Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Cancer Res ; 83(13): 2091-2092, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37403629

ABSTRACT

Statins are a class of cholesterol-lowering drugs that inhibit 3-hydroxy-3-methylglutaryl-CoA reductase, the rate-limiting enzyme of the mevalonate pathway. Evidence suggests that certain cancers depend on the mevalonate pathway for growth and survival, and thus blocking the mevalonate pathway with statins may offer a viable therapeutic approach for treating cancer, or at least enhance the efficacy of existing cancer drugs. In this issue of Cancer Research, Tran and colleagues showed that caffeine works jointly with FOXM1 inhibition to enhance the antitumor activity of statins in neuroblastoma cells. They found that caffeine synergizes with statins by suppressing statin-induced feedback activation of the mevalonate pathway. Here, we reflect on the potential of combining caffeine and statin drugs as a strategy for potentiating anticancer activity. See related article by Tran et al., p. 2248.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Neuroblastoma , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Caffeine/pharmacology , Mevalonic Acid/metabolism , Drug Repositioning , Friends , Neuroblastoma/drug therapy , Dietary Supplements , Forkhead Box Protein M1
2.
J Lipid Res ; 56(5): 1025-33, 2015 May.
Article in English | MEDLINE | ID: mdl-25773887

ABSTRACT

Atherosclerosis stems from imbalances in lipid metabolism and leads to maladaptive inflammatory responses. The AMP-activated protein kinase (Ampk) is a highly conserved serine/threonine kinase that regulates many aspects of lipid and energy metabolism, although its specific role in controlling macrophage cholesterol homeostasis remains unclear. We sought to address this question by testing the effects of direct Ampk activators in primary bone marrow-derived macrophages from Ampk ß1-deficient (ß1(-/-)) mice. Macrophages from Ampk ß1(-/-) mice had enhanced lipogenic capacity and diminished cholesterol efflux, although cholesterol uptake was unaffected. Direct activation of Ampk ß1 via salicylate (the unacetylated form of aspirin) or A-769662 (a small molecule activator), decreased the synthesis of FAs and sterols in WT but not Ampk ß1(-/-) macrophages. In lipid-laden macrophages, Ampk activation decreased cholesterol content (foam cell formation) and increased cholesterol efflux to HDL and apoA-I, effects that occurred in an Ampk ß1-dependent manner. Increased cholesterol efflux was also associated with increased gene expression of the ATP binding cassette transporters, Abcg1 and Abca1. Moreover, in vivo reverse cholesterol transport was suppressed in mice that received Ampk ß1(-/-) macrophages compared with the WT control. Our data highlight the therapeutic potential of targeting macrophage Ampk with new or existing drugs for the possible reduction in foam cell formation during the early stages of atherosclerosis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cholesterol/metabolism , Enzyme Activators/pharmacology , Foam Cells/enzymology , Salicylic Acid/pharmacology , Animals , Apolipoprotein A-I/metabolism , Atherosclerosis , Cells, Cultured , Cholesterol, HDL/metabolism , Drug Evaluation, Preclinical , Enzyme Activation , Foam Cells/drug effects , Homeostasis , Lipogenesis , Mice, Knockout
3.
J Lipid Res ; 50(12): 2486-501, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19461119

ABSTRACT

Recent studies have shown a link between obesity and endoplasmic reticulum (ER) stress. Perturbations in ER homeostasis cause ER stress and activation of the unfolded protein response (UPR). Adipocyte differentiation contributes to weight gain, and we have shown that markers of ER stress/UPR activation, including GRP78, phospho-eIF2, and spliced XBP1, are upregulated during adipogenesis. Given these findings, the objective of this study was to determine whether attenuation of UPR activation by the chemical chaperone 4-phenylbutyrate (4-PBA) inhibits adipogenesis. Exposure of 3T3-L1 preadipocytes to 4-PBA in the presence of differentiation media decreased expression of ER stress markers. Concomitant with the suppression of UPR activation, 4-PBA resulted in attenuation of adipogenesis as measured by lipid accumulation and adiponectin secretion. Consistent with these in vitro findings, female C57BL/6 mice fed a high-fat diet supplemented with 4-PBA showed a significant reduction in weight gain and had reduced fat pad mass, as compared with the high-fat diet alone group. Furthermore, 4-PBA supplementation decreased GRP78 expression in the adipose tissue and lowered plasma triglyceride, glucose, leptin, and adiponectin levels without altering food intake. Taken together, these results suggest that UPR activation contributes to adipogenesis and that blocking its activation with 4-PBA prevents adipocyte differentiation and weight gain in mice.


Subject(s)
Adipocytes/cytology , Adipocytes/drug effects , Adipogenesis/drug effects , DNA-Binding Proteins/chemistry , Phenylbutyrates/pharmacology , Protein Folding/drug effects , Transcription Factors/chemistry , 3T3-L1 Cells , Animals , Body Weight/drug effects , Cells, Cultured , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Dietary Supplements , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Female , Mice , Mice, Inbred C57BL , Phenylbutyrates/administration & dosage , Regulatory Factor X Transcription Factors , Transcription Factors/deficiency , Transcription Factors/metabolism , X-Box Binding Protein 1
4.
FASEB J ; 22(7): 2569-78, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18364397

ABSTRACT

A causal relationship between diet-induced hyperhomocysteinemia (HHcy) and accelerated atherosclerosis has been established in apolipoprotein E-deficient (apoE(-/-)) mice. However, it is not known whether the proatherogenic effect of HHcy in apoE(-/-) mice is independent of hyperlipidemia and/or deficiency of apoE. In this study, a comprehensive dietary approach using C57BL/6J mice was used to investigate whether HHcy is an independent risk factor for accelerated atherosclerosis or dependent on additional dietary factors that increase plasma lipids and/or inflammation. C57BL/6J mice at 4 wk of age were divided into 6 dietary groups: chow diet (C), chow diet + methionine (C+M), western-type diet (W), western-type diet + methionine (W+M), atherogenic diet (A), or atherogenic diet + methionine (A+M). After 2, 10, 20, or 40 wk on the diets, mice were sacrificed, and the levels of total plasma homocysteine, cysteine, and glutathione, as well as total plasma cholesterol and triglycerides were analyzed. Aortic root sections were examined for atherosclerotic lesions. HHcy was induced in all groups supplemented with methionine, compared to diet-matched control groups. Plasma total cholesterol was significantly increased in mice fed the W or A diet. However, the W diet increased LDL/IDL and HDL levels, while the A diet significantly elevated plasma VLDL and LDL/IDL levels without increasing HDL. No differences in plasma total cholesterol levels or lipid profiles were observed between methionine-supplemented groups and the diet-matched control groups. Early atherosclerotic lesions containing macrophage foam cells were only observed in mice fed the A or A + M diet. Furthermore, lesion size was significantly larger in the A + M group compared to the A group at 10 and 20 wk; however, mature lesions were never observed even after 40 wk on these diets. The presence of lymphocytes, increased hyaluronan staining, and the expression of endoplasmic reticulum (ER) stress markers were also increased in atherosclerotic lesions from the A + M group. Taken together, these results suggest that HHcy does not independently cause atherosclerosis in C57BL/6J mice even in the presence of increased total plasma lipids induced by the W diet. However, HHcy can accelerate atherosclerotic lesion development under dietary conditions that increase plasma VLDL levels and/or inflammation.


Subject(s)
Atherosclerosis/physiopathology , Hyperhomocysteinemia/chemically induced , Hyperhomocysteinemia/physiopathology , Methionine/pharmacology , Animals , Atherosclerosis/blood , Atherosclerosis/chemically induced , Atherosclerosis/pathology , Diet, Atherogenic , Dietary Supplements , Disease Models, Animal , Female , Homocysteine/blood , Hyaluronic Acid/metabolism , Hyperhomocysteinemia/blood , Hyperhomocysteinemia/pathology , Immunohistochemistry , Lipids/blood , Methionine/administration & dosage , Mice , Mice, Inbred C57BL
5.
Atherosclerosis ; 168(2): 255-62, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12801608

ABSTRACT

It has been demonstrated that hyperhomocysteinemia (HHcy) accelerates atherosclerosis in apolipoprotein E-deficient (apoE(-/-)) mice. In this study, vitamin-defined chow diets were used to induce HHcy in apoE(-/-) mice in an attempt to identify possible pathogenic pathways. Six-week-old female apoE(-/-) mice were divided into seven groups: vitamin-defined purified chow diet alone (control), or same diet supplemented with either D,L-homocysteine (upward arrow Hcy) or L-homocystine (upward arrow Hcy-Hcy), or diet high in L-methionine (upward arrow Met), or diet high in B-vitamins (upward arrow vitamin), or diets deficient in folate (downward arrow folate) or vitamin B(6) ( downward arrow B(6)). Eighteen weeks later, plasma total homocysteine (tHcy), lipids and atherosclerotic plaque burden (aortic root, aortic arch, and brachiocephalic trunk) were measured. tHcy levels were similar in the upward arrow vitamin, downward arrow folate, downward arrow B(6) and control groups (9.2-10.1 micromol/l, NS), but elevated mildly in the upward arrow Hcy-Hcy group (16.1 micromol/l) and moderately in the upward arrow Met and upward arrow Hcy groups (53.6 and 51.5 micromol/l, respectively). Mice in the latter two groups had significantly more atherosclerosis in the aortic root. Although B vitamin-supplementation failed to lower tHcy levels, mice had less atherosclerosis in the aortic arch. In summary, dietary methionine and homocysteine, but not homocystine, enhanced the development of atherosclerosis. Supplementation with B vitamins appeared to confer homocysteine-independent protection against atherosclerosis. These results suggest that (1) there may be a threshold level below which homocysteine is not atherogenic; (2) the atherogenic effect of HHcy may be mediated via an intracellular pathway; and/or (3) the anti-atherogenic effect of B vitamins in normohomocysteinemic mice is independent of tHcy levels.


Subject(s)
Apolipoproteins E/deficiency , Arteriosclerosis/etiology , Arteriosclerosis/prevention & control , Dietary Supplements , Hyperhomocysteinemia/complications , Vitamins/pharmacology , Animals , Aorta/pathology , Arteriosclerosis/pathology , Brachiocephalic Trunk/pathology , Diet , Female , Homocysteine/administration & dosage , Homocysteine/adverse effects , Homocysteine/blood , Hyperhomocysteinemia/blood , Lipids/blood , Methionine/administration & dosage , Methionine/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
J Biol Chem ; 278(32): 30317-27, 2003 Aug 08.
Article in English | MEDLINE | ID: mdl-12738777

ABSTRACT

Hyperhomocysteinemia is an independent risk factor for cardiovascular disease and accelerates atherosclerosis in apoE-/- mice. Despite the observations that homocysteine causes endoplasmic reticulum (ER) stress and programmed cell death (PCD) in cultured human vascular endothelial cells, the cellular factors responsible for this effect and their relevance to atherogenesis have not been completely elucidated. We report here that homocysteine induces the expression of T-cell death-associated gene 51 (TDAG51), a member of the pleckstrin homology-related domain family, in cultured human vascular endothelial cells. This effect was observed for other ER stress-inducing agents, including dithiothreitol and tunicamycin. TDAG51 expression was attenuated in homozygous A/A mutant eukaryotic translation initiation factor 2 alpha mouse embryonic fibroblasts treated with homocysteine or tunicamycin, suggesting that ER stress-induced phosphorylation of eukaryotic translation initiation factor 2 alpha is required for TDAG51 transcriptional activation. Transient overexpression of TDAG51 elicited significant changes in cell morphology, decreased cell adhesion, and promoted detachment-mediated PCD. In support of these in vitro findings, TDAG51 expression was increased and correlated with PCD in the atherosclerotic lesions from apoE-/- mice fed hyperhomocysteinemic diets, compared with mice fed a control diet. Collectively, these findings provide evidence that TDAG51 is induced by homocysteine, promotes detachment-mediated PCD, and contributes to the development of atherosclerosis observed in hyperhomocysteinemia.


Subject(s)
Apoptosis , Arteriosclerosis/pathology , Homocysteine/chemistry , Hyperhomocysteinemia/pathology , Transcription Factors/chemistry , Animals , Blotting, Northern , Cell Adhesion , Cell Nucleus/metabolism , Cells, Cultured , Cloning, Molecular , Crosses, Genetic , DNA, Complementary/metabolism , Endoplasmic Reticulum/metabolism , Endothelium, Vascular/cytology , Fluorescent Antibody Technique, Indirect , Gene Expression Profiling , Gene Library , Homocysteine/metabolism , Humans , Hyperhomocysteinemia/complications , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Subcellular Fractions , Time Factors , Tissue Distribution , Transcription Factors/metabolism , Transcriptional Activation , Transfection , Tunicamycin/pharmacology , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL