Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Arterioscler Thromb Vasc Biol ; 35(10): 2104-13, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26315403

ABSTRACT

OBJECTIVE: Oxidative stress associated with cardiovascular disease can produce various oxidized lipids, including cholesterol oxides, such as 7-hydroperoxide (7-OOH), 7-hydroxide (7-OH), and 7-ketone (7=O). Unlike 7=O and 7-OH, 7-OOH is redox active, giving rise to the others via potentially toxic-free radical reactions. We tested the novel hypothesis that under oxidative stress conditions, steroidogenic acute regulatory (StAR) family proteins not only deliver cholesterol to/into mitochondria of vascular macrophages, but also 7-OOH, which induces peroxidative damage that impairs early stage reverse cholesterol transport. APPROACH AND RESULTS: Stimulation of human monocyte-derived THP-1 macrophages with dibutyryl-cAMP resulted in substantial upregulation of StarD1 and ATP-binding cassette (ABC) transporter, ABCA1. Small interfering RNA-induced StarD1 knockdown before stimulation had no effect on StarD4, but reduced ABCA1 upregulation, linking the latter to StarD1 functionality. Mitochondria in stimulated StarD1-knockdown cells internalized 7-OOH slower than nonstimulated controls and underwent less 7-OOH-induced lipid peroxidation and membrane depolarization, as probed with C11-BODIPY (4,4-difluoro-5-(4-phenyl-1,3-butadienyl)-4-bora-3a,4a-diaza-s-inda-cene-3-undecanoic acid) and JC-1 (5,5',6,6'-tetrachloro-1,1',3,3'-tetraethyl-benzimidazolylcarbocyanine iodide), respectively. Major functional consequences of 7-OOH exposure were (1) loss of mitochondrial CYP27A1 activity, (2) reduced 27-hydroxycholesterol (27-OH) output, and (3) downregulation of cholesterol-exporting ABCA1 and ABCG1. Consistently, 7-OOH-challenged macrophages exported less cholesterol to apoA-I or high-density lipoprotein than did nonchallenged controls. StarD1-mediated 7-OOH transport was also found to be highly cytotoxic, whereas 7=O and 7-OH were minimally toxic. CONCLUSIONS: This study describes a previously unrecognized mechanism by which macrophage cholesterol efflux can be incapacitated under oxidative stress-linked disorders, such as chronic obesity and hypertension. Our findings provide new insights into the role of macrophage redox damage/dysfunction in atherogenesis.


Subject(s)
Atherosclerosis/metabolism , Cholesterol/analogs & derivatives , Lipid Peroxidation/physiology , Macrophages/metabolism , Oxidative Stress/physiology , Biological Transport , Cells, Cultured , Cholesterol/metabolism , Cyclic CMP/analogs & derivatives , Cyclic CMP/pharmacology , Humans , Macrophages/cytology , Mitochondria/metabolism , Protein Transport , Sensitivity and Specificity
2.
FEBS Lett ; 588(1): 65-70, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24269887

ABSTRACT

StAR family proteins in vascular macrophages participate in reverse cholesterol transport (RCT). We hypothesize that under pathophysiological oxidative stress, StARs will transport not only cholesterol to macrophage mitochondria, but also pro-oxidant cholesterol hydroperoxides (7-OOHs), thereby impairing early-stage RCT. Upon stimulation with dibutyryl-cAMP, RAW264.7 macrophages exhibited a strong time-dependent induction of mitochondrial StarD1 and plasma membrane ABCA1, which exports cholesterol. 7α-OOH uptake by stimulated RAW cell mitochondria (like cholesterol uptake) was strongly reduced by StarD1 knockdown, consistent with StarD1 involvement. Upon uptake by mitochondria, 7α-OOH (but not redox-inactive 7α-OH) triggered lipid peroxidation and membrane depolarization while reducing ABCA1 upregulation. These findings provide strong initial support for our hypothesis.


Subject(s)
Cholesterol/analogs & derivatives , Macrophages/metabolism , Mitochondria/metabolism , Phosphoproteins/metabolism , ATP Binding Cassette Transporter 1/metabolism , Animals , Biological Transport/drug effects , Blotting, Western , Bucladesine/pharmacology , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Cholesterol/metabolism , Cholesterol/pharmacology , Dose-Response Relationship, Drug , Lipid Peroxidation/drug effects , Macrophages/cytology , Macrophages/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Oxidative Stress , Phosphoproteins/genetics , RNA Interference , Time Factors
3.
J Biol Chem ; 288(16): 11509-19, 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-23467407

ABSTRACT

Steroidogenic acute regulatory (StAR) proteins in steroidogenic cells are implicated in the delivery of cholesterol (Ch) from internal or external sources to mitochondria (Mito) for initiation of steroid hormone synthesis. In this study, we tested the hypothesis that under oxidative stress, StAR-mediated trafficking of redox-active cholesterol hydroperoxides (ChOOHs) can result in site-specific Mito damage and dysfunction. Steroidogenic stimulation of mouse MA-10 Leydig cells with dibutyryl-cAMP (Bt2cAMP) resulted in strong expression of StarD1 and StarD4 proteins over insignificant levels in nonstimulated controls. During incubation with the ChOOH 3ß-hydroxycholest-5-ene-7α-hydroperoxide (7α-OOH) in liposomes, stimulated cells took up substantially more hydroperoxide in Mito than controls, with a resulting loss of membrane potential (ΔΨm) and ability to drive progesterone synthesis. 7α-OOH uptake and ΔΨm loss were greatly reduced by StarD1 knockdown, thus establishing the role of this protein in 7α-OOH delivery. Moreover, 7α-OOH was substantially more toxic to stimulated than nonstimulated cells, the former dying mainly by apoptosis and the latter dying by necrosis. Importantly, tert-butyl hydroperoxide, which is not a StAR protein ligand, was equally toxic to stimulated and nonstimulated cells. These findings support the notion that like Ch itself, 7α-OOH can be transported to/into Mito of steroidogenic cells by StAR proteins and therein induce free radical damage, which compromises steroid hormone synthesis.


Subject(s)
Cholesterol/analogs & derivatives , Gene Expression Regulation/drug effects , Leydig Cells/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mutagens/pharmacology , Phosphoproteins/biosynthesis , Animals , Biological Transport, Active/drug effects , Cell Line , Cholesterol/pharmacology , Gonadal Steroid Hormones/biosynthesis , Leydig Cells/pathology , Male , Mice , Mitochondria/pathology
4.
J Lipid Res ; 51(11): 3174-84, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20656919

ABSTRACT

Sterol carrier protein-2 (SCP-2) plays an important role in cholesterol trafficking and metabolism in mammalian cells. The purpose of this study was to determine whether SCP-2, under oxidative stress conditions, might also traffic hydroperoxides of cholesterol, thereby disseminating their cytotoxic effects. Two inhibitors, SCPI-1 and SCPI-3, known to block cholesterol binding by an insect SCP-2, were used to investigate this. A mouse fibroblast transfectant clone (SC2F) overexpressing SCP-2 was found to be substantially more sensitive to apoptotic killing induced by liposomal 7α-hydroperoxycholesterol (7α-OOH) than a wild-type control. 7α-OOH uptake by SC2F cells and resulting apoptosis were both inhibited by SCPI-1 or SCPI-3 at a subtoxic concentration. Preceding cell death, reactive oxidant accumulation and loss of mitochondrial membrane potential were also strongly inhibited. Similar SCPI protection against 7α-OOH was observed with two other types of SCP-2-expressing mammalian cells. In striking contrast, neither inhibitor had any effect on H(2)O(2)-induced cell killing. To learn whether 7α-OOH cytotoxicity is due to uptake/transport by SCP-2, we used a fluorescence-based competitive binding assay involving recombinant SCP-2, NBD-cholesterol, and SCPI-1/SCPI-3 or 7α-OOH. The results clearly showed that 7α-OOH binds to SCP-2 in SCPI-inhibitable fashion. Our findings suggest that cellular SCP-2 not only binds and translocates cholesterol but also cholesterol hydroperoxides, thus expanding their redox toxicity and signaling ranges under oxidative stress conditions.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cholesterol/analogs & derivatives , Acetanilides/metabolism , Acetanilides/pharmacology , Animals , Apoptosis/drug effects , Benzoxazines/chemistry , Benzoxazines/metabolism , Benzoxazines/pharmacology , Biological Transport/drug effects , Cell Line , Cholesterol/metabolism , Cholesterol/toxicity , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/toxicity , Membrane Potential, Mitochondrial/drug effects , Mice , Rats , Reactive Oxygen Species/metabolism , Thiazoles/metabolism , Thiazoles/pharmacology
5.
Photochem Photobiol ; 86(4): 747-51, 2010.
Article in English | MEDLINE | ID: mdl-20408976

ABSTRACT

Singlet oxygen attack on cholesterol (Ch), a prominent monounsaturated lipid of mammalian cell plasma membranes, gives rise to three hydroperoxide (ChOOH) isomers, 5alpha-OOH, 6alpha-OOH and 6beta-OOH, the latter two in lower yield than 5alpha-OOH, and 6alpha-OOH in lowest yield. A third possible positional isomer, 7alpha-OOH and 7beta-OOH, is produced by free radical attack. In the presence of iron and ascorbate (Fe/AH), 5alpha-OOH or 6beta-OOH in phosphatidylcholine/Ch/ChOOH (20:15:1 by mol) liposomes was reduced to its corresponding alcohol, the rate constant being approximately the same for both ChOOHs. Using [(14)C]Ch as an in situ probe, we found that liposomal 5alpha-OOH readily set off free radical-mediated (chain) peroxidation reactions when exposed to Fe/AH, whereas 6beta-OOH under the same conditions did not. Moreover, liposomal 5alpha-OOH triggered robust chain peroxidation in [(14)C]Ch-labeled L1210 cells, leading to cell death, whereas 6beta-OOH was essentially inert in this regard. Thus, 5alpha-OOH and 6beta-OOH undergo iron-catalyzed reductive turnover, but only the former can provoke toxic membrane damage. These novel findings have important implications for UVA-induced photodamage in Ch-rich tissues like skin and eye, where (1)O(2) often plays a major role.


Subject(s)
Cell Membrane/chemistry , Lipid Peroxidation , Singlet Oxygen/chemistry , Animals , Cell Membrane/metabolism , Cell Membrane/radiation effects , Cholesterol/analogs & derivatives , Cholesterol/chemistry , Free Radicals/chemistry , Free Radicals/metabolism , Liposomes/chemistry , Liposomes/metabolism , Liposomes/radiation effects , Mice , Peroxides/chemistry , Photochemistry , Singlet Oxygen/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...