigênio/metabolismo
7.
J Cereb Blood Flow Metab ; 37(3): 848-854, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27055779

RESUMO

According to recent reports, systemic treatment of rats with methylpalmoxirate (carnitine palmitoyltransferase-1 inhibitor) decreased peroxidation of polyunsaturated fatty acids in brain tissue. This was taken as evidence of mitochondrial ß-oxidation in brain, thereby contradicting long-standing paradigms of cerebral metabolism, which claim that ß-oxidation of activated fatty acids has minor importance for brain energy homeostasis. We addressed this controversy. Our experiments are the first direct experimental analysis of this question. We fueled isolated brain mitochondria or rat brain astrocytes with octanoic acid, but octanoic acid does not enhance formation of reactive oxygen species, neither in isolated brain mitochondria nor in astrocytes, even at limited hydrogen delivery to mitochondria. Thus, octanoic acid or l-octanoylcarnitine does not stimulate H2O2 release from brain mitochondria fueled with malate, in contrast to liver mitochondria (2.25-fold rise). This does obviously not support the possible occurrence of ß-oxidation of the fatty acid octanoate in the brain. We conclude that a proposed inhibition of ß-oxidation does not seem to be a helpful strategy for therapies aiming at lowering oxidative stress in cerebral tissue. This question is important, since oxidative stress is the cause of neurodegeneration in numerous neurodegenerative or inflammatory disease situations.


Assuntos
Encéfalo/metabolismo , Oxirredução , Estresse Oxidativo , Animais , Caprilatos/metabolismo , Carnitina/análogos & derivados , Carnitina/metabolismo , Metabolismo Energético , Homeostase , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Ratos
8.
Aging Dis ; 7(2): 136-49, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27114847

RESUMO

It is increasingly understood that in the aging brain, especially in the case of patients suffering from neurodegenerative diseases, some fatty acids at pathologically high concentrations exert detrimental activities. To study such activities, we here analyze genetic diseases, which are due to compromised metabolism of specific fatty acids, either the branched-chain phytanic acid or very long-chain fatty acids (VLCFAs). Micromolar concentrations of phytanic acid or of VLCFAs disturb the integrity of neural cells by impairing Ca(2+) homeostasis, enhancing oxidative stress or de-energizing mitochondria. Finally, these combined harmful activities accelerate cell death. Mitochondria are more severely targeted by phytanic acid than by VLCFAs. The insertion of VLCFAs into the inner membrane distorts the arrangement of membrane constituents and their functional interactions. Phytanic acid exerts specific protonophoric activity, induces reactive oxygen species (ROS) generation, and reduces ATP generation. A clear inhibition of the Na(+), K(+)-ATPase activity by phytanic acid has also been reported. In addition to the instantaneous effects, a chronic exposure of brain cells to low micromolar concentrations of phytanic acid may produce neuronal damage in Refsum disease by altering epigenetic transcriptional regulation. Myelin-producing oligodendrocytes respond with particular sensitivity to VLCFAs. Deleterious activity of VLCFAs on energy-dependent mitochondrial functions declines with increasing the hydrocarbon chain length (C22:0 > C24:0 > C26:0). In contrast, the reverse sequence holds true for cell death induction by VLCFAs (C22:0 < C24:0 < C26:0). In adrenoleukodystrophy, the uptake of VLCFAs by peroxisomes is impaired by defects of the ABCD1 transporter. Studying mitochondria from ABCD1-deficient and wild-type mice proves that the energy-dependent functions are not altered in the disease model. Thus, a defective ABCD1 apparently exerts no obvious adaptive pressure on mitochondria. Further research has to elucidate the detailed mechanistic basis for the failures causing fatty acid-mediated neurodegeneration and should help to provide possible therapeutic interventions.

9.
J Lipid Res ; 57(6): 943-54, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27080715

RESUMO

Short- and medium-chain fatty acids (SCFAs and MCFAs), independently of their cellular signaling functions, are important substrates of the energy metabolism and anabolic processes in mammals. SCFAs are mostly generated by colonic bacteria and are predominantly metabolized by enterocytes and liver, whereas MCFAs arise mostly from dietary triglycerides, among them milk and dairy products. A common feature of SCFAs and MCFAs is their carnitine-independent uptake and intramitochondrial activation to acyl-CoA thioesters. Contrary to long-chain fatty acids, the cellular metabolism of SCFAs and MCFAs depends to a lesser extent on fatty acid-binding proteins. SCFAs and MCFAs modulate tissue metabolism of carbohydrates and lipids, as manifested by a mostly inhibitory effect on glycolysis and stimulation of lipogenesis or gluconeogenesis. SCFAs and MCFAs exert no or only weak protonophoric and lytic activities in mitochondria and do not significantly impair the electron transport in the respiratory chain. SCFAs and MCFAs modulate mitochondrial energy production by two mechanisms: they provide reducing equivalents to the respiratory chain and partly decrease efficacy of oxidative ATP synthesis.


Assuntos
Trifosfato de Adenosina/metabolismo , Metabolismo Energético , Ácidos Graxos/metabolismo , Mitocôndrias/metabolismo , Acil Coenzima A/metabolismo , Trifosfato de Adenosina/biossíntese , Carnitina/metabolismo , Ácidos Graxos/biossíntese , Humanos , Oxirredução , Triglicerídeos/metabolismo
10.
Biochim Biophys Acta ; 1852(5): 925-36, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25583114

RESUMO

X-linked adrenoleukodystrophy (X-ALD) is a severe neurodegenerative disorder resulting from defective ABCD1 transport protein. ABCD1 mediates peroxisomal uptake of free very-long-chain fatty acids (VLCFA) as well as their CoA-esters. Consequently, VLCFA accumulate in patients' plasma and tissues, which is considered as pathogenic X-ALD triggering factor. Clinical symptoms are mostly manifested in neural tissues and adrenal gland. Here, we investigate astrocytes from wild-type control and a genetic X-ALD mouse model (Abcd1-knockout), exposed to supraphysiological VLCFA (C22:0, C24:0 and C26:0) concentrations. They exhibit multiple impairments of energy metabolism. Furthermore, brain mitochondria from Abcd1(-/-) mice and wild-type control respond similarly to VLCFA with increased ROS generation, impaired oxidative ATP synthesis and diminished Ca(2+) uptake capacity, suggesting that a defective ABCD1 exerts no adaptive pressure on mitochondria. In contrast, astrocytes from Abcd1(-/-) mice respond more sensitively to VLCFA than wild-type control astrocytes. Moreover, long-term application of VLCFA induces high ROS generation, and strong in situ depolarization of mitochondria, and, in Abcd1(-/-) astrocytes, severely diminishes the capability to revert oxidized pyridine nucleotides to NAD(P)H. In addition, observed differences in responses of mitochondria and astrocytes to the hydrocarbon chain length of VLCFA suggest that detrimental VLCFA activities in astrocytes involve defective cellular functions other than mitochondria. In summary, we clearly demonstrate that VLCFA increase the vulnerability of Abcd1(-/-) astrocytes.


Assuntos
Transportadores de Cassetes de Ligação de ATP/deficiência , Adrenoleucodistrofia/metabolismo , Astrócitos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Ácidos Graxos/farmacologia , Mitocôndrias/efeitos dos fármacos , Membro 1 da Subfamília D de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Adrenoleucodistrofia/genética , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Astrócitos/metabolismo , Cálcio/metabolismo , Cálcio/farmacocinética , Células Cultivadas , Ácidos Graxos/química , Transporte de Íons/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Mitocôndrias/metabolismo , NADP/metabolismo , Fosforilação Oxidativa/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Espectrometria de Fluorescência , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
11.
J Neurochem ; 131(2): 163-76, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24923354

RESUMO

Ca(2+) -independent phospholipase A2 (iPLA2 ) is hypothesized to control mitochondrial reactive oxygen species (ROS) generation. Here, we modulated the influence of iPLA2 -induced liberation of non-esterified free fatty acids on ROS generation associated with the electron transport chain. We demonstrate enzymatic activity of membrane-associated iPLA2 in native, energized rat brain mitochondria (RBM). Theoretically, enhanced liberation of free fatty acids by iPLA2 modulates mitochondrial ROS generation, either attenuating the reversed electron transport (RET) or deregulating the forward electron transport of electron transport chain. For mimicking such conditions, we probed the effect of docosahexaenoic acid (DHA), a major iPLA2 product on ROS generation. We demonstrate that the adenine nucleotide translocase partly mediates DHA-induced uncoupling, and that low micromolar DHA concentrations diminish RET-dependent ROS generation. Uncoupling proteins have no effect, but the adenine nucleotide translocase inhibitor carboxyatractyloside attenuates DHA-linked uncoupling effect on RET-dependent ROS generation. Under physiological conditions of forward electron transport, low micromolar DHA stimulates ROS generation. Finally, exposure of RBM to the iPLA2 inhibitor bromoenol lactone (BEL) enhanced ROS generation. BEL diminished RBM glutathione content. BEL-treated RBM exhibits reduced Ca(2+) retention capacity and partial depolarization. Thus, we rebut the view that iPLA2 attenuates oxidative stress in brain mitochondria. However, the iPLA2 inhibitor BEL has detrimental activities on energy-dependent mitochondrial functions. The Ca(2+) -independent phospholipase A2 (iPLA2 ), a FFA (free fatty acids)-generating membrane-attached mitochondrial phospholipase, is potential to regulate ROS (reactive oxygen species) generation by mitochondria. FFA can either decrease reversed electron transport (RET)-linked or enhance forward electron transport (FET)-linked ROS generation. In the physiological mode of FET, iPLA2 activity increases ROS generation. The iPLA2 inhibitor BEL exerts detrimental effects on energy-dependent mitochondrial functions.


Assuntos
Encéfalo/metabolismo , Cálcio/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Mitocôndrias/metabolismo , Naftalenos/metabolismo , Fosfolipases A2/metabolismo , Pironas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Cálcio/farmacologia , Ácidos Docosa-Hexaenoicos/farmacologia , Transporte de Elétrons/efeitos dos fármacos , Transporte de Elétrons/fisiologia , Mitocôndrias/efeitos dos fármacos , Naftalenos/farmacologia , Pironas/farmacologia , Ratos , Ratos Wistar
12.
J Cell Mol Med ; 17(12): 1619-31, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24118792

RESUMO

The production of hydrogen peroxide (H2 O2 ) drives tumourigenesis in ulcerative colitis (UC). Recently, we showed that H2 O2 activates DNA damage checkpoints in human colonic epithelial cells (HCEC) through c-Jun N-terminal Kinases (JNK) that induces p21(WAF1) . Moreover, caspases circumvented the G1/S and intra-S checkpoints, and cells accumulated in G2/M. The latter observation raised the question of whether repeated H2 O2 exposures alter JNK activation, thereby promoting a direct passage of cells from G2/M arrest to driven cell cycle progression. Here, we report that increased proliferation of repeatedly H2 O2 -exposed HCEC cells (C-cell cultures) was associated with (i) increased phospho-p46 JNK, (ii) decreased total JNK and phospho-p54 JNK and (iii) p21(WAF1) down-regulation. Altered JNK activation and p21(WAF1) down-regulation were accompanied by defects in maintaining G2/M and mitotic spindle checkpoints through adaptation, as well as by apoptosis resistance following H2 O2 exposure. This may cause increased proliferation of C-cell cultures, a defining initiating feature in the inflammation-carcinoma pathway in UC. We further suggest that dysregulated JNK activation is attributed to a non-apoptotic function of caspases, causing checkpoint adaptation in C-cell cultures. Additionally, loss of cell-contact inhibition and the overcoming of senescence, hallmarks of cancer, contributed to increased proliferation. Furthermore, there was evidence that p54 JNK inactivation is responsible for loss of cell-contact inhibition. We present a cellular model of UC and suggest a sinusoidal pattern of proliferation, which is triggered by H2 O2 -induced reactive oxygen species generation, involving an interplay between JNK activation/inactivation, p21(WAF1) , c-Fos, c-Jun/phospho-c-Jun, ATF2/phospho-ATF2, ß-catenin/TCF4-signalling, c-Myc, CDK6 and Cyclin D2, leading to driven cell cycle progression.


Assuntos
Ciclo Celular/efeitos dos fármacos , Colite Ulcerativa/patologia , Peróxido de Hidrogênio/farmacologia , Modelos Biológicos , Caspases/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Inibição de Contato/efeitos dos fármacos , Ciclina D2/metabolismo , Quinase 6 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição/metabolismo
13.
FEBS J ; 280(24): 6589-99, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24119167

RESUMO

Several studies have proposed that the antibiotic minocycline (MC) has cytoprotective activities. Nevertheless, when cells have been exposed to MC at micromolar concentrations, detrimental effects have been also observed. Despite the known inhibitory activity of MC on ATP synthesis and the Ca(2+) retention capacity of isolated rat liver and brain mitochondria, the underlying mechanism is still debated. Here, we present further arguments supporting our concept that MC acting on rat liver mitochondria suspended in KCl medium permeabilizes the inner membrane. Supplementation of the medium with cytochrome c and NAD(+) strongly enhanced the respiration of MC-treated mitochondria, thus partly preventing or reversing the inhibitory effect of MC on state 3 or uncoupled respiration. These results indicate that MC produced depletion of mitochondrial cytochrome c and NAD(+) , thus impairing mitochondrial respiration. In addition, NADH oxidation by alamethicin-permeabilized mitochondria supplemented with cytochrome c was insensitive to 200 µm MC, arguing against direct impairment of respiratory chain complexes by MC. Finally, a surprising feature of MC was its accumulation or binding by intact rat liver mitochondria, but not by mitochondria permeabilized with alamethicin or disrupted by freezing and thawing.


Assuntos
Antibacterianos/farmacologia , Respiração Celular/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Minociclina/farmacologia , Mitocôndrias Hepáticas/metabolismo , Dilatação Mitocondrial/efeitos dos fármacos , Alameticina/farmacologia , Animais , Carbonil Cianeto p-Trifluormetoxifenil Hidrazona/farmacologia , Citocromos c/metabolismo , Mitocôndrias Hepáticas/efeitos dos fármacos , Membranas Mitocondriais/efeitos dos fármacos , NAD/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Ionóforos de Próton/farmacologia , Ratos , Ratos Wistar
14.
J Cereb Blood Flow Metab ; 33(10): 1493-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23921897

RESUMO

It is puzzling that hydrogen-rich fatty acids are used only poorly as fuel in the brain. The long-standing belief that a slow passage of fatty acids across the blood-brain barrier might be the reason. However, this has been corrected by experimental results. Otherwise, accumulated nonesterified fatty acids or their activated derivatives could exert detrimental activities on mitochondria, which might trigger the mitochondrial route of apoptosis. Here, we draw attention to three particular problems: (1) ATP generation linked to ß-oxidation of fatty acids demands more oxygen than glucose, thereby enhancing the risk for neurons to become hypoxic; (2) ß-oxidation of fatty acids generates superoxide, which, taken together with the poor anti-oxidative defense in neurons, causes severe oxidative stress; (3) the rate of ATP generation based on adipose tissue-derived fatty acids is slower than that using blood glucose as fuel. Thus, in periods of extended continuous and rapid neuronal firing, fatty acid oxidation cannot guarantee rapid ATP generation in neurons. We conjecture that the disadvantages connected with using fatty acids as fuel have created evolutionary pressure on lowering the expression of the ß-oxidation enzyme equipment in brain mitochondria to avoid extensive fatty acid oxidation and to favor glucose oxidation in brain.


Assuntos
Encéfalo/metabolismo , Metabolismo Energético , Ácidos Graxos/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Glucose/metabolismo , Humanos , Mitocôndrias/metabolismo , Neurônios/metabolismo , Oxirredução , Estresse Oxidativo , Oxigênio/metabolismo
15.
J Cell Mol Med ; 17(8): 976-88, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23800081

RESUMO

Cancer cells showing low apoptotic effects following oxidative stress-induced DNA damage are mainly affected by growth arrest. Thus, recent studies focus on improving anti-cancer therapies by increasing apoptosis sensitivity. We aimed at identifying a universal molecule as potential target to enhance oxidative stress-based anti-cancer therapy through a switch from cell cycle arrest to apoptosis. A cDNA microarray was performed with hydrogen peroxide-treated oesophageal squamous epithelial cancer cells TE7. This cell line showed checkpoint activation via p21(WAF1) , but low apoptotic response following DNA damage. The potential target molecule was chosen depended on the following demands: it should regulate DNA damage response, cell cycle and apoptosis. As the transcription factor ATF2 is implicated in all these processes, we focused on this protein. We investigated checkpoint activation via ATF2. Indeed, ATF2 knockdown revealed ATF2-triggered p21(WAF1) protein expression, suggesting p21(WAF1) transactivation through ATF2. Using chromatin immunoprecipitation (ChIP), we identified a hitherto unknown ATF2-binding sequence in the p21(WAF1) promoter. p-ATF2 was found to interact with p-c-Jun, creating the AP-1 complex. Moreover, ATF2 knockdown led to c-Jun downregulation. This suggests ATF2-driven induction of c-Jun expression, thereby enhancing ATF2 transcriptional activity via c-Jun-ATF2 heterodimerization. Notably, downregulation of ATF2 caused a switch from cell cycle arrest to reinforced apoptosis, presumably via p21(WAF1) downregulation, confirming the importance of ATF2 in the establishment of cell cycle arrest. 1-Chloro-2,4-dinitrobenzene also led to ATF2-dependent G2/M arrest, suggesting that this is a general feature induced by oxidative stress. As ATF2 knockdown also increased apoptosis, we propose ATF2 as a target for combined oxidative stress-based anti-cancer therapies.


Assuntos
Fator 2 Ativador da Transcrição/metabolismo , Apoptose , Técnicas de Silenciamento de Genes , Estresse Oxidativo , Apoptose/efeitos dos fármacos , Apoptose/genética , Sítios de Ligação , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/farmacologia , Modelos Biológicos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-jun/metabolismo
16.
Biochem Biophys Res Commun ; 428(2): 234-8, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23068098

RESUMO

In this report, we show new experimental evidence that, in mouse brain mitochondria, uncoupling protein-2 (UCP2) can be involved in superoxide (O(2)(·-)) removal from the mitochondrial matrix. We found that the effect of guanosine 5'-diphosphate (GDP) on the rate of reactive oxygen species (ROS) release from brain mitochondria of UCP2 knockout mice was less pronounced compared to the wild type animals. This putative novel UCP2 activity, evaluated by the use of UCP2-knockout transgenic animals, along with the known antioxidant defence systems, may provide additional protection from ROS in brain mitochondria.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Canais Iônicos/fisiologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/fisiologia , Superóxidos/metabolismo , Animais , Guanosina Difosfato/farmacologia , Canais Iônicos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mitocondriais/genética , Espécies Reativas de Oxigênio/metabolismo , Proteína Desacopladora 2
18.
Biochim Biophys Acta ; 1817(10): 1747-53, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22414665

RESUMO

Existing literature on estradiol indicates that it affects mitochondrial functions at low micromolar concentrations. Particularly blockade of the permeability transition pore (PTP) or modulation of the enzymatic activity of one or more complexes of the respiratory chain were suspicious. We prepared mitoplasts from rat liver mitochondria (RLM) to study by single-channel patch-clamp techniques the PTP, and from rat astrocytes to study the potassium BK-channel said to modulate the PTP. Additionally, we measured respiration of intact RLM. After application of 17ß-estradiol (ßE) our single-channel results reveal a transient increase of activity of both, the BK-channel and the PTP followed by their powerful inhibition. Respiration measurements demonstrate inhibition of the Ca(2+)-induced permeability transition, as well, though only at higher concentrations (≥30µM). At lower concentrations, we observed an increase of endogenous- and state 2-respiration. Furthermore, we show that ßE diminishes the phosphorylating respiration supported by complex I-substrates (glutamate/malate) or by the complex II-substrate succinate. Taken together the results suggest that ßE affects mitochondria by several modes, including partial inhibition of the activities of ion channels of the inner membrane and of respiration. This article is part of a Special Issue entitled: 17th European Bioenergetics Conference (EBEC 2012).


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Mitocôndrias Hepáticas/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Membranas Mitocondriais/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Animais , Cálcio/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/antagonistas & inibidores , Poro de Transição de Permeabilidade Mitocondrial , Fosforilação/efeitos dos fármacos , Ratos
19.
Biochim Biophys Acta ; 1817(3): 410-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22226918

RESUMO

Mitochondria from brown adipose tissue (BATM) have a high enzymatic capacity for fatty acid oxidation and therefore are an ideal model to examine the sites of reactive oxygen species (ROS) generation during fatty acid oxidation. ROS generation by BATM (isolated from 3-week-old rats) was measured during acylcarnitine oxidation as release of H(2)O(2) into the medium and as inactivation of the matrix enzyme aconitase. The following results were obtained: (1) BATM release large amounts of H(2)O(2) in the coupled as well as in the uncoupled states, several times more than skeletal muscle mitochondria. (2) H(2)O(2) release is especially large with acylcarnitines of medium-chain fatty acids (e.g. octanoylcarnitine). (3) Reverse electron transport does not contribute in a significant extent to the overall ROS generation. (4) Despite the large release of H(2)O(2), the ROS-sensitive matrix enzyme aconitase is not inactivated during acylcarnitine oxidation. (5) In contrast to acylcarnitines, oxidation of α-glycerophosphate by BATM is characterized by large H(2)O(2) release and a pronounced aconitase inactivation. We hypothesize that acylcarnitine-supported ROS generation in BATM may be mainly associated with acyl-CoA dehydrogenase and electron transferring flavoprotein-ubiquinone reductase rather than with complexes of the respiratory chain.


Assuntos
Tecido Adiposo Marrom/metabolismo , Canais Iônicos/fisiologia , Mitocôndrias/metabolismo , Proteínas Mitocondriais/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Carnitina/análogos & derivados , Carnitina/metabolismo , Ácidos Graxos/metabolismo , Peróxido de Hidrogênio/metabolismo , Potencial da Membrana Mitocondrial , Nigericina/farmacologia , Oniocompostos/farmacologia , Oxirredução , Ratos , Rotenona/farmacologia , Proteína Desacopladora 1
20.
Lab Invest ; 91(12): 1766-76, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21863062

RESUMO

The mitochondrial protein frataxin (FXN) is known to be involved in mitochondrial iron homeostasis and iron-sulfur cluster biogenesis. It is discussed to modulate function of the electron transport chain and production of reactive oxygen species (ROS). FXN loss in neurons and heart muscle cells causes an autosomal-dominant mitochondrial disorder, Friedreich's ataxia. Recently, tumor induction after targeted FXN deletion in liver and reversal of the tumorigenic phenotype of colonic carcinoma cells following FXN overexpression were described in the literature, suggesting a tumor suppressor function. We hypothesized that a partial reversal of the malignant phenotype of glioma cells should occur after FXN transfection, if the mitochondrial protein has tumor suppressor functions in these brain tumors. In astrocytic brain tumors and tumor cell lines, we observed reduced FXN levels compared with non-neoplastic astrocytes. Mitochondrial content (citrate synthase activity) was not significantly altered in U87MG glioblastoma cells stably overexpressing FXN (U87-FXN). Surprisingly, U87-FXN cells exhibited increased cytoplasmic ROS levels, although mitochondrial ROS release was attenuated by FXN, as expected. Higher cytoplasmic ROS levels corresponded to reduced activities of glutathione peroxidase and catalase, and lower glutathione content. The defect of antioxidative capacity resulted in increased susceptibility of U87-FXN cells against oxidative stress induced by H(2)O(2) or buthionine sulfoximine. These characteristics may explain a higher sensitivity toward staurosporine and alkylating drugs, at least in part. On the other hand, U87-FXN cells exhibited enhanced growth rates in vitro under growth factor-restricted and hypoxic conditions and in vivo using tumor xenografts in nude mice. These data contrast to a general tumor suppressor function of FXN but suggest a dual, pro-proliferative but chemosensitizing role in astrocytic tumors.


Assuntos
Astrocitoma/metabolismo , Proteínas de Ligação ao Ferro/metabolismo , Animais , Antineoplásicos Alquilantes , Apoptose , Linhagem Celular Transformada , Linhagem Celular Tumoral , Técnicas de Transferência de Genes , Humanos , Proteínas de Ligação ao Ferro/genética , Camundongos , Camundongos Nus , Mitocôndrias/metabolismo , Neuroglia/metabolismo , Fosforilação Oxidativa , Estresse Oxidativo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Frataxina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA