Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Redox Biol ; 63: 102758, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37245287

RESUMO

Ferroptosis is an iron dependent form of cell death, that is triggered by the discoordination of iron, lipids, and thiols. Its unique signature that distinguishes it from other forms of cell death is the formation and accumulation of lipid hydroperoxides, particularly oxidized forms of polyunsaturated phosphatidylethanolamines (PEs), which drives cell death. These readily undergo iron-catalyzed secondary free radical reactions leading to truncated products which retain the signature PE headgroup and which can readily react with nucleophilic moieties in proteins via their truncated electrophilic acyl chains. Using a redox lipidomics approach, we have identified oxidatively-truncated PE species (trPEox) in enzymatic and non-enzymatic model systems. Further, using a model peptide we demonstrate adduct formation with Cys as the preferred nucleophilic residue and PE(26:2) +2 oxygens, as one of the most reactive truncated PE-electrophiles produced. In cells stimulated to undergo ferroptosis we identified PE-truncated species with sn-2 truncations ranging from 5 to 9 carbons. Taking advantage of the free PE headgroup, we have developed a new technology using the lantibiotic duramycin, to enrich and identify the PE-lipoxidated proteins. Our results indicate that several dozens of proteins for each cell type, are PE-lipoxidated in HT-22, MLE, and H9c2 cells and M2 macrophages after they were induced to undergo ferroptosis. Pretreatment of cells with the strong nucleophile, 2-mercaptoethanol, prevented the formation of PE-lipoxidated proteins and blocked ferroptotic death. Finally, our docking simulations showed that the truncated PE species bound at least as good to several of the lantibiotic-identified proteins, as compared to the non-truncated parent molecule, stearoyl-arachidonoyl PE (SAPE), indicating that these oxidatively-truncated species favor/promote the formation of PEox-protein adducts. The identification of PEox-protein adducts during ferroptosis suggests that they are participants in the ferroptotic process preventable by 2-mercaptoethanol and may contribute to a point of no return in the ferroptotic death process.


Assuntos
Ferroptose , Humanos , Mercaptoetanol , Oxirredução , Morte Celular , Ferro/metabolismo , Peroxidação de Lipídeos
2.
Free Radic Biol Med ; 147: 231-241, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31883467

RESUMO

High fidelity and effective adaptive changes of the cell and tissue metabolism to changing environments require strict coordination of numerous biological processes. Multicellular organisms developed sophisticated signaling systems of monitoring and responding to these different contexts. Among these systems, oxygenated lipids play a significant role realized via a variety of re-programming mechanisms. Some of them are enacted as a part of pro-survival pathways that eliminate harmful or unnecessary molecules or organelles by a variety of degradation/hydrolytic reactions or specialized autophageal processes. When these "partial" intracellular measures are insufficient, the programs of cells death are triggered with the aim to remove irreparably damaged members of the multicellular community. These regulated cell death mechanisms are believed to heavily rely on signaling by a highly diversified group of molecules, oxygenated phospholipids (PLox). Out of thousands of detectable individual PLox species, redox phospholipidomics deciphered several specific molecules that seem to be diagnostic of specialized death programs. Oxygenated cardiolipins (CLs) and phosphatidylethanolamines (PEs) have been identified as predictive biomarkers of apoptosis and ferroptosis, respectively. This has led to decoding of the enzymatic mechanisms of their formation involving mitochondrial oxidation of CLs by cytochrome c and endoplasmic reticulum-associated oxidation of PE by lipoxygenases. Understanding of the specific biochemical radical-mediated mechanisms of these oxidative reactions opens new avenues for the design and search of highly specific regulators of cell death programs. This review emphasizes the usefulness of such selective lipid peroxidation mechanisms in contrast to the concept of random poorly controlled free radical reactions as instruments of non-specific damage of cells and their membranes. Detailed analysis of two specific examples of phospholipid oxidative signaling in apoptosis and ferroptosis along with their molecular mechanisms and roles in reprogramming has been presented.


Assuntos
Ferroptose , Fosfolipídeos , Apoptose , Morte Celular , Oxirredução
3.
Free Radic Biol Med ; 133: 153-161, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30217775

RESUMO

Duality of iron as an essential cofactor of many enzymatic metabolic processes and as a catalyst of poorly controlled redox-cycling reactions defines its possible biological beneficial and hazardous role in the body. In this review, we discuss these two "faces" of iron in a newly conceptualized program of regulated cell death, ferroptosis. Ferroptosis is a genetically programmed iron-dependent form of regulated cell death driven by enhanced lipid peroxidation and insufficient capacity of thiol-dependent mechanisms (glutathione peroxidase 4, GPX4) to eliminate hydroperoxy-lipids. We present arguments favoring the enzymatic mechanisms of ferroptotically engaged non-heme iron of 15-lipoxygenases (15-LOX) in complexes with phosphatidylethanolamine binding protein 1 (PEBP1) as a catalyst of highly selective and specific oxidation reactions of arachidonoyl- (AA) and adrenoyl-phosphatidylethanolamines (PE). We discuss possible role of iron chaperons as control mechanisms for guided iron delivery directly to their "protein clients" thus limiting non-enzymatic redox-cycling reactions. We also consider opportunities of loosely-bound iron to contribute to the production of pro-ferroptotic lipid oxidation products. Finally, we propose a two-stage iron-dependent mechanism for iron in ferroptosis by combining its catalytic role in the 15-LOX-driven production of 15-hydroperoxy-AA-PE (HOO-AA-PE) as well as possible involvement of loosely-bound iron in oxidative cleavage of HOO-AA-PE to oxidatively truncated electrophiles capable of attacking nucleophilic targets in yet to be identified proteins leading to cell demise.


Assuntos
Ferroptose/genética , Radicais Livres/metabolismo , Ferro/metabolismo , Peroxidação de Lipídeos/genética , Animais , Araquidonato 15-Lipoxigenase/genética , Araquidonato 15-Lipoxigenase/metabolismo , Humanos , Oxirredução , Proteína de Ligação a Fosfatidiletanolamina/genética , Proteína de Ligação a Fosfatidiletanolamina/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo
4.
Biochemistry ; 47(51): 13699-710, 2008 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-19053260

RESUMO

Interaction of a mitochondria-specific anionic phospholipid, cardiolipin (CL), with an intermembrane protein, cytochrome c (cyt c), yields a peroxidase complex. During apoptosis, the complex induces accumulation of CL oxidation products that are essential for detachment of cyt c from the mitochondrial membrane, induction of permeability transition, and release of proapoptotic factors into the cytosol. Therefore, suppression of the peroxidase activity and prevention of CL oxidation may lead to discovery of new antiapoptotic drugs. Here, we report a new approach to regulate the cyt c peroxidase activity by using modified CL with an oxidizable and fluorescent 7-nitro-2,1,3-benzoxadiazole (NBD) moiety (NBD-CL). We demonstrate that NBD-CL forms high-affinity complexes with cyt c and blocks cyt c-catalyzed oxidation of several peroxidase substrates, cyt c self-oxidation, and, most importantly, inhibits cyt c-dependent oxidation of polyunsaturated tetralinoleoyl CL (TLCL) and accumulation of TLCL hydroperoxides. Electrospray ionization mass spectrometry and fluorescence analysis revealed that oxidation and cleavage of the NBD moiety of NBD-CL underlie the inhibition mechanism. We conclude that modified CL combining a nonoxidizable monounsaturated trioleoyl CL with a C(12)-NBD fragment undergoes a regiospecific oxidation thereby representing a novel inhibitor of cyt c peroxidase activity.


Assuntos
Apoptose , Cardiolipinas/química , Citocromos c/metabolismo , Oxidiazóis/química , Animais , Química Farmacêutica/métodos , Desenho de Fármacos , Corantes Fluorescentes/farmacologia , Cavalos , Humanos , Lipossomos/química , Miocárdio/metabolismo , Peroxidases/química , Espectrometria de Fluorescência/métodos , Espectrometria de Massas por Ionização por Electrospray
5.
Biochim Biophys Acta ; 1757(5-6): 648-59, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16740248

RESUMO

Since the (re)discovery of cytochrome c (cyt c) in the early 1920s and subsequent detailed characterization of its structure and function in mitochondrial electron transport, it took over 70 years to realize that cyt c plays a different, not less universal role in programmed cell death, apoptosis, by interacting with several proteins and forming apoptosomes. Recently, two additional essential functions of cyt c in apoptosis have been discovered that are carried out via its interactions with anionic phospholipids: a mitochondria specific phospholipid, cardiolipin (CL), and plasma membrane phosphatidylserine (PS). Execution of apoptotic program in cells is accompanied by substantial and early mitochondrial production of reactive oxygen species (ROS). Because antioxidant enhancements protect cells against apoptosis, ROS production was viewed not as a meaningless side effect of mitochondrial disintegration but rather playing some - as yet unidentified - role in apoptosis. This conundrum has been resolved by establishing that mitochondria contain a pool of cyt c, which interacts with CL and acts as a CL oxygenase. The oxygenase is activated during apoptosis, utilizes generated ROS and causes selective oxidation of CL. The oxidized CL is required for the release of pro-apoptotic factors from mitochondria into the cytosol. This redox mechanism of cyt c is realized earlier than its other well-recognized functions in the formation of apoptosomes and caspase activation. In the cytosol, released cyt c interacts with another anionic phospholipid, PS, and catalyzes its oxidation in a similar oxygenase reaction. Peroxidized PS facilitates its externalization essential for the recognition and clearance of apoptotic cells by macrophages. Redox catalysis of plasma membrane PS oxidation constitutes an important redox-dependent function of cyt c in apoptosis and phagocytosis. Thus, cyt c acts as an anionic phospholipid specific oxygenase activated and required for the execution of essential stages of apoptosis. This review is focused on newly discovered redox mechanisms of complexes of cyt c with anionic phospholipids and their role in apoptotic pathways in health and disease.


Assuntos
Citocromos c/metabolismo , Mitocôndrias/metabolismo , Fosfolipídeos/metabolismo , Sequência de Aminoácidos , Animais , Antioxidantes/metabolismo , Apoptose , Aterosclerose/metabolismo , Cardiolipinas/metabolismo , Membrana Celular/metabolismo , Transporte de Elétrons , Humanos , Membranas Mitocondriais/metabolismo , Dados de Sequência Molecular , Oxirredução , Oxigenases/metabolismo , Peroxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo
6.
Circ Res ; 88(11): 1210-5, 2001 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-11397789

RESUMO

The availability of nitric oxide (NO), which is required for the normal regulation of vascular tone, may be decreased in preeclampsia, thus contributing to the vascular pathogenesis of this pregnancy disorder. Because ascorbate is essential for the decomposition of S-nitrothiols and the release of NO, we speculated that the ascorbate deficiency typical of preeclampsia plasma might result in decreased rates of decomposition of S-nitrosothiols. We tested the hypothesis that total S-nitrosothiol and S-nitrosoalbumin concentrations are increased in preeclampsia plasma, reflecting a decreased release of NO from these major reservoirs of NO. Gestationally matched plasma samples were obtained (before labor or intravenous MgSO(4)) from 21 women with preeclampsia and 21 women with normal pregnancy, and plasma samples were also obtained from 12 nonpregnant women of similar age and body mass index during the follicular phase of the menstrual cycle. All were nonsmokers. The assay included ultraviolet-induced decomposition of S-nitrosothiols to liberate NO captured by a florigenic reagent, 4,5-diaminofluoresceine, to produce diaminofluoresceine-Triazole. Preeclampsia plasma contained significantly higher concentrations of total S-nitrosothiols (11.1+/-2.9 nmol/mL) than normal pregnancy samples (9.4+/-1.5 nmol/mL). Even greater differences were found between preeclampsia plasma and plasma samples from normal pregnancies and nonpregnant women (294+/-110, 186+/-25, and 151+/-25 pmol/mg protein, respectively) when S-nitrosothiol content was expressed per milligram protein. The albumin fraction contained 49.4% of total plasma S-nitrosothiols in the control samples and 53.7% and 56.8% of plasma S-nitrosothiols in normal pregnancy and preeclampsia, respectively. The level of S-nitrosoalbumin was significantly higher in preeclampsia than in normal pregnancy or nonpregnancy plasma (6.3+/-1.4, 5.1+/-0.7, and 4.2+/-1.0 nmol/mL, respectively). The increased concentration of S-nitrosoalbumin in preeclampsia almost completely accounted for the increased levels of S-nitrosothiols in total plasma. Due to combined increases in nitrosothiols and decreases in protein, the preeclampsia plasma concentration of S-nitrosoalbumin was greatly increased on a per milligram of protein basis (271% and 186% compared with normal nonpregnancy and normal pregnancy plasma, respectively). We conclude that S-nitrosoalbumin and total S-nitrosothiol concentrations are significantly increased in preeclampsia plasma and may reflect insufficient release of NO groups in this condition.


Assuntos
Mercaptoetanol , Óxido Nítrico/metabolismo , Compostos Nitrosos/sangue , Pré-Eclâmpsia/sangue , S-Nitrosotióis , Soroalbumina Bovina/metabolismo , Adulto , Análise de Variância , Deficiência de Ácido Ascórbico , Proteínas Sanguíneas/análise , Índice de Massa Corporal , Eletroforese em Gel de Poliacrilamida , Feminino , Corantes Fluorescentes , Fluorometria , Humanos , Estresse Oxidativo , Pré-Eclâmpsia/diagnóstico , Pré-Eclâmpsia/etiologia , Gravidez , Segundo Trimestre da Gravidez , Terceiro Trimestre da Gravidez , Valores de Referência , Sensibilidade e Especificidade , Albumina Sérica/análise , Albumina Sérica/metabolismo , Soroalbumina Bovina/análise
7.
Chem Biol Interact ; 163(1-2): 15-28, 2006 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-16797512

RESUMO

One of the prominent consequences of the symbiogenic origin of eukaryotic cells is the unique presence of one particular class of phospholipids, cardiolipin (CL), in mitochondria. As the product originated from the evolution of symbiotic bacteria, CL is predominantly confined to the inner mitochondrial membrane in normally functioning cells. Recent findings identified CL and its oxidation products as important participants and signaling molecules in the apoptotic cell death program. Early in apoptosis, massive membrane translocations of CL take place resulting in its appearance in the outer mitochondrial membrane. Consequently, significant amounts of CL become available for the interactions with cyt c, one of the major proteins of the intermembrane space. Binding of CL with cytochrome c (cyt c) yields the cyt c/CL complex that acts as a potent CL-specific peroxidase and generates CL hydroperoxides. In this review, we discuss the catalytic mechanisms of CL oxidation by the peroxidase activity of cyt c as well as the role of oxidized CL (CLox) in the release of pro-apoptotic factors from mitochondria into the cytosol. Potential implications of cyt c/CL peroxidase intracellular complexes in disease conditions (cancer, neurodegeneration) are also considered. The discovery of the new role of cyt c/CL complexes in early mitochondrial apoptosis offers interesting opportunities for new targets in drug discovery programs. Finally, exit of cyt c from damaged and/or dying (apoptotic) cells into extracellular compartments and its accumulation in biofluids is discussed in lieu of the formation of its peroxidase complexes with negatively charged lipids and their significance in the development of systemic oxidative stress in circulation.


Assuntos
Apoptose/fisiologia , Cardiolipinas/metabolismo , Citocromos c/metabolismo , Mitocôndrias Cardíacas/fisiologia , Transdução de Sinais/fisiologia , Animais , Humanos , Mitocôndrias Cardíacas/metabolismo , Membranas Mitocondriais/metabolismo , Oxirredução
8.
Toxicol Lett ; 165(1): 88-100, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16527436

RESUMO

Single-walled carbon nanotubes (SWCNT), nano-cylinders with an extremely small diameter (1-2 nm) and high aspect ratio, have unique physico-chemical, electronic and mechanical properties and may exhibit unusual interactions with cells and tissues, thus necessitating studies of their toxicity and health effects. Manufactured SWCNT usually contain significant amounts of iron that may act as a catalyst of oxidative stress. Because macrophages are the primary responders to different particles that initiate and propagate inflammatory reactions and oxidative stress, we utilized two types of SWCNT: (1) iron-rich (non-purified) SWCNT (26 wt.% of iron) and (2) iron-stripped (purified) SWCNT (0.23 wt.% of iron) to study their interactions with RAW 264.7 macrophages. Ultrasonication resulted in predominantly well-dispersed and separated SWCNT strands as evidenced by scanning electron microscopy. Neither purified nor non-purified SWCNT were able to generate intracellular production of superoxide radicals or nitric oxide in RAW 264.7 macrophages as documented by flow-cytometry and fluorescence microscopy. SWCNT with different iron content displayed different redox activity in a cell-free model system as revealed by EPR-detectable formation of ascorbate radicals resulting from ascorbate oxidation. In the presence of zymosan-stimulated RAW 264.7 macrophages, non-purified iron-rich SWCNT were more effective in generating hydroxyl radicals (documented by EPR spin-trapping with 5,5-dimethyl-1-pyrroline-N-oxide, DMPO) than purified SWCNT. Similarly, EPR spin-trapping experiments in the presence of zymosan-stimulated RAW 264.7 macrophages showed that non-purified SWCNT more effectively converted superoxide radicals generated by xanthine oxidase/xanthine into hydroxyl radicals as compared to purified SWCNT. Iron-rich SWCNT caused significant loss of intracellular low molecular weight thiols (GSH) and accumulation of lipid hydroperoxides in both zymosan-and PMA-stimulated RAW 264.7 macrophages. Catalase was able to partially protect macrophages against SWCNT induced elevation of biomarkers of oxidative stress (enhancement of lipid peroxidation and GSH depletion). Thus, the presence of iron in SWCNT may be important in determining redox-dependent responses of macrophages.


Assuntos
Ferro , Macrófagos Alveolares/efeitos dos fármacos , Nanotubos de Carbono/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Animais , Linhagem Celular , Citometria de Fluxo , Ferro/química , Macrófagos Alveolares/metabolismo , Camundongos , Microscopia de Fluorescência , Nanotubos de Carbono/química , Óxido Nítrico/metabolismo , Detecção de Spin , Superóxidos/metabolismo
9.
Cell Death Differ ; 23(7): 1140-51, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26742431

RESUMO

Mitophagy is critical for cell homeostasis. Externalization of the inner mitochondrial membrane phospholipid, cardiolipin (CL), to the surface of the outer mitochondrial membrane (OMM) was identified as a mitophageal signal recognized by the microtubule-associated protein 1 light chain 3. However, the CL-translocating machinery remains unknown. Here we demonstrate that a hexameric intermembrane space protein, NDPK-D (or NM23-H4), binds CL and facilitates its redistribution to the OMM. We found that mitophagy induced by a protonophoric uncoupler, carbonyl cyanide m-chlorophenylhydrazone (CCCP), caused externalization of CL to the surface of mitochondria in murine lung epithelial MLE-12 cells and human cervical adenocarcinoma HeLa cells. RNAi knockdown of endogenous NDPK-D decreased CCCP-induced CL externalization and mitochondrial degradation. A R90D NDPK-D mutant that does not bind CL was inactive in promoting mitophagy. Similarly, rotenone and 6-hydroxydopamine triggered mitophagy in SH-SY5Y cells was also suppressed by knocking down of NDPK-D. In situ proximity ligation assay (PLA) showed that mitophagy-inducing CL-transfer activity of NDPK-D is closely associated with the dynamin-like GTPase OPA1, implicating fission-fusion dynamics in mitophagy regulation.


Assuntos
Cardiolipinas/metabolismo , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Mitofagia , Nucleosídeo Difosfato Quinase D/metabolismo , Animais , Autofagia/efeitos dos fármacos , Carbonil Cianeto m-Clorofenil Hidrazona/toxicidade , Cardiolipinas/análise , Linhagem Celular , GTP Fosfo-Hidrolases/metabolismo , Células HeLa , Humanos , Lisossomos/metabolismo , Lisossomos/patologia , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/patologia , Mitofagia/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Nucleosídeo Difosfato Quinase D/antagonistas & inibidores , Nucleosídeo Difosfato Quinase D/genética , Oxidopamina/farmacologia , Ligação Proteica , Interferência de RNA , Rotenona/farmacologia
10.
Biochim Biophys Acta ; 1620(1-3): 72-84, 2003 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-12595076

RESUMO

Myeloperoxidase (MPO)-catalyzed one-electron oxidation of endogenous phenolic constituents (e.g., antioxidants, hydroxylated metabolites) and exogenous compounds (e.g., drugs, environmental chemicals) generates free radical intermediates: phenoxyl radicals. Reduction of these intermediates by endogenous reductants, i.e. recycling, may enhance their antioxidant potential and/or prevent their potential cytotoxic and genotoxic effects. The goal of this work was to determine whether generation and recycling of MPO-catalyzed phenoxyl radicals of a vitamin E homologue, 2,2,5,7,8-pentamethyl-6-hydroxychromane (PMC), by physiologically relevant intracellular reductants such as ascorbate/lipoate could be demonstrated in intact MPO-rich human leukemia HL-60 cells. A model system was developed to show that MPO/H(2)O(2)-catalyzed PMC phenoxyl radicals (PMC*) could be recycled by ascorbate or ascorbate/dihydrolipoic acid (DHLA) to regenerate the parent compound. Absorbance measurements demonstrated that ascorbate prevents net oxidation of PMC by recycling the phenoxyl radical back to the parent compound. The presence of DHLA in the reaction mixture containing ascorbate extended the recycling reaction through regeneration of ascorbate. DHLA alone was unable to prevent PMC oxidation. These conclusions were confirmed by direct detection of PMC* and ascorbate radicals formed during the time course of the reactions by EPR spectroscopy. Based on results in the model system, PMC* and ascorbate radicals were identified by EPR spectroscopy in ascorbate-loaded HL-60 cells after addition of H(2)O(2) and the inhibitor of catalase, 3-aminotriazole (3-AT). The time course of PMC* and ascorbate radicals was found to follow the same reaction sequence as during their recycling in the model system. Recycling of PMC by ascorbate was also confirmed by HPLC assays in HL-60 cells. Pre-loading of HL-60 cells with lipoic acid regenerated ascorbate and thus increased the efficiency of ascorbate in recycling PMC*. Lipoic acid had no effect on PMC oxidation in the absence of ascorbate. Thus PMC phenoxyl radical does not directly oxidize thiols but can be recycled by dihydrolipoate in the presence of ascorbate. The role of phenoxyl radical recycling in maintaining antioxidant defense and protecting against cytotoxic and genotoxic phenolics is discussed.


Assuntos
Ácido Ascórbico/metabolismo , Cromanos/metabolismo , Radicais Livres/metabolismo , Peroxidase/metabolismo , Ácido Tióctico/análogos & derivados , Ácido Tióctico/metabolismo , Antioxidantes/metabolismo , Sobrevivência Celular , Cromatografia Líquida de Alta Pressão , Espectroscopia de Ressonância de Spin Eletrônica , Células HL-60 , Humanos , Peróxido de Hidrogênio/farmacologia , Oxirredução , Fenóis/metabolismo , Espectrofotometria , Ciclização de Substratos/efeitos dos fármacos
11.
Free Radic Res ; 49(5): 681-91, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25740198

RESUMO

Exposure to rotenone in vivo results in selective degeneration of dopaminergic neurons and development of neuropathologic features of Parkinson's disease (PD). As rotenone acts as an inhibitor of mitochondrial respiratory complex I, we employed oxidative lipidomics to assess oxidative metabolism of a mitochondria-specific phospholipid, cardiolipin (CL), in substantia nigra (SN) of exposed animals. We found a significant reduction in oxidizable polyunsaturated fatty acid (PUFA)-containing CL molecular species. We further revealed increased contents of mono-oxygenated CL species at late stages of the exposure. Notably, linoleic acid in sn-1 position was the major oxidation substrate yielding its mono-hydroxy- and epoxy-derivatives whereas more readily "oxidizable" fatty acid residues (arachidonic and docosahexaenoic acids) remained non-oxidized. Elevated levels of PUFA CLs were detected in plasma of rats exposed to rotenone. Characterization of oxidatively modified CL molecular species in SN and detection of PUFA-containing CL species in plasma may contribute to better understanding of the PD pathogenesis and lead to the development of new biomarkers of mitochondrial dysfunction associated with this disease.


Assuntos
Cardiolipinas/metabolismo , Cromatografia Líquida de Alta Pressão , Espectrometria de Massas , Mitocôndrias/metabolismo , Transtornos Parkinsonianos/metabolismo , Rotenona , Substância Negra/metabolismo , Animais , Ácido Araquidônico/metabolismo , Biomarcadores/metabolismo , Cardiolipinas/sangue , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/metabolismo , Ácido Linoleico/metabolismo , Masculino , Oxirredução , Transtornos Parkinsonianos/sangue , Transtornos Parkinsonianos/induzido quimicamente , Ratos Endogâmicos Lew , Fatores de Tempo
12.
J Invest Dermatol ; 114(2): 354-64, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10651998

RESUMO

A variety of phenolic compounds are utilized for industrial production of phenol-formaldehyde resins, paints, lacquers, cosmetics, and pharmaceuticals. Skin exposure to industrial phenolics is known to cause skin rash, dermal inflammation, contact dermatitis, leucoderma, and cancer promotion. The biochemical mechanisms of cytotoxicity of phenolic compounds are not well understood. We hypothesized that enzymatic one-electron oxidation of phenolic compounds resulting in the generation of phenoxyl radicals may be an important contributor to the cytotoxic effects. Phenoxyl radicals are readily reduced by thiols, ascorbate, and other intracellular reductants (e.g., NADH, NADPH) regenerating the parent phenolic compound. Hence, phenolic compounds may undergo enzymatically driven redox-cycling thus causing oxidative stress. To test the hypothesis, we analyzed endogenous thiols, lipid peroxidation, and total antioxidant reserves in normal human keratinocytes exposed to phenol. Using a newly developed cis-parinaric acid-based procedure to assay site-specific oxidative stress in membrane phospholipids, we found that phenol at subtoxic concentrations (50 microM) caused oxidation of phosphatidylcholine and phosphatidylethanolamine (but not of phosphatidylserine) in keratinocytes. Phenol did not induce peroxidation of phospholipids in liposomes prepared from keratinocyte lipids labeled by cis-parinaric acid. Measurements with ThioGlo-1 showed that phenol depleted glutathione but did not produce thiyl radicals as evidenced by our high-performance liquid chromatography measurements of GS.-5, 5-dimethyl1pyrroline N-oxide nitrone. Additionally, phenol caused a significant decrease of protein SH groups. Luminol-enhanced chemiluminescence assay demonstrated a significant decrease in total antioxidant reserves of keratinocytes exposed to phenol. Incubation of ascorbate-preloaded keratinocytes with phenol produced an electron paramagnetic resonance-detectable signal of ascorbate radicals, suggesting that redox-cycling of one-electron oxidation products of phenol, its phenoxyl radicals, is involved in the oxidative effects. As no cytotoxicity was observed in keratinocytes exposed to 50 microM or 500 microM phenol, we conclude that phenol at subtoxic concentrations causes significant oxidative stress.


Assuntos
Ácido Ascórbico/farmacologia , Queratinócitos/metabolismo , Antioxidantes/análise , Apoptose/efeitos dos fármacos , Compostos Azo/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Óxidos N-Cíclicos/análise , Espectroscopia de Ressonância de Spin Eletrônica , Ácidos Graxos Insaturados/análise , Corantes Fluorescentes/análise , Radicais Livres , Glutationa/análogos & derivados , Glutationa/análise , Humanos , Queratinócitos/química , Queratinócitos/efeitos dos fármacos , Microscopia Eletrônica , Nitrilas/farmacologia , Organelas/ultraestrutura , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Fenol/farmacologia , Fenóis/metabolismo , Fenóis/farmacologia , Fosfolipídeos/análise , Fosfolipídeos/isolamento & purificação , Marcadores de Spin , Compostos de Sulfidrila/análise
13.
FEBS Lett ; 428(1-2): 43-6, 1998 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-9645471

RESUMO

We investigated the ability of plasma membrane CoQ reductase (PMQR) purified from pig liver to reduce phenoxyl radicals of a vitamin E homologue, Trolox. We report that NADH-driven one-electron reduction of CoQ0 catalyzed by PMQR produced CoQ0 semiquinone radical and CoQoH2. These in turn, recycle vitamin E homologue, Trolox, via reducing its phenoxyl radical. A significant part of NADH/PMQR-catalyzed reduction of CoQ0 (and Trolox recycling) was superoxide-dependent. Overall, our results demonstrate that PMQR in the model system used can act as an antioxidant enzyme that recycles water-soluble homologues of coenzyme Q and vitamin E.


Assuntos
Benzoquinonas/metabolismo , NADH NADPH Oxirredutases/metabolismo , Superóxidos/metabolismo , Vitamina E/metabolismo , Animais , Catálise , Membrana Celular , Cromanos/metabolismo , Complexo I de Transporte de Elétrons , Ácido Linoleico/metabolismo , Lipoxigenase/metabolismo , Oxirredução , Fenóis , Suínos
14.
Free Radic Biol Med ; 9(2): 117-26, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2227528

RESUMO

Ubiquinones (CoQn) are intrinsic lipid components of many membranes. Besides their role in electron-transfer reactions they may act as free radical scavengers, yet their antioxidant function has received relatively little study. The efficiency of ubiquinols of varying isoprenoid chain length (from Q0 to Q10) in preventing (Fe2+ + ascorbate)-dependent or (Fe2+ + NADPH)-dependent lipid peroxidation was investigated in rat liver microsomes and brain synaptosomes and mitochondria. Ubiquinols, the reduced forms of CoQn, possess much greater antioxidant activity than the oxidized ubiquinone forms. In homogenous solution the radical scavenging activity of ubiquinol homologues does not depend on the length of their isoprenoid chain. However in membranes ubiquinols with short isoprenoid chains (Q1-Q4) are much more potent inhibitors of lipid peroxidation than the longer chain homologues (Q5-Q10). It is found that: i) the inhibitory action, that is, antioxidant efficiency of short-chain ubiquinols decreases in order Q1 greater than Q2 greater than Q3 greater than Q4; ii) the antioxidant efficiency of long-chain ubiquinols is only slightly dependent on their concentrations in the order Q5 greater than Q6 greater than Q7 greater than Q8 greater than Q9 greater than Q10 and iii) the antioxidant efficiency of Q0 is markedly less than that of other homologues. Interaction of ubiquinols with oxygen radicals was followed by their effects on luminol-activated chemiluminescence. Ubiquinols Q1-Q4 at 0.1 mM completely inhibit the luminol-activated NADPH-dependent chemiluminescent response of microsomes, while homologues Q6-Q10 exert no effect. In contrast to ubiquinol Q10 (ubiquinone Q10) ubiquinone Q1 synergistically enhances NADPH-dependent regeneration of endogenous vitamin E in microsomes thus providing for higher antioxidant protection against lipid peroxidation. The differences in the antioxidant potency of ubiquinols in membranes are suggested to result from differences in partitioning into membranes, intramembrane mobility and non-uniform distribution of ubiquinols resulting in differing efficiency of interaction with oxygen and lipid radicals as well as different efficiency of ubiquinols in regeneration of endogenous vitamin E.


Assuntos
Antioxidantes , Microssomos Hepáticos/metabolismo , Terpenos/química , Ubiquinona/farmacologia , Vitamina E/metabolismo , Animais , Transporte Biológico , Sistema Enzimático do Citocromo P-450/metabolismo , Radicais Livres , Peroxidação de Lipídeos , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , NADP/farmacologia , Oxirredução , Oxigênio/metabolismo , Ratos , Ratos Endogâmicos , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/enzimologia , Sinaptossomos/metabolismo
15.
FEBS Lett ; 477(1-2): 1-7, 2000 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-10899301

RESUMO

Active maintenance of membrane phospholipid asymmetry is universal in normal cell membranes and its disruption with subsequent externalization of phosphatidylserine is a hallmark of apoptosis. Externalized phosphatidylserine appears to serve as an important signal for targeting recognition and elimination of apoptotic cells by macrophages, however, the molecular mechanisms responsible for phosphatidylserine translocation during apoptosis remain unresolved. Studies have focused on the function of aminophospholipid translocase and phospholipid scramblase as mediators of this process. Here we present evidence that unique oxidative events, represented by selective oxidation of phosphatidylserine, occur during apoptosis that could promote phosphatidylserine externalization. We speculate that selective phosphatidylserine oxidation could affect phosphatidylserine recognition by aminophospholipid translocase and/or directly result in enzyme inhibition. The potential interactions between the anionic phospholipid phosphatidylserine and the redox-active cationic protein effector of apoptosis, cytochrome c, are presented as a potential mechanism to account for selective oxidation of phosphatidylserine during apoptosis. Thus, cytochrome c-mediated phosphatidylserine oxidation may represent an important component of the apoptotic pathway.


Assuntos
Apoptose , Membrana Celular/metabolismo , Estresse Oxidativo/fisiologia , Fosfatidilserinas/metabolismo , Transdução de Sinais , Animais , Grupo dos Citocromos c/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Oxidantes/metabolismo , Especificidade por Substrato
16.
Free Radic Biol Med ; 27(9-10): 1050-63, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10569638

RESUMO

Various types of cancer occur in peroxidase-rich target tissues of animals exposed to aryl alcohols and amines. Unlike biotransformation by cytochrome P450 enzymes, peroxidases activate most substrates by one-electron oxidation via radical intermediates. This work analyzed the peroxidase-dependent formation of phenoxyl radicals in HL-60 cells and its contribution to cytotoxicity and genotoxicity. The results showed that myeloperoxidase-catalyzed redox cycling of phenol in HL-60 cells led to intracellular formation of glutathionyl radicals detected as GS-DMPO nitrone. Formation of thiyl radicals was accompanied by rapid oxidation of glutathione and protein-thiols. Analysis of protein sulfhydryls by SDS-PAGE revealed a significant oxidation of protein SH-groups in HL-60 cells incubated in the presence of phenol/H2O2 that was inhibited by cyanide and azide. Additionally, cyanide- and azide-sensitive generation of EPR-detectable ascorbate radicals was observed during incubation of HL-60 cell homogenates in the presence of ascorbate and H2O2. Oxidation of thiols required addition of H2O2 and was inhibited by pretreatment of cells with the inhibitor of heme synthesis, succinylacetone. Radical-driven oxidation of thiols was accompanied by a trend toward increased content of 8-oxo-7,8-dihydro-2'-deoxyguanosine in the DNA of HL-60 cells. Membrane phospholipids were also sensitive to radical-driven oxidation as evidenced by a sensitive fluorescence HPLC-assay based on metabolic labeling of phospholipids with oxidation-sensitive cis-parinaric acid. Phenol enhanced H2O2-dependent oxidation of all classes of phospholipids including cardiolipin, but did not oxidize parinaric acid-labeled lipids without addition of H2O2. Induction of a significant hypodiploid cell population, an indication of apoptosis, was detected after exposure to H2O2 and was slightly but consistently and significantly higher after exposure to H2O2/phenol. The clonogenicity of HL-60 cells decreased to the same extent after exposure to H2O2 or H2O2/phenol. Treatment of HL-60 cells with either H2O2 or H2O2/phenol at concentrations adequate for lipid peroxidation did not cause a detectable increase in chromosomal breaks. Detection of thiyl radicals as well as rapid oxidation of thiols and phospholipids in viable HL-60 cells provide strong evidence for redox cycling of phenol in this bone marrow-derived cell line.


Assuntos
Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidase/metabolismo , Fenol/metabolismo , Fenol/toxicidade , Compostos de Sulfidrila/metabolismo , 8-Hidroxi-2'-Desoxiguanosina , Apoptose/efeitos dos fármacos , Ácido Ascórbico/metabolismo , Dano ao DNA , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Radicais Livres/metabolismo , Células HL-60 , Humanos , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/toxicidade , Oxirredução , Estresse Oxidativo , Fosfolipídeos/metabolismo , Cianeto de Potássio/farmacologia , Azida Sódica/farmacologia , Especificidade por Substrato
17.
Antioxid Redox Signal ; 2(2): 251-62, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11229530

RESUMO

High affinity for NADH, and low affinity for NADPH, for reduction of endogenous coenzyme Q10 (CoQ10) by pig liver plasma membrane is reported in the present work. CoQ reduction in plasma membrane is carried out, in addition to other mechanisms, by plasma membrane coenzyme Q reductase (PMQR). We show that PMQR-catalyzed reduction of CoQ0 by both NADH and NADPH is accompanied by generation of CoQ0 semiquinone radicals in a superoxide-dependent reaction. In the presence of a water-soluble vitamin E homologue, Trolox, this reduction leads to quenching of the Trolox phenoxyl radicals. The involvement of PMQR versus DT-diaphorase under the conditions of vitamin E and selenium sufficiency and deficiency was evaluated for CoQ reduction by plasma membranes. The data presented here suggest that both nucleotides (NADH and NADPH) can be accountable for CoQ reduction by PMQR on the basis of their physiological concentrations within the cell. The enzyme is primarily responsible for CoQ reduction in plasma membrane under normal (nonoxidative stress-associated) conditions.


Assuntos
Membrana Celular/enzimologia , Fígado/enzimologia , NADP/metabolismo , NAD/metabolismo , Ubiquinona/metabolismo , Animais , Antioxidantes/farmacologia , Cromanos/farmacologia , Coenzimas , Grupo dos Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Espectroscopia de Ressonância de Spin Eletrônica , Transporte de Elétrons , Cinética , Masculino , Estresse Oxidativo , Ratos , Ratos Long-Evans , Selênio/metabolismo , Superóxidos/metabolismo , Suínos , Ubiquinona/análogos & derivados , Vitamina E/metabolismo
18.
Antioxid Redox Signal ; 1(3): 349-64, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-11229446

RESUMO

Metallothioneins (MTs) are cysteine-rich metal-binding proteins that exert cytoprotection during metal exposure and oxidative stress. The roles of MT in copper (Cu) binding and release and modulation of redox cycling are unresolved. We hypothesized that Cu-binding to MT renders Cu redox inactive, but that oxidation of free thiols critical for metal binding can reduce MT/Cu interactions and potentiate Cu redox cycling. Overexpression of MT in cells by cadmium pretreatment or ectopic overexpression by gene transfer confers protection from Cu-dependent lipid oxidation and cytotoxicity. Using a chemically defined model system (Cu/ascorbate/H2O2) to study Cu/MT interactions, we observed that MT inhibited Cu-dependent oxidation of luminol. In the absence of H2O2, MT blocked Cu-dependent ascorbyl radical production with a stoichiometry corresponding to Cu/MT ratios < or = 12. In the presence of H2O2, Cu-dependent hydroxyl radical formation was inhibited only up to Cu/MT ratios < or = 6. Using low-temperature EPR of free Cu2+ to assess Cu/MT physical interactions, we observed that the maximal amount of Cu1+ bound to MT corresponded to 12 molar equivalents of Cu/MT with Cu and ascorbate alone and was reduced in the presence of H2O2. 2,2'-Dithiodipyridine titration of MT SH-groups revealed a 50% decrease after H2O2, which could be regenerated by dihydrolipoic acid (DHLA). DHLA regeneration of thiols in MT was accompanied by restoration of MT's ability to inhibit Cu-dependent oxidation of ascorbate. Thus, optimum ability of MT to inhibit Cu-redox cycling directly correlates with its ability to bind Cu. Some of this Cu, however, appears releasable following oxidation of the thiolate metal-binding clusters. We speculate that redox-dependent release of Cu from MT serves both as a mechanism for physiological delivery of Cu to specific target proteins, as well as potentiation of cellular damage during oxidative stress.


Assuntos
Antioxidantes/metabolismo , Cobre/metabolismo , Cobre/farmacologia , Metalotioneína/metabolismo , Oxidantes/metabolismo , Ácido Tióctico/análogos & derivados , Animais , Antioxidantes/farmacologia , Cádmio/farmacologia , Sobrevivência Celular , Cobre/antagonistas & inibidores , Cobre/toxicidade , Espectroscopia de Ressonância de Spin Eletrônica , Endotélio Vascular/efeitos dos fármacos , Células HL-60 , Humanos , Peróxido de Hidrogênio/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado , Luminol/metabolismo , Metalotioneína/genética , Metalotioneína/farmacologia , Oxidantes/farmacologia , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Coelhos , Compostos de Sulfidrila/metabolismo , Ácido Tióctico/farmacologia , Transfecção
19.
Biochem Pharmacol ; 54(8): 937-45, 1997 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-9354594

RESUMO

The antifungal activity of amphotericin B (AmB) and its side-effects (e.g. nephrotoxicity and hemolytic action) are suggested to be associated with its prooxidant effects in target cells. To test this hypothesis, we have undertaken studies to examine the role of AmB in oxidative stress in cultured rat aortic smooth muscle cells (SMC) incubated in the absence or in the presence of a lipid-soluble azo-initiator of peroxyl radicals, 2,2'-azobis(2,4-dimethylvaleronitrile) (AMVN). No changes in the pattern of membrane phospholipids could be detected by two-dimensional high performance thin-layer chromatography (HPTLC) after oxidative stress induced by AMVN in which the cells remained viable, as judged by trypan blue exclusion. To improve the sensitivity of detection of oxidative stress in the cells, cis-parinaric acid (PnA) was incorporated biosynthetically into the membrane phospholipids [using PnA-human serum albumin (hSA) complex]. Incubation of the cells under aerobic conditions in the presence of up to 10 microM AmB showed no significant change in the pattern of PnA-labeled phospholipids, suggesting that AmB was not affecting the oxidative state of the cells. In contrast, treatment with AMVN (0.5 mM, incubation in the dark for 2 hr at 37 degrees--conditions in which the viability of the cells was maintained) caused a significant reduction of all fluorescently labeled phospholipid fractions separated by HPLC. When PnA-labeled cells were subjected to oxidative stress by incubation with 0.5 mM AMVN in the presence of AmB, the loss of fluorescent phospholipids was reduced in a concentration-dependent manner over a concentration range of 0.25 to 10 microM. Thus, AmB does not produce any prooxidant effect but rather acts as an intracellular antioxidant.


Assuntos
Anfotericina B/metabolismo , Antioxidantes , Compostos Azo/antagonistas & inibidores , Peróxidos Lipídicos/metabolismo , Lipídeos de Membrana/metabolismo , Nitrilas/antagonistas & inibidores , Fosfolipídeos/metabolismo , Animais , Aorta , Compostos Azo/farmacologia , Ácidos Graxos Insaturados/metabolismo , Músculo Liso Vascular/metabolismo , Nitrilas/farmacologia , Estresse Oxidativo , Ratos , Ratos Sprague-Dawley
20.
Biochem Pharmacol ; 57(9): 989-1001, 1999 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10796069

RESUMO

The goal of the present work was to determine whether nitric oxide (NO) released from different donors (NONOates and nitrosothiols) can act as a protective antioxidant against oxidative stress and cytotoxicity induced by extracellular hemoglobin/tert-butyl hydroperoxide (Hb/tert-BuOOH) in vascular smooth muscle cells (VSMCs). No changes in phospholipid composition were found in VSMCs incubated with oxyhemoglobin (oxyHb)/tert-BuOOH. Using our newly developed HPLC-fluorescence technique for measurement of site-specific oxidative stress in membrane phospholipids, we produced VSMCs in which endogenous phospholipids were metabolically labeled with an oxidation-sensitive fluorescent fatty acid, cis-parinaric acid. In these cells, we were able to reliably quantitate oxidative stress in major phospholipid classes-phosphatidylethanolamine, phosphatidylcholine, phosphatidylserine, and phosphatidylinositol-induced by tert-BuOOH in the presence of oxyHb or methemoglobin (metHb). The oxidative stress was accompanied by cytotoxic effects of oxyHb/tert-BuOOH and metHb/tert-BuOOH on VSMCs. We further found that an NO donor, (Z)-1-[N-(3-ammoniopropyl)-N-(n-propyl)amino]diazen 1-ium-1,2-diolate (PAPANONO), but not nitrosothiols, protected VSMCs against oxidative stress and cytotoxicity induced by Hb/tert-BuOOH. The protective effect of PAPANONO was most likely due to its ability to form NO-heme Hb (detectable by low temperature EPR spectroscopy and visible spectrophotometry). These findings are important for further understanding the physiological antioxidant role of NO against oxidative stress induced by hemoproteins as well as for pathological hypertensive events induced by extracellular Hb via NO depletion.


Assuntos
Hemoglobinas/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Óxido Nítrico/metabolismo , Estresse Oxidativo/efeitos dos fármacos , terc-Butil Hidroperóxido/farmacologia , Animais , Azetidinas/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Interações Medicamentosas , Peroxidação de Lipídeos/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Doadores de Óxido Nítrico/farmacologia , Oxiemoglobinas/metabolismo , Fosfolipídeos/metabolismo , Substâncias Protetoras/farmacologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA